Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Cereb Blood Flow Metab ; 30(2): 352-60, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19861974

ABSTRACT

Diabetes is an increased risk factor for stroke and results in increased brain damage in experimental animals and humans. The precise mechanisms are unclear, but our earlier studies in the db/db mice suggested that the cerebral inflammatory response initiating recovery was both delayed and diminished in the diabetic mice compared with the nondiabetic db/+ mice. In this study, we investigated the actions of the peroxisome proliferator-activated receptor (PPAR)-gamma agonist darglitazone in treating diabetes and promoting recovery after a hypoxic-ischemic (H/I) insult in the diabetic ob/ob mouse. Male ob/+ and ob/ob mice received darglitazone (1 mg/kg) for 7 days before induction of H/I. Darglitazone restored euglycemia and normalized elevated corticosterone, triglycerides, and very-low-density lipoprotein levels. Darglitazone dramatically reduced the infarct size in the ob/ob mice at 24 h of recovery compared with the untreated group (30+/-13% to 3.3+/-1.6%, n=6 to 8) but did not show any significant effect in the ob/+ mice. Microglial and astrocytic activation monitored by cytokine expression (interleukin-1beta and tumor necrosis factor-alpha) and in situ hybridization studies (bfl1 and glial fibrillary acidic protein) suggest a biphasic inflammatory response, with darglitazone restoring the compromised proinflammatory response(s) in the diabetic mouse at 4 h but suppressing subsequent inflammatory responses at 8 and 24 h in both control and diabetic mice.


Subject(s)
Diabetes Mellitus, Experimental/immunology , Hypoglycemic Agents/pharmacology , Hypoxia-Ischemia, Brain/immunology , Inflammation/immunology , PPAR gamma/agonists , Thiazolidinediones/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Blood Glucose/analysis , Corticosterone/blood , Diabetes Mellitus, Experimental/complications , Hypoxia-Ischemia, Brain/complications , Hypoxia-Ischemia, Brain/metabolism , In Situ Hybridization , Lipoproteins, VLDL/blood , Male , Mice , Microglia/drug effects , Microglia/immunology , Microglia/metabolism , PPAR gamma/drug effects , Radioimmunoassay , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/blood
2.
J Neurosci Res ; 86(7): 1538-47, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18214991

ABSTRACT

Studies have shown that cytokines released following CNS injury can affect the supportive or cytotoxic functions of microglia. Interleukin-6 (IL-6)-family cytokines are among the injury factors released. To understand how microglia respond to IL-6 family cytokines, we examined the effects of ciliary neurotrophic factor (CNTF) and IL-6 on primary cultures of rat microglia. To assess the functional state of the cells, we assayed the expression of tumor necrosis factor-alpha (TNFalpha), interleukin-1beta (IL-1beta), and cyclooxygenase 2 (COX-2) following stimulation. We show that CNTF reduces COX-2 levels, whereas IL-6 increases the expression of IL-1beta, TNFalpha, and Cox-2. We also examined trophic factor expression and found that CNTF enhances glial cell-line derived neurotrophic factor (GDNF) mRNA and protein secretion, whereas IL-6 has no effect. Correspondingly, conditioned media from CNTF-stimulated microglia promote motor neuron survival threefold beyond controls, whereas IL-6-stimulated microglia decrease neuronal survival twofold. To understand better the signaling mechanisms responsible for the opposite responses of these IL-6-family cytokines, we examined STAT-3 and ERK phosphorylation in CNTF- and IL-6-stimulated microglia. IL-6 markedly increases STAT-3 and ERK phosphorylation after 20 min of treatment, whereas these signal transducers are weakly stimulated by CNTF across a range of doses. We conclude that CNTF modifies microglial activation to support neuronal survival and that IL-6 enhances their capacity to do harm, as a result of different modes of intracellular signaling.


