Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis ; 69(2): 463-478, 2019.
Article in English | MEDLINE | ID: mdl-31006686

ABSTRACT

There is increasing evidence suggesting that amyloidogenic proteins might form deposits in non-neuronal tissues in neurodegenerative disorders such as Alzheimer's or Parkinson's diseases. However, the detection of these aggregation-prone proteins within the human skin has been controversial. Using immunohistochemistry (IHC) and mass spectrometry tissue imaging (MALDI-MSI), fresh frozen human skin samples were analyzed for the expression and localization of neurodegenerative disease-related proteins. While α-synuclein was detected throughout the epidermal layer of the auricular samples (IHC and MALDI-MSI), tau and Aß34 were also localized to the epidermal layer (IHC). In addition to Aß peptides of varying length (e.g., Aß40, Aß42, Aß34), we also were able to detect inflammatory markers within the same sample sets (e.g., thymosin ß-4, psoriasin). While previous literature has described α-synuclein in the nucleus of neurons (e.g., Parkinson's disease), our current detection of α-synuclein in the nucleus of skin cells is novel. Imaging of α-synuclein or tau revealed that their presence was similar between the young and old samples in our present study. Future work may reveal differences relevant for diagnosis between these proteins at the molecular level (e.g., age-dependent post-translational modifications). Our novel detection of Aß34 in human skin suggests that, just like in the brain, it may represent a stable intermediate of the Aß40 and Aß42 degradation pathway.


Subject(s)
Amyloid beta-Peptides/metabolism , Epidermis/metabolism , Inflammation Mediators/metabolism , Neurodegenerative Diseases/metabolism , Peptide Fragments/metabolism , alpha-Synuclein/metabolism , tau Proteins/metabolism , Aged , Amyloid beta-Peptides/analysis , Child , Epidermis/chemistry , Epidermis/pathology , Female , Humans , Inflammation Mediators/analysis , Male , Middle Aged , Neurodegenerative Diseases/pathology , Peptide Fragments/analysis , Skin/chemistry , Skin/metabolism , Skin/pathology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , alpha-Synuclein/analysis , tau Proteins/analysis
2.
ACS Chem Neurosci ; 9(2): 260-271, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29078046

ABSTRACT

Dendritic polyglycerols (dPG), particularly dendritic polyglycerol sulfates (dPGS), have been intensively studied due to their intrinsic anti-inflammatory activity. As related to brain pathologies involving neuroinflammation, the current study examined if dPG and dPGS can (i) regulate neuroglial activation, and (ii) normalize the morphology and function of excitatory postsynaptic dendritic spines adversely affected by the neurotoxic 42 amino acid amyloid-ß (Aß42) peptide of Alzheimer disease (AD). The exact role of neuroglia, such as microglia and astrocytes, remains controversial especially their positive and negative impact on inflammatory processes in AD. To test dPGS effectiveness in AD models we used primary neuroglia and organotypic hippocampal slice cultures exposed to Aß42 peptide. Overall, our data indicate that dPGS is taken up by both microglia and astrocytes in a concentration- and time-dependent manner. The mechanism of action of dPGS involves binding to Aß42, i.e., a direct interaction between dPGS and Aß42 species interfered with Aß fibril formation and reduced the production of the neuroinflammagen lipocalin-2 (LCN2) mainly in astrocytes. Moreover, dPGS normalized the impairment of neuroglia and prevented the loss of dendritic spines at excitatory synapses in the hippocampus. In summary, dPGS has desirable therapeutic properties that may help reduce amyloid-induced neuroinflammation and neurotoxicity in AD.


