Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters










Publication year range
1.
Brain Behav Immun ; 73: 670-681, 2018 10.
Article in English | MEDLINE | ID: mdl-30063972

ABSTRACT

Ischemic stroke is amongst the leading causes of death and disabilities. The available treatments are suitable for only a fraction of patients and thus novel therapies are urgently needed. Blockage of one of the cerebral arteries leads to massive and persisting inflammatory reaction contributing to the nearby neuronal damage. Targeting the detrimental pathways of neuroinflammation has been suggested to be beneficial in conditions of ischemic stroke. Nuclear receptor 4A-family (NR4A) member Nurr1 has been shown to be a potent modulator of harmful inflammatory reactions, yet the role of Nurr1 in cerebral stroke remains unknown. Here we show for the first time that an agonist for the dimeric transcription factor Nurr1/retinoid X receptor (RXR), HX600, reduces microglia expressed proinflammatory mediators and prevents inflammation induced neuronal death in in vitro co-culture model of neurons and microglia. Importantly, HX600 was protective in a mouse model of permanent middle cerebral artery occlusion and alleviated the stroke induced motor deficits. Along with the anti-inflammatory capacity of HX600 in vitro, treatment of ischemic mice with HX600 reduced ischemia induced Iba-1, p38 and TREM2 immunoreactivities, protected endogenous microglia from ischemia induced death and prevented leukocyte infiltration. These anti-inflammatory functions were associated with reduced levels of brain lysophosphatidylcholines (lysoPCs) and acylcarnitines, metabolites related to proinflammatory events. These data demonstrate that HX600 driven Nurr1 activation is beneficial in ischemic stroke and propose that targeting Nurr1 is a novel candidate for conditions involving neuroinflammatory component.


Subject(s)
Dibenzazepines/pharmacology , Nerve Degeneration/prevention & control , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Animals , Brain/metabolism , Brain Ischemia/metabolism , Brain Ischemia/physiopathology , Disease Models, Animal , Infarction, Middle Cerebral Artery/metabolism , Inflammation/metabolism , Membrane Glycoproteins/analysis , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neurons/metabolism , Neuroprotective Agents/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 2/agonists , Primary Cell Culture , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Immunologic/analysis , Receptors, Immunologic/metabolism , Retinoid X Receptors/agonists , Retinoid X Receptors/physiology , Stroke/metabolism
2.
CNS Drugs ; 31(12): 1057-1082, 2017 12.
Article in English | MEDLINE | ID: mdl-29260466

ABSTRACT

Over the past few decades, research on Alzheimer's disease (AD) has focused on pathomechanisms linked to two of the major pathological hallmarks of extracellular deposition of beta-amyloid peptides and intra-neuronal formation of neurofibrils. Recently, a third disease component, the neuroinflammatory reaction mediated by cerebral innate immune cells, has entered the spotlight, prompted by findings from genetic, pre-clinical, and clinical studies. Various proteins that arise during neurodegeneration, including beta-amyloid, tau, heat shock proteins, and chromogranin, among others, act as danger-associated molecular patterns, that-upon engagement of pattern recognition receptors-induce inflammatory signaling pathways and ultimately lead to the production and release of immune mediators. These may have beneficial effects but ultimately compromise neuronal function and cause cell death. The current review, assembled by participants of the Chiclana Summer School on Neuroinflammation 2016, provides an overview of our current understanding of AD-related immune processes. We describe the principal cellular and molecular players in inflammation as they pertain to AD, examine modifying factors, and discuss potential future therapeutic targets.


Subject(s)
Alzheimer Disease/drug therapy , Inflammation/drug therapy , Molecular Targeted Therapy , Alzheimer Disease/immunology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Humans , Immunity, Innate/immunology , Inflammation/immunology , Inflammation/physiopathology
3.
Sci Rep ; 7: 42270, 2017 02 16.
Article in English | MEDLINE | ID: mdl-28205585