Subject(s)
Ciliary Neurotrophic Factor/pharmacology , Interleukin-6/pharmacology , Microglia/drug effects , Animals , Animals, Newborn , Cells, Cultured , Cerebral Cortex , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Embryo, Mammalian , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Motor Neurons/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
3.
J Cereb Blood Flow Metab ; 27(4): 710-8, 2007 Apr.
Article in English | MEDLINE | ID: mdl-16926846

ABSTRACT

Impaired peripheral wound healing is a hallmark of diabetics pathology and has been attributed to compromised macrophage activation. Stroke is another component of diabetic pathology, with increased tissue infarction and worsened recovery although the mechanisms remain unresolved. In this study, we investigated whether a compromised glial/macrophage response might contribute to cerebral hypoxic-ischemic (H/I) brain damage in diabetic (db/db), relative to their normoglycemic db/+ mice. Hypoxia-ischemia was induced in 8-week-old male db/db and db/+ mice by the ligation of right common carotid artery followed by systemic hypoxia (8% O2: 92% N2) for 17 mins. Mice were killed at specific intervals of reperfusion/recovery and the brains analyzed by in situ hybridization or total RNA isolation. In situ hybridization using bfl-1 (microglia) and glial fibrillary acidic protein (GFAP) (astrocytes) revealed expression of both bfl-1 and GFAP in the ipsilateral hemisphere at 4 h in the db/+ mice, which was delayed and minimal in the db/db mice. RNase protection assays showed a robust increase in expression of the proinflammatory cytokines tumor necrosis factor-alpha (TNFalpha), interleukin-1 IL-1alpha, and IL-1beta mRNA in the db/+ mice at 6 to 8 h of reperfusion peaking at 8 to 12 h; in db/db mice expression was markedly delayed and diminished. Real-time-polymerase chain reaction (RT-PCR) confirmed the reduced and delayed expression TNFalpha, IL-1alpha, IL-1beta, and the growth factors insulin-like growth factor-1 and ciliary neurotrophic factor in the db/db mice; enzyme-linked immunosorbent assays confirmed the reduced and delayed translation of IL-1beta protein. These findings suggest that a compromised inflammatory response may underlie the greater infarct associated with diabetic db/db mice compared with their nondiabetic littermates following a hypoxic/ischemic insult.


Subject(s)
Diabetes Mellitus/genetics , Diabetes Mellitus/pathology , Hypoxia-Ischemia, Brain/pathology , Wound Healing/physiology , Animals , Astrocytes/physiology , Ciliary Neurotrophic Factor/biosynthesis , Enzyme-Linked Immunosorbent Assay , Glial Fibrillary Acidic Protein/biosynthesis , In Situ Hybridization , Insulin-Like Growth Factor I/biosynthesis , Interleukin-1alpha/biosynthesis , Interleukin-1alpha/physiology , Interleukin-1beta/biosynthesis , Interleukin-1beta/physiology , Male , Mice , Mice, Inbred C57BL , Microglia/physiology , Minor Histocompatibility Antigens , Nuclease Protection Assays , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/biosynthesis
4.
Diabetes ; 55(9): 2401-11, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16936187

ABSTRACT

Diabetic retinopathy remains a frightening prospect to patients and frustrates physicians. Destruction of damaged retina by photocoagulation remains the primary treatment nearly 50 years after its introduction. The diabetes pandemic requires new approaches to understand the pathophysiology and improve the detection, prevention, and treatment of retinopathy. This perspective considers how the unique anatomy and physiology of the retina may predispose it to the metabolic stresses of diabetes. The roles of neural retinal alterations and impaired retinal insulin action in the pathogenesis of early retinopathy and the mechanisms of vision loss are emphasized. Potential means to overcome limitations of current animal models and diagnostic testing are also presented with the goal of accelerating therapies to manage retinopathy in the face of ongoing diabetes.


Subject(s)
Diabetic Retinopathy , Diabetic Retinopathy/complications , Diabetic Retinopathy/etiology , Glucose/adverse effects , Humans , Microcirculation/drug effects , Receptor, Insulin/physiology , Retina/anatomy & histology , Retina/physiology , Retinitis/physiopathology , Vision Disorders/etiology
5.
J Neuroinflammation ; 3: 15, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16808851