Subject(s)
Dendrimers/pharmacology , Dendritic Spines/drug effects , Glycerol/analogs & derivatives , Glycerol/pharmacology , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Synapses/drug effects , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Dendritic Spines/metabolism , Dendritic Spines/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Humans , Lipocalin-2/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Atomic Force , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Neuroglia/metabolism , Neuroglia/pathology , Neuroimmunomodulation/drug effects , Neuroimmunomodulation/physiology , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Protein Aggregation, Pathological/drug therapy , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Surface Plasmon Resonance , Synapses/metabolism , Synapses/pathology , Tissue Culture Techniques
3.
PLoS One ; 12(8): e0182372, 2017.
Article in English | MEDLINE | ID: mdl-28792512

ABSTRACT

Within the microenvironment of multiple sclerosis lesions, oligodendrocytes are subject to metabolic stress reflecting effects of focal ischemia and inflammation. Previous studies have shown that under optimal conditions in vitro, the respiratory activity of human adult brain-derived oligodendrocytes is lower and more predominantly glycolytic compared to oligodendrocytes differentiated in vitro from post natal rat brain oligodendrocyte progenitor cells. In response to sub-lethal metabolic stress, adult human oligodendrocytes reduce overall energy production rate impacting the capacity to maintain myelination. Here, we directly compare the metabolic profiles of oligodendrocytes derived from adult rat brain with oligodendrocytes newly differentiated in vitro from oligodendrocyte progenitor cells obtained from the post natal rat brain, under both optimal culture and metabolic stress (low/no glucose) conditions. Oxygen consumption and extracellular acidification rates were measured using a Seahorse extracellular flux analyzer. Our findings indicate that under optimal conditions, adult rat oligodendrocytes preferentially use glycolysis whereas newly differentiated post natal rat oligodendrocytes, and the oligodendrocyte progenitor cells from which they are derived, mainly utilize oxidative phosphorylation to produce ATP. Metabolic stress increases the rate of ATP production via oxidative phosphorylation and significantly reduces glycolysis in adult oligodendrocytes. The rate of ATP production was relatively unchanged in newly differentiated post natal oligodendrocytes under these stress conditions, while it was significantly reduced in oligodendrocyte progenitor cells. Our study indicates that both age and maturation influence the metabolic profile under optimal and stressed conditions, emphasizing the need to consider these variables for in vitro studies that aim to model adult human disease.


Subject(s)
Brain/growth & development , Brain/metabolism , Cell Differentiation/physiology , Oligodendroglia/metabolism , Stress, Physiological/physiology , Adenosine Triphosphate/metabolism , Animals , Animals, Newborn , Brain/cytology , Cell Survival/physiology , Cells, Cultured , Cellular Senescence/physiology , Extracellular Space/metabolism , Glucose/deficiency , Glycolysis/physiology , Neural Stem Cells/metabolism , Oligodendroglia/cytology , Oxidation-Reduction , Oxygen Consumption/physiology , Phosphorylation/physiology , Rats, Sprague-Dawley
4.
Biochemistry ; 55(12): 1839-49, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26900939

ABSTRACT

Alzheimer's disease is characterized by deposition of the amyloid ß-peptide (Aß) in brain tissue of affected individuals. In recent years, many potential lead structures have been suggested that can potentially be used for diagnosis and therapy. However, the mode of action of these compounds is so far not understood. Among these small molecules, the nonsteroidal anti-inflammatory drug (NSAID) sulindac sulfide received a lot of attention. In this manuscript, we characterize the interaction between the monomeric Aß peptide and the NSAID sulindac sulfide. We find that sulindac sulfide efficiently depletes the pool of toxic oligomers by enhancing the rate of fibril formation. In vitro, sulindac sulfide forms colloidal particles which catalyze the formation of fibrils. Aggregation is immediate, presumably by perturbing the supersaturated Aß solution. We find that sulindac sulfide induced Aß aggregates are structurally homogeneous. The C-terminal part of the peptide adopts a ß-sheet structure, whereas the N-terminus is disordered. The salt bridge between D23 and K28 is present, similar as in wild type fibril structures. (13)C-(19)F transferred echo double resonance experiments suggest that sulindac sulfide colocalizes with the Aß peptide in the aggregate.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Peptide Fragments/metabolism , Protein Aggregates/physiology , Sulindac/analogs & derivatives , Amino Acid Sequence , Amyloid beta-Peptides/toxicity , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Molecular Sequence Data , Peptide Fragments/toxicity , Protein Aggregates/drug effects , Sulindac/pharmacology
5.
Sci Rep ; 5: 15410, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26510576