ABSTRACT

Alzheimer's disease (AD) is characterized by extensive neuron loss that accompanies profound impairments in memory and cognition. We examined the neuronally directed effects of the retinoid X receptor agonist bexarotene in an aggressive model of AD. We report that a two week treatment of 3.5 month old 5XFAD mice with bexarotene resulted in the clearance of intraneuronal amyloid deposits. Importantly, neuronal loss was attenuated by 44% in the subiculum in mice 4 months of age and 18% in layer V of the cortex in mice 8 months of age. Moreover, bexarotene treatment improved remote memory stabilization in fear conditioned mice and improved olfactory cross habituation. These improvements in neuron viability and function were correlated with significant increases in the levels of post-synaptic marker PSD95 and the pre-synaptic marker synaptophysin. Moreover, bexarotene pretreatment improved neuron survival in primary 5XFAD neurons in vitro in response to glutamate-induced excitotoxicity. The salutary effects of bexarotene were accompanied by reduced plaque burden, decreased astrogliosis, and suppression of inflammatory gene expression. Collectively, these data provide evidence that bexarotene treatment reduced neuron loss, elevated levels of markers of synaptic integrity that was linked to improved cognition and in an aggressive model of AD.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Neurons/metabolism , Retinoid X Receptors/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Bexarotene , Biomarkers/metabolism , Cell Survival/drug effects , Cytokines/metabolism , Disease Models, Animal , Female , Gliosis/complications , Gliosis/drug therapy , Gliosis/pathology , Glutamic Acid/toxicity , Habituation, Psychophysiologic/drug effects , Hippocampus/pathology , Inflammation/pathology , Male , Membrane Transport Proteins/metabolism , Memory/drug effects , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , Neurotoxins/toxicity , Olfactory Bulb/drug effects , Olfactory Bulb/pathology , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Receptors, LDL/metabolism , Synapses/drug effects , Synapses/metabolism , Tetrahydronaphthalenes/pharmacology , Tetrahydronaphthalenes/therapeutic use
4.
Neuroscience ; 170(3): 846-57, 2010 Oct 27.
Article in English | MEDLINE | ID: mdl-20691766

ABSTRACT

Stroke is a leading cause of death and disability but has limited therapeutic options. Thiazolidinediones (TZDs), agonists for the nuclear receptor, peroxisome proliferator-activated receptor (PPAR)γ, reduce infarct volume and improve neurologic function following transient middle cerebral artery occlusion (MCAO) in rats. Translation of these findings into clinical therapy will require careful assessment of dosing paradigms and effective time windows for treatment. Understanding the mechanisms by which TZDs protect the brain provides insight into how time windows for neuroprotection might be extended. We find that two TZDs, pioglitazone and rosiglitazone, significantly reduce infarct volume at doses similar to those used clinically (1 mg/kg for pioglitazone and 0.1 mg/kg for rosiglitazone). We also find that pioglitazone reduces infarction volume in a transient, but not a permanent MCAO model suggesting that reperfusion plays an important role in TZD mediated neuroprotection. Since PPARγ agonists reduce inflammation and oxidative stress, both of which are exacerbated by reperfusion, we hypothesized that TZDs would be most effective if administered prior to reperfusion. We administered TZDs 3 h after MCAO and found that infarction volume and neurologic function are significantly improved in animals reperfused at 3 h and 15 min (after TZD treatment), but not in animals reperfused at 2 h (before TZD treatment) when assessed either 24 h or 3 weeks after MCAO. While TZDs reduce intercellular adhesion molecule (ICAM) expression to a similar extent regardless of the time of reperfusion, leukocyte entry into brain parenchyma is more dramatically reduced when reperfusion is delayed until after drug treatment. The finding that delaying reperfusion until after TZD treatment is beneficial despite a longer period of ischemia, is dramatic given the widely held view that duration of ischemia is the most important determinate of injury.


Subject(s)
Brain Ischemia/drug therapy , Neuroprotective Agents/administration & dosage , Reperfusion/methods , Stroke/drug therapy , Thiazolidinediones/administration & dosage , Animals , Behavior, Animal/drug effects , Blood Pressure/drug effects , Brain/blood supply , Brain/drug effects , Brain/metabolism , Brain/pathology , Brain Ischemia/pathology , Cell Adhesion Molecules/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Leukocytes/drug effects , Male , Pioglitazone , Rats , Rats, Wistar , Regional Blood Flow/drug effects , Rosiglitazone
5.
Mol Psychiatry ; 14(4): 448-61, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18227838

ABSTRACT

The extracellular signal-regulated kinase (ERK) pathway mediates neuronal plasticity in the CNS. The mood stabilizers lithium and valproate activate the ERK pathway in prefrontal cortex and hippocampus and potentiate ERK pathway-mediated neurite growth, neuronal survival and hippocampal neurogenesis. Here, we examined the role of the ERK pathway in behavioral plasticity related to facets of bipolar disorder. Mice with ERK1 ablation acquired reduced phosphorylation of RSK1, an ERK substrate, in prefrontal cortex and striatum, but not in hippocampus or cerebellum, indicating the ablation-induced brain region-specific ERK signaling deficits. ERK1 ablation produced a behavioral excitement profile similar to that induced by psychostimulants. The profile is characterized by hyperactivity, enhanced goal-directed activity and increased pleasure-related activity with potential harmful consequence. ERK1-ablated mice were hyperactive in multiple tests and resistant to behavioral despair in the forced swim test. These mice displayed more home-cage voluntary wheel running activities, rearings in a large arena and open-arm visits in an elevated plus maze. Treatments with valproate and olanzapine, but not lithium reduced baseline activities in ERK1-ablated mice. All three treatments attenuated amphetamine-induced hyperactivity in ablated mice. These data indicate a profound involvement of ERK1 signaling in behavioral excitement and in the behavioral action of antimanic agents. The extent to which ERK pathway perturbation contributes to the susceptibility, mood switch mechanism(s) and symptom pathophysiology of bipolar disorder requires further investigation. Whether there is a shared mechanism through which mood stabilizers produce their clinical actions on mood, thought and behavioral symptoms of mania also requires further investigation.