ABSTRACT

The cytokines IL-1alpha and IL-1beta are induced rapidly after insults to the CNS, and their subsequent signaling through the type 1 IL-1 receptor (IL-1R1) has been regarded as essential for a normal astroglial and microglial/macrophage response. To determine whether abrogating signaling through the IL-1R1 will alter the cardinal astrocytic responses to injury, we analyzed molecules characteristic of activated astrocytes in response to a penetrating stab wound in wild type mice and mice with a targeted deletion of IL-1R1. Here we show that after a stab wound injury, glial fibrillary acidic protein (GFAP) induction on a per cell basis is delayed in the IL-1R1-null mice compared to wild type counterparts. However, the induction of chondroitin sulfate proteoglycans, tenascin, S-100B as well as glutamate transporter proteins, GLAST and GLT-1, and glutamine synthetase are independent of IL-1RI signaling. Cumulatively, our studies on gliosis in the IL-1R1-null mice indicate that abrogating IL-1R1 signaling delays some responses of astroglial activation; however, many of the important neuroprotective adaptations of astrocytes to brain trauma are preserved. These data recommend the continued development of therapeutics to abrogate IL-1R1 signaling to treat traumatic brain injuries. However, astroglial scar related proteins were induced irrespective of blocking IL-1R1 signaling and thus, other therapeutic strategies will be required to inhibit glial scarring.

6.
Stroke ; 36(10): 2226-31, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16179572

ABSTRACT

BACKGROUND AND PURPOSE: Interleukin-1 (IL-1) is a proinflammatory cytokine implicated in multiple neurodegenerative diseases, including stroke. However, to date, there is no consensus regarding which receptor(s) mediates the detrimental effects of IL-1. We hypothesized that abrogating IL-1 type 1 receptor (IL-1R1) signaling would reduce edema, chemokine expression, and leukocyte infiltration; lower levels of iNOS; and, consequently, decrease free radical damage after mild hypoxia/ischemia (H/I), thus preserving brain cells. METHODS: IL-1R1 null mice and wild-type mice were subjected to a mild H/I insult. MRI was used to measure the area affected at 30 minutes and 48 hours after H/I. An RNAse protection assay was used to evaluate changes in chemokine mRNA expression. RT-PCR was used to assess inducible nitric oxide synthase (iNOS) and endothelial nitric oxide synthase mRNA levels. Immunohistochemistry was used to assess leukocyte infiltration. Western blots were used to assess iNOS and glutamate aspartate transporter protein levels. RESULTS: IL-1R1 null mice had reduced cytotoxic and vasogenic edema. The volume of hyperintense signal on T2-weighted images was reduced on average by 90% at 48 hours after H/I. The induction of multiple chemokine mRNAs was significantly reduced in IL-1R1 null mice compared with wild-type mice at 18 and 72 hours after H/I, which correlated with fewer infiltrating CD3+ leukocytes. Levels of iNOS protein and mRNA (but not glutamate aspartate transporter) were significantly reduced in the IL-1R1 mouse brain. CONCLUSIONS: These findings indicate that abrogating IL-1R1 signaling could protect brain cells subsequent to a mild stroke by reducing edema and immune cell recruitment, as well as by limiting iNOS-mediated free radical damage.


Subject(s)
Edema/pathology , Inflammation/pathology , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/physiology , Amino Acid Transport System X-AG/metabolism , Animals , Blotting, Western , Brain/pathology , CD3 Complex/biosynthesis , Chemokines/metabolism , Crosses, Genetic , Cytokines/metabolism , DNA Primers/chemistry , Diffusion Magnetic Resonance Imaging , Disease Models, Animal , Free Radicals , Hypoxia/pathology , Hypoxia-Ischemia, Brain/pathology , Immunohistochemistry , Interleukin-1/metabolism , Leukocytes/cytology , Leukocytes/metabolism , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitric Oxide Synthase Type II/metabolism , RNA, Messenger/metabolism , Reperfusion Injury/prevention & control , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleases/metabolism , Signal Transduction , Stroke/pathology , Time Factors
7.
Invest Ophthalmol Vis Sci ; 46(6): 2210-8, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15914643