ABSTRACT

The amyloid-ß42 (Aß42) peptide is believed to be the main culprit in the pathogenesis of Alzheimer disease (AD), impairing synaptic function and initiating neuronal degeneration. Soluble Aß42 oligomers are highly toxic and contribute to progressive neuronal dysfunction, loss of synaptic spine density, and affect long-term potentiation (LTP). We have characterized a short, L-amino acid Aß-oligomer Interacting Peptide (AIP) that targets a relatively well-defined population of low-n Aß42 oligomers, rather than simply inhibiting the aggregation of Aß monomers into oligomers. Our data show that AIP diminishes the loss of Aß42-induced synaptic spine density and rescues LTP in organotypic hippocampal slice cultures. Notably, the AIP enantiomer (comprised of D-amino acids) attenuated the rough-eye phenotype in a transgenic Aß42 fly model and significantly improved the function of photoreceptors of these flies in electroretinography tests. Overall, our results indicate that specifically "trapping" low-n oligomers provides a novel strategy for toxic Aß42-oligomer recognition and removal.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/antagonists & inhibitors , Oligopeptides/pharmacology , Peptide Fragments/antagonists & inhibitors , Protein Aggregation, Pathological/drug therapy , Synapses/metabolism , Synaptic Transmission/drug effects , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Animals , Peptide Fragments/metabolism , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Rats , Rats, Wistar , Synapses/pathology
6.
Neuropharmacology ; 79: 506-14, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24412648

ABSTRACT

Prolonged exposure of oligodendrocyte progenitor cultures to non-toxic concentrations of glutamate receptor agonists for 24 h decreased cellular proliferation mediated by α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors. Since prolonged agonist stimulation can regulate the expression of various families of receptors, we examined this possibility. Pretreatment of progenitor cultures with 100 µM kainic acid (KA) for 1-24 h caused a time-dependent decrease in AMPA receptor activity, determined by agonist-induced (45)Ca(2+) uptake. The maximum effect (70-80% decrease), observed in the 24 h-pretreated cells, was accompanied by a significant reduction in AMPA receptor subunits, as determined by Western blotting. GluR2/3 and GluR4 subunits were the most affected. Receptor down-regulation and (45)Ca(2+) uptake were only partially reversible upon KA removal. Furthermore, 24 h co-treatment of cultures with CNQX blocked the KA-induced decreases in calcium uptake. To address whether calpain, a calcium-activated protease, was implicated in the regulation of the AMPA receptor subunits, cultures were treated with the specific inhibitor PD150606 alone or in combination with KA for 24 h. Calpain inhibition significantly increased GluR1 in both conditions and partly reversed downregulation of GluR4 by KA. Collectively, these results indicate that calpain is not involved in the agonist-induced down-regulation of AMPA receptors subunits 2/3 in oligodendrocyte progenitors, while it downregulates GluR1 and GluR4.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Kainic Acid/pharmacology , Oligodendroglia/drug effects , Receptors, AMPA/metabolism , Stem Cells/drug effects , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Acrylates/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Calcium/metabolism , Calpain/antagonists & inhibitors , Calpain/metabolism , Cells, Cultured , Down-Regulation/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Oligodendroglia/metabolism , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/metabolism , Stem Cells/metabolism , Time Factors
7.
Glia ; 62(1): 64-77, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166839