Subject(s)
Behavior, Animal/physiology , Extracellular Signal-Regulated MAP Kinases/physiology , Signal Transduction/physiology , Adjuvants, Immunologic , Administration, Oral , Amphetamine/pharmacology , Analysis of Variance , Animals , Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Benzodiazepines/pharmacology , Central Nervous System Stimulants/pharmacology , Enzyme Inhibitors/pharmacology , Lithium Chloride/administration & dosage , Locomotion/drug effects , Locomotion/genetics , Maze Learning/drug effects , Maze Learning/physiology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/deficiency , Olanzapine , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Signal Transduction/genetics , Swimming , Valproic Acid/pharmacology
6.
Eur J Neurosci ; 24(6): 1653-63, 2006 Sep.
Article in English | MEDLINE | ID: mdl-17004929

ABSTRACT

Stroke is a devastating disease with limited treatment options. Recently, we found that the peroxisome proliferator-activated receptor-gamma (PPARgamma) agonists troglitazone and pioglitazone reduce injury and inflammation in a rat model of transient cerebral ischemia. The mechanism of this protection is unclear, as these agents can act through PPAR-gamma activation or through PPAR-gamma-independent mechanisms. Therefore, we examined PPAR-gamma expression, DNA binding and transcriptional activity following stroke. In addition, we used a PPAR-gamma antagonist, T0070907, to determine the role of PPAR-gamma during ischemia. Using immunohistochemical techniques and real-time PCR, we found low levels of PPAR-gamma mRNA and PPAR-gamma immunoreactivity in nonischemic brain; however, PPAR-gamma expression dramatically increased in ischemic neurons, peaking 24 h following middle cerebral artery occlusion. Interestingly, we found that in both vehicle- and agonist-treated brains, DNA binding was reduced in the ischemic hemisphere relative to the contralateral hemisphere. Expression of a PPAR-gamma target gene, lipoprotein lipase, was also reduced in ischemic relative to nonischemic brain. Both DNA binding and lipoprotein lipase expression were increased by the addition of the PPAR-gamma agonist rosiglitazone. Finally, we found that rosiglitazone-mediated protection after stroke was reversed by the PPAR-gamma antagonist T0070907. Interestingly, infarction size was also increased by T0070907 in the absence of PPAR-gamma agonist, suggesting that endogenous PPAR-gamma ligands may mitigate the effects of cerebral ischemia.


Subject(s)
Gene Expression Regulation/physiology , Ischemic Attack, Transient/metabolism , PPAR gamma/metabolism , Animals , Benzamides/pharmacology , Enzyme Activation/physiology , Gene Expression Regulation/drug effects , Immunohistochemistry/methods , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/pathology , Male , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , PPAR gamma/genetics , Protein Binding/physiology , Pyridines/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction/methods , Rosiglitazone , Thiazolidinediones/therapeutic use , Time Factors
7.
Neuroscience ; 130(3): 685-96, 2005.
Article in English | MEDLINE | ID: mdl-15590152

ABSTRACT

Newly developed insulin-sensitizing agents, which target the nuclear receptor peroxisome proliferator-activated receptor-gamma have recently been appreciated to exhibit potent anti-inflammatory actions. Since stroke is associated with an intense inflammatory response, we reasoned that these agents may ameliorate injury from stroke. We report that administration of troglitazone or pioglitazone 24 h before and at the time of cerebral infarction dramatically reduced infarction volume and improved neurological function following middle cerebral artery occlusion in rats. Furthermore, we find that delayed therapy also significantly reduced infarct volume. The brains of the drug-treated animals displayed reduced inflammation as evidenced by decreased immunoreactivity for microglial/macrophage markers and reduced protein and mRNA for interleukin-1beta, cyclooxygenase-2 and inducible nitric oxide synthase. We argue that the beneficial effects of these drugs are likely due to reduced expression of these inflammatory mediators, which are known to exacerbate ischemic injury following stroke. These results are of particular relevance to diabetic patients chronically treated with these agents who may benefit from the neuroprotective actions of these drugs.