ABSTRACT

PURPOSE: This study tested the Ins2(Akita) mouse as an animal model of retinal complications in diabetes. The Ins2(Akita) mutation results in a single amino acid substitution in the insulin 2 gene that causes misfolding of the insulin protein. The mutation arose and is maintained on the C57BL/6J background. Male mice heterozygous for this mutation have progressive loss of beta-cell function, decreased pancreatic beta-cell density, and significant hyperglycemia, as early as 4 weeks of age. METHODS: Heterozygous Ins2(Akita) mice were bred to C57BL/6J mice, and male offspring were monitored for hyperglycemia, beginning at 4.5 weeks of age. After 4 to 36 weeks of hyperglycemia, the retinas were analyzed for vascular permeability, vascular lesions, leukostasis, morphologic changes of micro- and macroglia, apoptosis, retinal degeneration, and insulin receptor kinase activity. RESULTS: The mean blood glucose of Ins2(Akita) mice was significantly elevated, whereas the body weight at death was reduced compared with that of control animals. Compared with sibling control mice, the Ins2(Akita) mice had increased retinal vascular permeability after 12 weeks of hyperglycemia (P < 0.005), a modest increase in acellular capillaries after 36 weeks of hyperglycemia (P < 0.0008), and alterations in the morphology of astrocytes and microglia, but no changes in expression of Muller cell glial fibrillary acidic protein. Increased apoptosis was identified by immunoreactivity for active caspase-3 after 4 weeks of hyperglycemia (P < 0.01). After 22 weeks of hyperglycemia, there was a 16.7% central and 27% peripheral reduction in the thickness of the inner plexiform layer, a 15.6% peripheral reduction in the thickness of the inner nuclear layer (P < 0.001), and a 23.4% reduction in the number of cell bodies in the retinal ganglion cell layer (P < 0.005). In vitro insulin receptor kinase activity was reduced (P < 0.05) after 12 weeks of hyperglycemia. CONCLUSIONS: The retinas of heterozygous male Ins2(Akita) mice exhibit vascular, neural, and glial abnormalities generally consistent with clinical observations and other animal models of diabetes. In light of the relatively early, spontaneous onset of the disease and the popularity of the C57BL/6J inbred strain as a background for the generation and study of other genetic alterations, combining the Ins2(Akita) mutation with other engineered mutations will be of great use for studying the molecular basis of retinal complications of diabetes.


Subject(s)
Diabetic Retinopathy/physiopathology , Disease Models, Animal , Animals , Apoptosis , Astrocytes/metabolism , Astrocytes/pathology , Blood Glucose/metabolism , Body Weight , Capillary Permeability , Caspase 3 , Caspases/metabolism , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/physiopathology , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hyperglycemia/pathology , Insulin/genetics , Male , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Neuroglia/metabolism , Neuroglia/pathology , Receptor, Insulin/metabolism , Retinal Degeneration , Retinal Vessels/pathology
8.
Diabetes ; 54(5): 1559-65, 2005 May.
Article in English | MEDLINE | ID: mdl-15855346

ABSTRACT

Diabetes leads to vascular leakage, glial dysfunction, and neuronal apoptosis within the retina. The goal of the studies reported here was to determine the role that retinal microglial cells play in diabetic retinopathy and assess whether minocycline can decrease microglial activation and alleviate retinal complications. Immunohistochemical analyses showed that retinal microglia are activated early in diabetes. Furthermore, mRNAs for interleukin-1beta and tumor necrosis factor-alpha, proinflammatory mediators known to be released from microglia, are also increased in the retina early in the course of diabetes. Using an in vitro bioassay, we demonstrated that cytokine-activated microglia release cytotoxins that kill retinal neurons. Furthermore, we showed that neuronal apoptosis is increased in the diabetic retina, as measured by caspase-3 activity. Minocycline represses diabetes-induced inflammatory cytokine production, reduces the release of cytotoxins from activated microglia, and significantly reduces measurable caspase-3 activity within the retina. These results indicate that inhibiting microglial activity may be an important strategy in the treatment of diabetic retinopathy and that drugs such as minocycline hold promise in delaying or preventing the loss of vision associated with this disease.


Subject(s)
Caspases/metabolism , Cytokines/genetics , Diabetes Mellitus, Experimental/immunology , Diabetic Retinopathy/pathology , Inflammation/immunology , Microglia/pathology , Minocycline/therapeutic use , Animals , Caspase 3 , Caspases/drug effects , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/prevention & control , Disease Models, Animal , Inflammation/prevention & control , Microglia/drug effects , Rats , Rats, Sprague-Dawley
9.
J Cereb Blood Flow Metab ; 25(1): 17-29, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15678109