ABSTRACT

Oligodendrocytes (OLGs) produce and maintain myelin in the central nervous system (CNS). In the demyelinating autoimmune disease multiple sclerosis, OLGs are damaged and those remaining fail to fully remyelinate CNS lesions. Therefore, current therapies directed to restrain the inflammation process with approaches that protect and reconstitute oligodendrocyte density would be essential to pave the way of myelin repair. A critical signal for oligodendrocytes is insulin-like growth factor-1 (IGF-1), which promotes their development and ultimately myelin formation. PTEN inhibits the phosphoinositide 3-kinase (PI3K)/Akt signaling, a convergence downstream pathway for growth factors such as IGF-1. In this report, we temporarily inhibited PTEN activity by treating rat and human oligodendrocyte progenitors (OLPs) cultured alone or with dorsal root ganglion neurons (DRGNs) with bisperoxovanadium (phen). Our findings show that phen potentiates IGF-1 actions by increasing proliferation of OLPs in a concentration-dependent manner, and caused a sustained and time-dependent activation of the main pathways: PI3K/Akt/mammalian target of rapamycin (mTOR) and MEK/ERK. At low concentrations, IGF-1 and phen stimulated the differentiation of rat and human OLPs. Concordantly, the PTEN inhibitor together with IGF-1 robustly augmented myelin basic protein accumulation in rat newborn and human fetal OLGs co-cultured with DRGNs in a longer timeframe by promoting the elaboration of organized myelinated fibers as evidenced by confocal microscopy. Thus, our results suggest that a transient suppression of a potential barrier for myelination in combination with other therapeutic approaches including growth factors may be promising to improve the functional recovery of CNS injuries.


Subject(s)
Enzyme Inhibitors/pharmacology , Insulin-Like Growth Factor I/metabolism , Myelin Sheath/metabolism , Oligodendroglia/drug effects , Signal Transduction/drug effects , Vanadium Compounds/pharmacology , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Fetus/cytology , Ganglia, Spinal/cytology , Gene Expression Regulation/drug effects , Humans , Myelin Basic Protein/metabolism , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects
8.
Glia ; 60(7): 1130-44, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22511272

ABSTRACT

We previously reported that addition of extracellular matrix (ECM) extracts to rat Schwann cell-dorsal root ganglion neuron (DRGN) co-cultures activated mitogen-activated protein kinase (MAPK) p38, whereas inhibition blocked myelination. Here, we used p38 pharmacological inhibitors and gene silencing to assess their effects on downstream kinases and key transcription factors. We show that p38α regulates expression of the master transcription factor, Krox-20, required for the onset of myelination in Schwann cell-DRGNs, as assessed by immunocytochemistry and qRT-PCR. p38 activity is also required for the expression of the cell cycle inhibitor p27(kip1) , associated with Schwann cell differentiation. Three potential effectors of p38 were explored: MAPK-activated protein kinase-2 (MK2), mitogen and stress-activated protein kinase-1 (MSK-1), and the transcription factor cAMP response element-binding protein (CREB). Inhibition of MK2 with CMPD1 or gene knockdown with siRNAs reduced numbers of Krox-20-positive Schwann cells and expression of myelin proteins MBP and MAG. ECM activated CREB and increased Krox-20 expression, whereas CREB1 gene silencing reduced Krox-20. Furthermore, two nonselective inhibitors of MSK-1 (H89 and R0-318820) decreased ECM-induced CREB phosphorylation and, similar to anti-MSK-1 siRNAs, reduced Krox-20-positive cells. In addition, p38 modulated the expression of two transcription factors involved in the regulation of Krox-20 [suppressed cAMP-inducible protein (SCIP) and Sox10], but not Sox2, an antagonist of Krox-20. Collectively, our results show that p38 primarily directs Schwann cell differentiation and peripheral myelination by regulating Krox-20 expression through its downstream effectors, MK2 and MSK-1/CREB, and transcription factors SCIP and Sox10.