Subject(s)
Chromans/therapeutic use , Encephalitis/drug therapy , Encephalitis/pathology , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/drug therapy , Ischemic Attack, Transient/pathology , PPAR gamma/drug effects , Thiazolidinediones/therapeutic use , Animals , Blood Glucose/metabolism , Brain Chemistry/drug effects , Brain Chemistry/genetics , Cell Count , Cerebrovascular Circulation/physiology , Dose-Response Relationship, Drug , Encephalitis/etiology , Immunohistochemistry , Infarction, Middle Cerebral Artery/etiology , Ischemic Attack, Transient/metabolism , Ligands , Macrophages/drug effects , Male , Microglia/drug effects , Middle Cerebral Artery/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Pioglitazone , Psychomotor Performance/drug effects , Psychomotor Performance/physiology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Troglitazone
8.
J Neurochem ; 82(3): 615-24, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12153485

ABSTRACT

We examined the effect of pioglitazone, a peroxisome proliferator-activated receptor-gamma (PPARgamma) agonist of the thiazolidinedione class, on dopaminergic nerve cell death and glial activation in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease. The acute intoxication of C57BL/6 mice with MPTP led to nigrostriatal injury, as determined by tyrosine hydroxylase (TH) immunocytochemistry, and HPLC detection of striatal dopamine and metabolites. Damage to the nigrostriatal dopamine system was accompanied by a transient activation of microglia, as determined by macrophage antigen-1 (Mac-1) and inducible nitric oxide synthase (iNOS) immunoreactivity, and a prolonged astrocytic response. Orally administered pioglitazone (approximately 20 mg/kg/day) attenuated the MPTP-induced glial activation and prevented the dopaminergic cell loss in the substantia nigra pars compacta (SNpc). In contrast, there was little reduction of MPTP-induced dopamine depletion, with no detectable effect on loss of TH immunoreactivity and glial response in the striatum of pioglitazone-treated animals. Low levels of PPARgamma expression were detected in the ventral mesencephalon and striatum, and were unaffected by MPTP or pioglitazone treatment. Since pioglitazone affects primarily the SNpc in our model, different PPARgamma-independent mechanisms may regulate glial activation in the dopaminergic terminals compared with the dopaminergic cell bodies after acute MPTP intoxication.


Subject(s)
Parkinsonian Disorders/prevention & control , Receptors, Cytoplasmic and Nuclear/agonists , Thiazoles/pharmacology , Thiazolidinediones , Transcription Factors/agonists , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , 3,4-Dihydroxyphenylacetic Acid/metabolism , Administration, Oral , Animals , Cell Count , Cell Survival/drug effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/metabolism , Homovanillic Acid/metabolism , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Immunohistochemistry , Macrophage-1 Antigen/biosynthesis , Male , Mice , Mice, Inbred C57BL , Neuroglia/drug effects , Neuroglia/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Pioglitazone , Receptors, Cytoplasmic and Nuclear/biosynthesis , Substantia Nigra/drug effects , Substantia Nigra/metabolism , Substantia Nigra/pathology , Thiazoles/administration & dosage , Transcription Factors/biosynthesis , Tyrosine 3-Monooxygenase/metabolism
9.
Neurochem Int ; 39(5-6): 449-57, 2001.
Article in English | MEDLINE | ID: mdl-11578780

ABSTRACT

Amyloid deposition within the brains of Alzheimer's Disease patients results in the activation of microglial cells and the induction of a local inflammatory response. The interaction of microglia or monocytes with beta-amyloid (A beta) fibrils elicits the activation a complex tyrosine kinase-based signal transduction cascade leading to stimulation of multiple independent signaling pathways and ultimately to changes in proinflammatory gene expression. The A beta-stimulated expression of proinflammatory genes in myeloid lineage cells is antagonized by the action of a family of ligand-activated nuclear hormone receptors, the peroxisome proliferator-activated receptors (PPARs). We report that THP-1 monocytes express predominantly PPAR gamma isoform and lower levels of PPAR alpha and PPAR delta isoforms. PPAR mRNA levels are not affected by differentiation of the cells into a macrophage phenotype, nor are they altered following exposure to the classical immune stimulus, lipopolysaccharide. Previous studies have found that PPAR gamma agonists act broadly to inhibit inflammatory responses. The present study explored the action of the PPAR alpha isoform and found that PPAR alpha agonists inhibited the A beta-stimulated expression of TNFalpha and IL-6 reporter genes in a dose-dependent manner. Moreover, the PPAR alpha agonist WY14643 inhibited macrophage differentiation and COX-2 gene expression. However, the PPAR alpha agonists failed to inhibit A beta-stimulated elaboration of neurotoxic factors by THP-1 cells. These findings demonstrate that PPAR alpha acts to suppress a diverse array of inflammatory responses in monocytes.