ABSTRACT

Excessive inflammation has been implicated in the progressive neurodegeneration that occurs in multiple neurological diseases, including cerebral ischemia, and elevated levels of the proinflammatory cytokine interleukin-1 (IL-1) have been shown to exacerbate brain damage, whereas diminishing IL-1 levels limits the extent of injury. However, to date there is no consensus regarding which receptor(s) mediates the detrimental effects of IL-1. Because we have previously demonstrated that signaling through the IL-1 type 1 receptor (IL-1R1) is necessary for microglial activation and because results from other studies have implicated microglia as effectors of neurodegeneration, we hypothesized that inactivating the IL-1R1 would decrease the extent of damage caused by a hypoxic-ischemic (H/I) insult. It is shown that a mild insult initiates progressive neurodegeneration that leads to cystic infarcts, which can be prevented by inactivating the IL-1R1. The IL-1R1 null mice also show preserved sensorimotor function at 1 month's recovery. The mild insult induces multiple proinflammatory cytokines and activates microglia, and these responses are dramatically curtailed in mice lacking the IL-1R1. Importantly, the neuroinflammation precedes the progressive enlargement of the infarct, suggesting that the inflammation is causal rather than a consequence of the brain damage. These findings show that abrogating the inflammation consequent to a mild H/I insult will prevent brain damage and preserve neurological function. Additionally, these data incriminate the IL-1R1 as a master proinflammatory cytokine receptor.


Subject(s)
Brain Injuries/metabolism , Heredodegenerative Disorders, Nervous System/metabolism , Hypoxia-Ischemia, Brain/metabolism , Receptors, Interleukin-1/metabolism , Recovery of Function/physiology , Animals , Brain Injuries/etiology , Brain Injuries/pathology , Heredodegenerative Disorders, Nervous System/etiology , Heredodegenerative Disorders, Nervous System/pathology , Hypoxia-Ischemia, Brain/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Receptors, Interleukin-1/genetics , Recovery of Function/genetics
10.
J Neurosci Res ; 78(2): 151-6, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15378607

ABSTRACT

Interleukins 1alpha and 1beta (IL-1) are very potent signaling molecules that are expressed normally at low levels, but are induced rapidly in response to local or peripheral insults. IL-1 coordinates systemic host defense responses to pathogens and to injury and not surprisingly it has similar effects within the central nervous system (CNS). Numerous reports have correlated the presence of IL-1 in the injured or diseased brain, and its effects on neurons and nonneuronal cells in the CNS, but it is only recently that the importance of IL-1 signaling has been recognized. This article reviews studies that demonstrate that IL-1 is at or near the top of the hierarchical cytokine signaling cascade in the CNS that results in the activation of endogenous microglia and vascular endothelial cells to recruit peripheral leukocytes (i.e., neuroinflammation). The IL-1 system thus provides an attractive target for therapeutic intervention to ameliorate the destructive consequences of neuroinflammation.


Subject(s)
Central Nervous System Diseases/immunology , Interleukin-1/physiology , Animals , Central Nervous System Diseases/therapy , Humans , Inflammation/immunology , Neurodegenerative Diseases/immunology , Signal Transduction/physiology
11.
Brain Res Mol Brain Res ; 117(1): 58-67, 2003 Sep 10.
Article in English | MEDLINE | ID: mdl-14499481

ABSTRACT

Synaptic plasticity is important for formation of long-term memories and in re-establishment of function following injury. Seven cDNAs enriched following lesion in the hippocampus of the rat have been isolated using a PCR-based cDNA suppression subtraction hybridization. Sequence analysis resulted in the identification of two genes with known roles in synaptic development and neuronal activities: astrotactin and calcineurin. These two neuron-specific genes have established roles in development or synaptogenesis. Sequence analysis of the other five additional genes shows that two are likely to be involved in G-protein signaling pathways, one is a WD repeat protein, and the remaining two are entirely novel. All seven candidates are expressed in the hippocampus and, in some cases, cortical layers of adult brains. RT-PCR data show that expression increases following synaptogenic lesion. Immunocytochemical analysis in primary hippocampal neurons showed that Calcineurin immunoreactivity was redistributed in neurons during 2 weeks in culture. This redistribution suggests that Calcineurin's role changes during neurite outgrowth immediately prior to synapse formation in vitro. In addition, inhibiting Calcineurin activity with cyclosporin A enhanced neurite outgrowth, suggesting that Calcineurin has a regulatory role in axon sprouting. The discovery of previously unknown genes involved in the response to neurodegeneration will contribute to our understanding of neural development, responses to CNS trauma, and neurodegenerative diseases.