Subject(s)
Cell Differentiation/physiology , Early Growth Response Protein 2/metabolism , Myelin Sheath/metabolism , Schwann Cells/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Coculture Techniques , Cyclic AMP Response Element-Binding Protein/metabolism , Early Growth Response Protein 2/genetics , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Myelin Sheath/genetics , Neurons/metabolism , Octamer Transcription Factor-6/genetics , Octamer Transcription Factor-6/metabolism , Phosphorylation , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
9.
Exp Neurol ; 226(1): 47-57, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20696161

ABSTRACT

Fyn, a nonreceptor Src-like tyrosine kinase (SLK), plays an important role in oligodendrocyte differentiation and myelination in the brain. However, its role in myelination of peripheral nerves remains undefined. Here we report that selective inhibitors of SLKs (PP2 and SU6656) caused a dose-dependent decrease in the accumulation of several myelin proteins, including myelin basic protein (MBP), protein zero (P0) and myelin-associated glycoprotein (MAG) in rat Schwann cell-dorsal root ganglion neuron (SC-DRGN) co-cultures. Interestingly, SLK inhibition was insufficient to completely abrogate myelin synthesis, as removal of PP2 after several days of treatment permitted a partial recovery of myelin proteins expression. Furthermore, fewer and shorter myelinated segments formed in the continuous presence of PP2, although the myelin formed was normally compacted. PP2 also decreased the number of SCs expressing Krox-20, a master-regulatory transcription factor expressed by myelinating SCs, by 50%. These results were corroborated by selective knockdown of Fyn and Lyn kinases using siRNA. Extracellular matrix is important to SC differentiation and peripheral myelination. Using phospho-specific antibodies, we showed that addition of extracellular matrix extracts to SC-DRGN co-cultures resulted in the activation of ERK, Akt and p38 MAPK, three protein kinases involved in SC proliferation, differentiation and peripheral myelination. PP2 blocked the phosphorylation of all three kinases. Our results support a role for SLKs in the initiation of peripheral myelination via the activation of p38, Akt and ERK, which regulate Krox-20 expression and peripheral myelination.


Subject(s)
Myelin Sheath/enzymology , Myelin Sheath/physiology , Peripheral Nerves/enzymology , Peripheral Nerves/physiology , src-Family Kinases/physiology , Animals , Axons/metabolism , Blotting, Western , Cells, Cultured , Early Growth Response Protein 2/biosynthesis , Early Growth Response Protein 2/genetics , Extracellular Matrix/metabolism , Ganglia, Spinal/cytology , Ganglia, Spinal/enzymology , Immunohistochemistry , Microscopy, Electron , Myelin Proteins/biosynthesis , Myelin Proteins/genetics , Myelin Sheath/ultrastructure , Peripheral Nerves/ultrastructure , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-fyn/biosynthesis , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Schwann Cells/enzymology , Schwann Cells/ultrastructure , Sodium Channels/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases/biosynthesis , p38 Mitogen-Activated Protein Kinases/genetics , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
10.
Neurotoxicology ; 30(4): 544-54, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19523979

ABSTRACT

Cadmium toxicity has been associated with learning disabilities and Parkinsonian symptoms in humans. We have previously shown that cultured oligodendrocytes are directly damaged by cadmium exposure. Here, we characterized the molecular mechanisms underlying cadmium-induced cell death in oligodendrocyte progenitors (OLP). Cadmium caused a concentration-dependent decrease in cell viability as assessed by mitochondrial dehydrogenase activity and by the cellular release of lactate dehydrogenase (LDH). A short exposure (1h) to cadmium (25-100 microM), followed by several hours of recovery, produced a predominant apoptotic mechanism of cell death, involving the mitochondrial intrinsic pathway, as evidenced by nuclear condensation, DNA fragmentation, bax integration into the outer mitochondrial membrane, cytochrome c release, and activation of caspases-9 and -3. Pretreatment of OLPs with the pan-caspase inhibitor, zVAD-fmk, prevented caspase-3 activation but only slightly reduced cell death 11h after cadmium exposure and failed to prevent cadmium-induced bax insertion into the mitochondrial membrane. In contrast, the anti-oxidant N-acetyl cysteine blocked caspase-3 activation and significantly protected OLPs from cadmium-induced cell death. Continuous exposure (18-48 h) of OLPs to low micromolar concentrations (0.001-25 microM) of cadmium significantly decreased mitochondrial metabolic activity, increased LDH leakage starting at 5 microM and maximally activated caspase-3. These results suggest that cadmium induces OLP cell death mainly by apoptosis, and at higher concentrations or with prolonged exposure to the heavy metal there is an increase in cytoplasmic membrane damage, an index of necrosis. More importantly, transient exposure to cadmium is sufficient to damage OLPs and could in principle impair myelination in the neonate.