Subject(s)
Amyloid beta-Peptides/physiology , Inflammation/etiology , Membrane Proteins , Receptors, Cytoplasmic and Nuclear/physiology , Receptors, Lipoprotein , Transcription Factors/physiology , Amyloid/pharmacology , Cell Differentiation/drug effects , Cell Line , Cyclooxygenase 2 , Humans , Interleukin-6/antagonists & inhibitors , Isoenzymes/metabolism , Macrophages/cytology , Microglia/physiology , Monocytes/cytology , Monocytes/physiology , Neurotoxins/antagonists & inhibitors , Prostaglandin-Endoperoxide Synthases/metabolism , Protein-Tyrosine Kinases/physiology , Receptor for Advanced Glycation End Products , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Immunologic/physiology , Receptors, Scavenger , Scavenger Receptors, Class B , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factors/agonists , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors
10.
Microsc Res Tech ; 54(2): 59-70, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11455613

ABSTRACT

The etiology of Alzheimer's disease (AD) involves a significant inflammatory component as evidenced by the presence of elevated levels of a diverse range of proinflammatory molecules in the AD brain. These inflammatory molecules are produced principally by activated microglia, which are found to be clustered within and adjacent to the senile plaque. Moreover, long-term treatment of patients with non-steroidal anti-inflammatory drugs has been shown to reduce risk and incidence of AD and delay disease progression. The microglia respond to beta-amyloid (Abeta) deposition in the brain through the interaction of fibrillar forms of amyloid with cell surface receptors, leading to the activation of intracellular signal transduction cascades. The activation of multiple independent signaling pathways ultimately leads to the induction of proinflammatory gene expression and production of reactive oxygen and nitrogen species. These microglial inflammatory products act in concert to produce neuronal toxicity and death. Therapeutic approaches focused on inhibition of the microglial-mediated local inflammatory response in the AD brain offer new opportunities to intervene in the disease.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Inflammation , Microglia/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Alzheimer Disease/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Humans , Signal Transduction
11.
J Biol Chem ; 276(21): 18169-77, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11278445

ABSTRACT

In PC12 cells, epidermal growth factor (EGF) transiently stimulates the mitogen-activated protein (MAP) kinases, ERK1 and ERK2, and provokes cellular proliferation. In contrast, nerve growth factor (NGF) stimulation leads to the sustained activation of the MAPKs and subsequently to neuronal differentiation. It has been shown that both the magnitude and longevity of MAPK activation governs the nature of the cellular response. The activations of MAPKs are dependent upon two distinct small G-proteins, Ras and Rap1, that link the growth factor receptors to the MAPK cascade by activating c-Raf and B-Raf, respectively. We found that Ras was transiently stimulated upon both EGF and NGF treatment of PC12 cells. However, EGF transiently activated Rap1, whereas NGF stimulated prolonged Rap1 activation. The activation of the ERKs was due almost exclusively (>90%) to the action of B-Raf. The transient activation of the MAPKs by EGF was a consequence of the formation of a short lived complex assembling on the EGF receptor itself, composed of Crk, C3G, Rap1, and B-Raf. In contrast, NGF stimulation of the cells resulted in the phosphorylation of FRS2. FRS2 scaffolded the assembly of a stable complex of Crk, C3G, Rap1, and B-Raf resulting in the prolonged activation of the MAPKs. Together, these data provide a signaling link between growth factor receptors and MAPK activation and a mechanistic explanation of the differential MAPK kinetics exhibited by these growth factors.


Subject(s)
Epidermal Growth Factor/pharmacology , MAP Kinase Signaling System/drug effects , Nerve Growth Factor/pharmacology , Animals , PC12 Cells , Rats , Signal Transduction/drug effects
12.
Learn Mem ; 8(1): 11-9, 2001.
Article in English | MEDLINE | ID: mdl-11160759