Subject(s)
Brain Injuries/metabolism , DNA, Complementary/metabolism , Hippocampus/metabolism , Neurons/metabolism , Animals , Animals, Newborn , Blotting, Northern , Brain/metabolism , Brain Injuries/chemically induced , Brain Injuries/genetics , Calcineurin/genetics , Cell Division , Cell Size/physiology , Cells, Cultured , Cloning, Molecular , Colchicine/toxicity , Cyclosporine/administration & dosage , DNA, Complementary/genetics , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Gene Library , Hippocampus/pathology , Immunohistochemistry , In Situ Hybridization , Neurites/metabolism , RNA, Messenger/biosynthesis , Rats , Rats, Inbred F344 , Reverse Transcriptase Polymerase Chain Reaction , Synapsins/metabolism , Time Factors
12.
Glia ; 44(1): 76-84, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12951659

ABSTRACT

IL-1alpha and IL-1beta are induced immediately after insults to the brain, and signaling through the type 1 IL-1 receptor is essential for a normal microglial and astroglial response to injury. To better understand which genes are induced in astrocytes by IL-1beta, we used the unbiased technique of differential display to analyze mouse astroglial gene expression after IL-1beta treatment. Two novel genes were induced, as well as the gene for ceruloplasmin, a ferroxidase with antioxidant properties. Ceruloplasmin was analyzed further by Northern and Western blot. RNA and protein levels of ceruloplasmin were increased when astrocytes were treated with IL-1beta. To determine whether the IL-1 type 1 receptor (IL-1R1) is essential for the injury-induced expression of ceruloplasmin, a Western blot analysis was performed after a traumatic brain injury on mice that were IL-1R1-deficient. Ceruloplasmin increased significantly above controls after injury; however, injury-induced levels of ceruloplasmin were lower in IL-1R1-deficient (2.7-fold increase) than in the wild-type animals (3.5-fold increase). These data indicate that while IL-1R1 deletion has a slight effect on ceruloplasmin expression, it is not essential for either the basal or the induced expression of ceruloplasmin in vivo. Since ceruloplasmin buffers free copper, oxidizes ferrous iron, and catalyzes the dismutation of free radicals, increased levels of ceruloplasmin likely protect neurons and glia from sustaining damage after injury. Furthermore, as the IL-1R1 has been proposed to be a target for achieving neuroprotection after injury, these data suggest that the protection afforded by ceruloplasmin will be retained even when the IL-1R1 is antagonized.


Subject(s)
Astrocytes/metabolism , Brain Injuries/genetics , Ceruloplasmin/metabolism , Interleukin-1/metabolism , Receptors, Interleukin-1/deficiency , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/immunology , Brain Injuries/immunology , Brain Injuries/metabolism , Cells, Cultured , Ceruloplasmin/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gliosis/genetics , Gliosis/immunology , Gliosis/metabolism , Interleukin-1/immunology , Interleukin-1/pharmacology , Mice , Mice, Inbred C57BL , Nerve Degeneration/genetics , Nerve Degeneration/immunology , Nerve Degeneration/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1 Type I
13.
Glia ; 40(1): 11-24, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12237840

ABSTRACT

Protein tyrosine kinase (PTK) activity is abundant in microglia, but the PTKs that participate in their activation have not been identified. For these studies, we used three paradigms to characterize PTK expression during microglial activation: resting and activated microglia were bulk fractionated from the adult brain, cultured newborn microglia were treated with lipopolysaccharide (LPS) to model the transition from activated toward phagocytic microglia, and PTK expression was examined in activated microglia in situ after facial nerve axotomy. Two PCR-based strategies were used to show that 21 different PTK genes are expressed by rat brain microglia: 5 receptor PTKs, 10 nonreceptor PTKs, and 6 members of the src family. Seven of the 21 PTKs were examined in greater detail. Five PTK mRNAs (fgr, hck, fak, jak-2, and flk-1) increased expression across all three models of activation. We conclude that they represent key components in the cascades that participate in microglial activation. In contrast, expression of fes and fms correlated with stimuli that affect microglial proliferation. Four of the PTKs (hck, fgr, fes, and fms) are believed to be myeloid cell specific and were not expressed by cultured astrocytes. HCK and FAK protein were also not expressed in lysates of immature astrocytes and oligodendrocytes. Because of their putative specificity, these kinases represent potential targets for inhibitors of microglial activation. Because reactive microglia can exacerbate the severity of neurological diseases, the identification of specific kinases that participate in microglial activation represents an important advance toward the development of new therapeutics.