Subject(s)
Apoptosis/drug effects , Cadmium/toxicity , Mitochondria/drug effects , Oligodendroglia/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Analysis of Variance , Animals , Animals, Newborn , Brain/cytology , Caspase 3/metabolism , Cells, Cultured , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , In Situ Nick-End Labeling/methods , L-Lactate Dehydrogenase/metabolism , Neuroprotective Agents/pharmacology , Propidium , Rats , Rats, Sprague-Dawley , Time Factors
11.
J Mol Neurosci ; 35(1): 23-33, 2008 May.
Article in English | MEDLINE | ID: mdl-17994198

ABSTRACT

The p38 mitogen-activated protein kinase family is emerging as a crucial signaling molecule for a vast number of cellular functions including cell migration, proliferation, and differentiation. The function of p38 in myelination has only been recently addressed. Using pyridinyl imidazole-based p38 alpha/beta selective inhibitors, we have reported a critical role for this kinase in the regulation of myelination, specifically, in controlling the differentiation of Schwann cells, and oligodendrocytes, the myelinating glia of the peripheral and central nervous systems, respectively. These compounds inhibited the accumulation of myelin-cell-specific markers, including myelin-specific glycosphingolipids, myelin-associated glycoprotein, and myelin basic protein. More significantly, myelination of dorsal root ganglia neurons by oligodendrocytes was irreversibly blocked by p38 inhibitors. Our current studies are focusing on the molecular mechanisms by which p38 regulates oligodendrocyte and Schwann cell differentiation and its role in models of myelination and remyelination.


Subject(s)
Myelin Sheath/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Cycle/physiology , Cell Differentiation/physiology , Enzyme Inhibitors/metabolism , Ganglia, Spinal/cytology , MAP Kinase Signaling System/physiology , Neurons/cytology , Neurons/physiology , Oligodendroglia/cytology , Oligodendroglia/physiology , Schwann Cells/cytology , Schwann Cells/physiology , Stem Cells/cytology , Stem Cells/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
12.
J Biol Chem ; 277(43): 40775-81, 2002 Oct 25.
Article in English | MEDLINE | ID: mdl-12198121

ABSTRACT

Human neutrophil granulocytes die rapidly, and their survival is contingent upon rescue from programmed cell death by signals from the environment. Here we report that a novel signal for delaying neutrophil apoptosis is the classic acute phase reactant, C-reactive protein (CRP). However, this anti-apoptotic activity is expressed only when the cyclic pentameric structure of CRP is lost, resulting in formation of modified or monomeric CRP (mCRP), which may be formed in inflamed tissues. By contrast, native pentameric CRP and CRP peptides 77-82, 174-185, and 201-206 failed to affect neutrophil apoptosis. The apoptosis delaying action of mCRP was markedly attenuated by an antibody against the low affinity IgG immune complex receptor (CD16) but not by an anti-CD32 antibody. mCRP evoked a transient concurrent activation of the extracellular signal-regulated kinase (ERK) and phosphatidylinositol 3-kinase/Akt signaling pathways, leading to inhibition of caspase-3 and consequently to delaying apoptosis. Consistently, pharmacological inhibition of either ERK or Akt reversed the anti-apoptotic action of mCRP; however, they did not produce additive inhibition. Thus, mCRP, but not pentameric CRP or peptides derived from CRP, promotes neutrophil survival and may therefore contribute to amplification of the inflammatory response.


Subject(s)
Apoptosis/physiology , C-Reactive Protein/metabolism , Neutrophils/cytology , Adult , C-Reactive Protein/chemistry , C-Reactive Protein/physiology , Cells, Cultured , Enzyme Activation , Female , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , Protein Binding , Receptors, IgG/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...