ABSTRACT

The extracellular signal-regulated kinases (ERKs) are members of the mitogen-activated protein kinase (MAPK) superfamily of enzymes and have recently garnered considerable attention in the field of learning and memory. ERK activation has been shown to be required for the induction of long-term potentiation (LTP) in the rat hippocampus and for the formation of associative and spatial memories in both the rat and the mouse. However, the individual roles for the two isoforms of ERK have yet to be deciphered. To investigate the specific contribution of the ERK1 (p44) isoform of MAPK to mammalian learning, we performed a general behavioral and physiological characterization of mice lacking the ERK1 gene. The ERK1-null animals demonstrated significantly higher levels of activity in the open field test. However, we observed no other discernible deficits in the ERK1 knockout mice in our behavioral testing. Specifically, no differences were observed in the acquisition or retention (24 h and 2 wk after training) of either contextual or cue fear conditioning between the ERK1(-/-) and their wild-type littermate controls. In addition, no learning phenotype was observed in the passive avoidance test. When hippocampal slices were analyzed, we found no deficits in baseline synaptic transmission or in tetanus-induced LTP in hippocampal area CA1. We found no apparent compensatory changes in the expression of ERK2 (p42 MAPK). We conclude that hippocampus- and amygdala-dependent emotional learning does not depend critically on the activity of ERK1.


Subject(s)
Emotions/physiology , Learning/physiology , Mitogen-Activated Protein Kinases/physiology , Animals , Hippocampus/physiology , Mice , Mice, Knockout/genetics , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/genetics , Motor Activity/physiology , Reference Values , Sensation/physiology
14.
J Neurosci ; 21(4): 1179-88, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11160388

ABSTRACT

Reactive microglia associated with the beta-amyloid plaques in Alzheimer's disease (AD) brains initiate a sequence of inflammatory events integral to the disease process. We have observed that fibrillar beta-amyloid peptides activate a tyrosine kinase-based signaling response in primary mouse microglia and the human monocytic cell line, THP-1, resulting in production of neurotoxic secretory products, proinflammatory cytokines, and reactive oxygen species. We report that most of the amyloid-induced tyrosine kinase activity was stimulated after activation of Src family members such as Lyn. However, transduction of the signaling response required for increased production of the cytokines TNFalpha and IL1-beta was mediated by the nonreceptor tyrosine kinase, Syk. Additionally, beta-amyloid stimulated an NFkappaB-dependent pathway in parallel that was required for cytokine production. Importantly, TNFalpha generated by the monocytes and microglia was responsible for the majority of the neuorotoxic activity secreted by these cells after beta-amyloid stimulation but must act in concert with other factors elaborated by microglia to elicit neuronal death. Moreover, we observed that the neuronal loss was apoptotic in nature and involved increased neuronal expression of inducible nitric oxide synthase and subsequent peroxynitrite production. Selective inhibitors of inducible nitric oxide synthase effectively protected cells from toxicity associated with the microglial and monocytic secretory products. This study demonstrates a functional linkage between beta-amyloid-dependent activation of microglia and several characteristic markers of neuronal death occurring in Alzheimer's disease brains.


Subject(s)
Amyloid beta-Peptides/metabolism , Microglia/metabolism , Monocytes/metabolism , Neurons/metabolism , Nitric Oxide Synthase/metabolism , Tumor Necrosis Factor-alpha/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Apoptosis , Cells, Cultured , Contraindications , Enzyme Precursors/metabolism , Humans , Inflammation/immunology , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins , Mice , Mice, Inbred C57BL , Microglia/cytology , Microglia/drug effects , Monocytes/cytology , Monocytes/drug effects , NF-kappa B/metabolism , Neurons/cytology , Neurons/drug effects , Nitric Oxide Synthase Type II , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Syk Kinase , Transcription Factors/metabolism , src-Family Kinases/metabolism
15.
Neurobiol Aging ; 22(6): 937-44, 2001.
Article in English | MEDLINE | ID: mdl-11755002

ABSTRACT

The role of inflammatory processes in the brains of Alzheimer's Disease (AD) patients has recently attracted considerable interest. Indeed, the only demonstrated effective therapy for AD patients is long-term treatment with non-steroidal anti-inflammatory drugs (NSAIDs). The mechanistic basis of the efficacy of NSAIDs in AD remains unclear. However, the recent recognition that NSAIDs can bind to and activate the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma), has offered an explanation for the action of these drugs in AD. PPARgamma activation leads to the inhibition of microglial activation and the expression of a broad range of proinflammatory molecules. The newly appreciated anti-inflammatory actions of PPARgamma agonists may allow novel therapies for AD and other CNS indications with an inflammatory component.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Inflammation/prevention & control , Receptors, Cytoplasmic and Nuclear/agonists , Transcription Factors/agonists , Animals , Humans , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/physiology , Transcription Factors/biosynthesis , Transcription Factors/physiology
16.
J Cell Sci ; 113 ( Pt 18): 3241-53, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10954422