Subject(s)
Antigens, CD , Antigens, Neoplasm , Antigens, Surface , Avian Proteins , Blood Proteins , Brain/enzymology , Encephalitis/enzymology , Gene Expression Regulation, Enzymologic/genetics , Microglia/enzymology , Protein-Tyrosine Kinases/genetics , Animals , Animals, Newborn , Axotomy , Basigin , Brain/immunology , Brain/physiopathology , Cells, Cultured , Encephalitis/chemically induced , Encephalitis/genetics , Facial Nerve Injuries/enzymology , Facial Nerve Injuries/genetics , Facial Nerve Injuries/physiopathology , Immunohistochemistry , Lipopolysaccharides , Membrane Glycoproteins/metabolism , Microglia/cytology , Microglia/metabolism , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Signal Transduction/physiology , src-Family Kinases/genetics , src-Family Kinases/metabolism
14.
Glia ; 40(1): 109-20, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12237848

ABSTRACT

Microglia rapidly respond to CNS injury, yet the mechanisms leading to their activation and inactivation remain poorly defined. In particular, few studies have established how interactions between inflammatory mediators affect the innate immune response of microglia. To begin to establish how microglia integrate signals from multiple inflammatory mediators, we examined the effects of interleukin 1beta (IL-1beta), interleukin 6 (IL-6), tumor necrosis factor alpha (TNFalpha), interferon gamma (IFN-gamma), and transforming growth factor beta1 (TGFbeta1) on both newborn and bulk-isolated adult microglia. To assess the functional state of the cells, we assayed the expression of cyclooxygenase 2 (Cox-2), interleukin 6, and tumor necrosis factor alpha, and two protein tyrosine kinases that have been implicated in microglial responses to activational stimuli, HCK and FAK. These studies demonstrated that IL-1beta, TNFalpha, IL-6, but not IFN-gamma increase the expression of Cox-2, whereas they all increase the expression of HCK and FAK. In these studies, TGFbeta1 either had no effect, or it decreased basal levels of these proteins. TGFbeta1 blocked activation by IL-1beta when given prior to, or simultaneously with, IL-1beta. TGFbeta1 blocked the induction of the tyrosine kinases, Cox-2, and the induction of IL-6 and TNFalpha mRNAs. However, TGFbeta1 was ineffective in antagonizing the induction of Cox-2 by either IL-6 or TNFalpha. We conclude that the TGFbeta receptor signaling cascades intersect with IL-1, but they may not interact with IL-6 or TNFalpha signaling pathways that lead to activation.


Subject(s)
Brain Injuries/metabolism , Brain/metabolism , Chemotaxis/immunology , Encephalitis/metabolism , Inflammation Mediators/immunology , Microglia/metabolism , Animals , Animals, Newborn , Brain/drug effects , Brain/immunology , Brain Injuries/immunology , Brain Injuries/physiopathology , Cells, Cultured , Chemotaxis/drug effects , Cyclooxygenase 2 , Drug Interactions/immunology , Encephalitis/immunology , Encephalitis/physiopathology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Inflammation Mediators/pharmacology , Interleukin-1/genetics , Interleukin-1/immunology , Interleukin-1/pharmacology , Interleukin-6/genetics , Interleukin-6/immunology , Interleukin-6/pharmacology , Isoenzymes/drug effects , Isoenzymes/immunology , Male , Microglia/drug effects , Microglia/immunology , Prostaglandin-Endoperoxide Synthases/drug effects , Prostaglandin-Endoperoxide Synthases/immunology , Protein-Tyrosine Kinases/drug effects , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Proto-Oncogene Proteins/drug effects , Proto-Oncogene Proteins/immunology , Proto-Oncogene Proteins c-hck , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/immunology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/drug effects , Up-Regulation/immunology
15.
Glia ; 39(1): 85-97, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12112378