ABSTRACT

CLK1 was one of the first identified dual specificity kinases and is the founding member of the 'LAMMER' family of kinases. We have established the substrate site specificity of CLK1. We report here that truncation of the N terminus of CLK1 resulted in a dramatic increase in CLK1 enzymatic activity, indicating that the N terminus acts as a negative regulatory domain. The N-terminal truncation resulted in a 45-fold increase in V(max), suggesting that this domain does not contain a pseudo-substrate motif, but may act to conformationally constrain the catalytic activity of CLK1. Tyrosine phosphorylation has been proposed to be critical for CLK1 activity, however, CLK1 activity was unaffected by exposure to tyrosine phosphatases. Treatment of CLK1 with the serine/threonine specific phosphatase PP2A, resulted in a 2- to 6-fold increase in enzymatic activity. Incubation of CLK1 with tyrosine phosphatases in combination with PP2A abolished CLK1 activity. These data suggest that CLK1 is regulated by three distinct mechanisms that serve to both positively and negatively regulate CLK1 activity. CLK1 activity is positively regulated by phosphorylation on either tyrosine residues or serine/threonine residues, and is negatively regulated by steric constraints mediated by the N-terminal domain, as well as, by phosphorylation on a subset of serine/threonine residues within the catalytic domain. CLK1 mRNA is expressed at low levels in all tissues and cell lines examined. The full-length and truncated splice forms are expressed at roughly equivalent levels in most tissues. The ratio of the two splice variants of CLK1 can be altered by treatment with cycloheximide. CLK1 protein expression is limited to a small subset of highly localized neuronal populations in the rat brain. Contrary to previous studies using overexpression systems, we show that CLK1 protein is primarily found in the cytoplasm of these cells, with only a small fraction localized to the nucleus.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Binding Sites , Blotting, Western/methods , Brain/enzymology , Brain/pathology , Consensus Sequence , Mice , Molecular Sequence Data , Myelin Basic Protein/metabolism , PC12 Cells , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , RNA Splicing , RNA, Messenger , Rabbits , Rats , Reverse Transcriptase Polymerase Chain Reaction , Staining and Labeling/methods , Substrate Specificity , Transfection
17.
Neurobiol Aging ; 21(3): 383-421, 2000.
Article in English | MEDLINE | ID: mdl-10858586

ABSTRACT

Inflammation clearly occurs in pathologically vulnerable regions of the Alzheimer's disease (AD) brain, and it does so with the full complexity of local peripheral inflammatory responses. In the periphery, degenerating tissue and the deposition of highly insoluble abnormal materials are classical stimulants of inflammation. Likewise, in the AD brain damaged neurons and neurites and highly insoluble amyloid beta peptide deposits and neurofibrillary tangles provide obvious stimuli for inflammation. Because these stimuli are discrete, microlocalized, and present from early preclinical to terminal stages of AD, local upregulation of complement, cytokines, acute phase reactants, and other inflammatory mediators is also discrete, microlocalized, and chronic. Cumulated over many years, direct and bystander damage from AD inflammatory mechanisms is likely to significantly exacerbate the very pathogenic processes that gave rise to it. Thus, animal models and clinical studies, although still in their infancy, strongly suggest that AD inflammation significantly contributes to AD pathogenesis. By better understanding AD inflammatory and immunoregulatory processes, it should be possible to develop anti-inflammatory approaches that may not cure AD but will likely help slow the progression or delay the onset of this devastating disorder.


Subject(s)
Alzheimer Disease/pathology , Inflammation/pathology , Brain/pathology , Humans
18.
J Neurosci ; 20(2): 558-67, 2000 Jan 15.
Article in English | MEDLINE | ID: mdl-10632585

ABSTRACT

Alzheimer's disease (AD) is characterized by the extracellular deposition of beta-amyloid fibrils within the brain and the subsequent association and phenotypic activation of microglial cells associated with the amyloid plaque. The activated microglia mount a complex local proinflammatory response with the secretion of a diverse range of inflammatory products. Nonsteroidal anti-inflammatory drugs (NSAIDs) are efficacious in reducing the incidence and risk of AD and significantly delaying disease progression. A recently appreciated target of NSAIDs is the ligand-activated nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma). PPARgamma is a DNA-binding transcription factor whose transcriptional regulatory actions are activated after agonist binding. We report that NSAIDs, drugs of the thiazolidinedione class, and the natural ligand prostaglandin J2 act as agonists for PPARgamma and inhibit the beta-amyloid-stimulated secretion of proinflammatory products by microglia and monocytes responsible for neurotoxicity and astrocyte activation. The activation of PPARgamma also arrested the differentiation of monocytes into activated macrophages. PPARgamma agonists were shown to inhibit the beta-amyloid-stimulated expression of the cytokine genes interleukin-6 and tumor necrosis factor alpha. Furthermore, PPARgamma agonists inhibited the expression of cyclooxygenase-2. These data provide direct evidence that PPARgamma plays a critical role in regulating the inflammatory responses of microglia and monocytes to beta-amyloid. We argue that the efficacy of NSAIDs in the treatment of AD may be a consequence of their actions on PPARgamma rather than on their canonical targets the cyclooxygenases. Importantly, the efficacy of these agents in inhibiting a broad range of inflammatory responses suggests PPARgamma agonists may provide a novel therapeutic approach to AD.