ABSTRACT

We have used a model of hypoxic-ischemic brain injury in adult male C57BL/6 mice to study insulin-like growth factor-I (IGF-I) and IGF-binding protein (IGFBP) expression in response to cerebral hypoxia-ischemia (H/I) in the adult mouse. A period of 20 min of H/I that resulted in histopathology in cortex, striatum, and thalamus was correlated with induction of mRNA for IGF-I, IGFBP-2, IGFBP-3, IGFBP-5, and glial fibrillary acidic protein (GFAP) by 4 days of recovery. Increased IGF-I mRNA was located within damaged regions and was surrounded by IGFBP-2 mRNA expression. The results of combined immunostaining/in situ hybridzation showed that the cells expressing IGFBP-2 mRNA were also GFAP-positive and comprised a subset of activated astrocytes immediately surrounding areas of damage. In contrast, staining within damaged regions showed high numbers of cells immunopositive for F4/80 and lectin B(4) indicative of microglia and macrophages but no cells immunopositive for the astrocytic proteins GFAP or S-100beta. Microglia/macrophages within the damaged areas expressed IGF-I mRNA and were also immunopositive for the proliferating cell nuclear antigen. To determine whether expression of IGF-I could contribute to proliferation of microglia, we treated purified cultures of adult brain microglia with IGF-I in the presence of (3)H-thymidine. IGF-I stimulated a twofold increase in DNA synthesis in cultures of adult brain microglia. Taken together with previous data demonstrating that IGF-I promotes proliferation of peripheral macrophages, these data support the hypothesis that IGF-I is an autocrine/paracrine mitogen for microglia/macrophages after H/I.


Subject(s)
Brain Ischemia/metabolism , Brain Ischemia/pathology , Insulin-Like Growth Factor I/biosynthesis , Macrophages/metabolism , Macrophages/pathology , Microglia/metabolism , Microglia/pathology , Animals , Astrocytes/metabolism , Cell Division , Cells, Cultured , DNA Replication , Insulin-Like Growth Factor Binding Protein 2/biosynthesis , Insulin-Like Growth Factor Binding Protein 2/genetics , Insulin-Like Growth Factor I/genetics , Macrophage Activation , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/biosynthesis
16.
J Neurosci ; 22(14): 6071-82, 2002 Jul 15.
Article in English | MEDLINE | ID: mdl-12122068

ABSTRACT

Interleukin-1 (IL-1) is induced immediately after insults to the brain, and elevated levels of IL-1 have been strongly implicated in the neurodegeneration that accompanies stroke, Alzheimer's disease, and multiple sclerosis. In animal models, antagonizing IL-1 has been shown to reduce cell death; however, the basis for this protection has not been elucidated. Here we analyzed the response to penetrating brain injury in mice lacking the type 1 IL-1 receptor (IL-1R1) to determine which cellular and molecular mediators of tissue damage require IL-1 signaling. At the cellular level, fewer amoeboid microglia/macrophages appeared adjacent to the injured brain tissue in IL-1R1 null mice, and those microglia present at early postinjury intervals retained their resting morphology. Astrogliosis also was mildly abrogated. At the molecular level, cyclooxygenase-2 (Cox-2) and IL-6 expression were depressed and delayed. Interestingly, basal levels of Cox-2, IL-1, and IL-6 were significantly lower in the IL-1R1 null mice. In addition, stimulation of vascular cell adhesion molecule-1 mRNA was depressed in the IL-1R1 null mice, and correspondingly, there was reduced diapedesis of peripheral macrophages in the IL-1R1 null brain after injury. This observation correlated with a reduced number of Cox-2+ amoeboid phagocytes adjacent to the injury. In contrast, several molecular aspects of the injury response were normal, including expression of tumor necrosis factor-alpha and the production of nerve growth factor. Because antagonizing IL-1 protects neural cells in experimental models of stroke and multiple sclerosis, our data suggest that cell preservation is achieved by abrogating microglial/macrophage activation and the subsequent self-propagating cycle of inflammation.


Subject(s)
Head Injuries, Penetrating/physiopathology , Inflammation Mediators/metabolism , Microglia/metabolism , Receptors, Interleukin-1/metabolism , Animals , Cell Count , Cyclooxygenase 2 , Cyclophilins/genetics , Cyclophilins/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Gliosis/pathology , Gliosis/prevention & control , Interleukin-1/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Macrophage Activation , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Prostaglandin-Endoperoxide Synthases/genetics , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/metabolism , RNA, Messenger/metabolism , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1 Type I , Signal Transduction , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...