Subject(s)
Alzheimer Disease/physiopathology , Amyloid beta-Peptides/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Astrocytes/physiology , Microglia/physiology , Peptide Fragments/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Thiazolidinediones , Transcription Factors/agonists , Animals , Animals, Newborn , Astrocytes/cytology , Brain/cytology , Brain/physiology , Cell Differentiation , Chromans/pharmacology , Cyclooxygenase 2 , Dinoprost/pharmacology , Genes, Reporter , Humans , Inflammation , Interleukin-6/genetics , Isoenzymes/metabolism , Membrane Proteins , Mice , Mice, Inbred C57BL , Microbodies/physiology , Microglia/cytology , Microglia/drug effects , Monocytes/cytology , Monocytes/drug effects , Monocytes/physiology , Prostaglandin-Endoperoxide Synthases/metabolism , Recombinant Proteins/biosynthesis , Tetradecanoylphorbol Acetate/pharmacology , Thiazoles/pharmacology , Transfection , Troglitazone , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/genetics
19.
J Biol Chem ; 274(53): 37965-73, 1999 Dec 31.
Article in English | MEDLINE | ID: mdl-10608864

ABSTRACT

L1-mediated axon growth involves intracellular signaling, but the precise mechanisms involved are not yet clear. We report a role for the mitogen-activated protein kinase (MAPK) cascade in L1 signaling. L1 physically associates with the MAPK cascade components Raf-1, ERK2, and the previously identified p90(rsk) in brain. In vitro, ERK2 can phosphorylate L1 at Ser(1204) and Ser(1248) of the L1 cytoplasmic domain. These two serines are conserved in the L1 family of cell adhesion molecules, also being found in neurofascin and NrCAM. The ability of ERK2 to phosphorylate L1 suggests that L1 signaling could directly regulate L1 function by phosphorylation of the L1 cytoplasmic domain. In L1-expressing 3T3 cells, L1 cross-linking can activate ERK2. Remarkably, the activated ERK localizes with endocytosed vesicular L1 rather than cell surface L1, indicating that L1 internalization and signaling are coupled. Inhibition of L1 internalization with dominant-negative dynamin prevents activation of ERK. These results show that L1-generated signals activate the MAPK cascade in a manner most likely to be important in regulating L1 intracellular trafficking.


Subject(s)
Endocytosis , MAP Kinase Signaling System , Membrane Glycoproteins/metabolism , Neural Cell Adhesion Molecules/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Chick Embryo , Enzyme Activation , Leukocyte L1 Antigen Complex , Membrane Glycoproteins/chemistry , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Neural Cell Adhesion Molecules/chemistry , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-raf/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism
20.
Exp Cell Res ; 253(2): 463-73, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10585269

ABSTRACT

CLK3, a member of the LAMMER family of dual-specificity protein kinases, is abundantly expressed in the reproductive system of male mice. Specifically, high levels of CLK3 protein expression are found in mature spermatozoa in the testis and epididymis. The majority of the CLK3 protein in the testis is a full-length kinase-containing form, and only a small amount of a catalytically inactive N-terminally truncated splice variant protein product is observed. Within the mature spermatozoa CLK3 is localized to the acrosome and tail. CLK3 is expelled from the sperm following the acrosome reaction and inactivated, likely by degradation by the proteases released by the sperm during the acrosome reaction. The CLK family of kinases has previously been implicated in mRNA splicing; however, the bulk of the CLK3 protein in these cells is located in the cytoplasm, suggesting that CLK3 may have additional roles in the cell.


Subject(s)
Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Spermatogenesis/physiology , Spermatozoa/enzymology , 3T3 Cells , Acrosome Reaction/physiology , Age Factors , Animals , Antibodies , Blotting, Western , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Male , Mice , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/immunology , Protein-Tyrosine Kinases/analysis , Protein-Tyrosine Kinases/immunology , RNA, Messenger/analysis , Testis/cytology , Testis/enzymology , Transcription, Genetic/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...