Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
Add more filters










Publication year range
1.
Cells ; 10(5)2021 05 19.
Article in English | MEDLINE | ID: mdl-34069691

ABSTRACT

Heterogeneity of glia in different CNS regions may contribute to the selective vulnerability of neuronal populations in neurodegenerative conditions such as amyotrophic lateral sclerosis (ALS). Here, we explored regional variations in the expression of heat shock protein 25 in glia under conditions of acute and chronic stress. Hsp27 (Hsp27; murine orthologue: Hsp25) fulfils a number of cytoprotective functions and may therefore be a possible therapeutic target in ALS. We identified a subpopulation of astrocytes in primary murine mixed glial cultures that expressed Hsp25. Under basal conditions, the proportion of Hsp25-positive astrocytes was twice as high in spinal cord cultures than in cortical cultures. To explore the physiological role of the elevated Hsp25 expression in spinal cord astrocytes, we exposed cortical and spinal cord glia to acute stress, using heat stress and pro-inflammatory stimuli. Surprisingly, we observed no stress-induced increase in Hsp25 expression in either cortical or spinal cord astrocytes. Similarly, exposure to endogenous stress, as modelled in glial cultures from SOD1 G93A-ALS mice, did not increase Hsp25 expression above that observed in astrocytes from wild-type mice. In vivo, Hsp25 expression was greater under conditions of chronic stress present in the spinal cord of SOD1 G93A mice than in wild-type mice, although this increase in expression is likely to be due to the extensive gliosis that occurs in this model. Together, these results show that there are differences in the expression of Hsp25 in astrocytes in different regions of the central nervous system, but Hsp25 expression is not upregulated under acute or chronic stress conditions.


Subject(s)
Astrocytes/enzymology , Cerebral Cortex/enzymology , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Spinal Cord/enzymology , Superoxide Dismutase-1/metabolism , Animals , Astrocytes/drug effects , Astrocytes/pathology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Female , Gene Expression Regulation , Gliosis/enzymology , Gliosis/pathology , Heat-Shock Proteins/genetics , Heat-Shock Response , Humans , Lipopolysaccharides/pharmacology , Male , Mice, Inbred C57BL , Mice, Transgenic , Molecular Chaperones/genetics , Phenotype , Spinal Cord/drug effects , Spinal Cord/pathology , Superoxide Dismutase-1/genetics , Tumor Necrosis Factor-alpha/pharmacology
2.
Int J Mol Sci ; 20(7)2019 Apr 05.
Article in English | MEDLINE | ID: mdl-30959793

ABSTRACT

Theiler's murine encephalomyelitis (TME) represents a versatile animal model for studying the pathogenesis of demyelinating diseases such as multiple sclerosis. Hallmarks of TME are demyelination, astrogliosis, as well as inflammation. Previous studies showed that matrix metalloproteinase 12 knockout (Mmp12-/-) mice display an ameliorated clinical course associated with reduced demyelination. The present study aims to elucidate the impact of MMP12 deficiency in TME with special emphasis on astrogliosis, macrophages infiltrating the central nervous system (CNS), and the phenotype of microglia/macrophages (M1 or M2). SJL wild-type and Mmp12-/- mice were infected with TME virus (TMEV) or vehicle (mock) and euthanized at 28 and 98 days post infection (dpi). Immunohistochemistry or immunofluorescence of cervical and thoracic spinal cord for detecting glial fibrillary acidic protein (GFAP), ionized calcium-binding adaptor molecule 1 (Iba1), chemokine receptor 2 (CCR2), CD107b, CD16/32, and arginase I was performed and quantitatively evaluated. Statistical analyses included the Kruskal⁻Wallis test followed by Mann⁻Whitney U post hoc tests. TMEV-infected Mmp12-/- mice showed transiently reduced astrogliosis in association with a strong trend (p = 0.051) for a reduced density of activated/reactive microglia/macrophages compared with wild-type mice at 28 dpi. As astrocytes are an important source of cytokine production, including proinflammatory cytokines triggering or activating phagocytes, the origin of intralesional microglia/macrophages as well as their phenotype were determined. Only few phagocytes in wild-type and Mmp12-/- mice expressed CCR2, indicating that the majority of phagocytes are represented by microglia. In parallel to the reduced density of activated/reactive microglia at 98 dpi, TMEV-infected Mmp12-/- showed a trend (p = 0.073) for a reduced density of M1 (CD16/32- and CD107b-positive) microglia, while no difference regarding the density of M2 (arginase I- and CD107b-positive) cells was observed. However, a dominance of M1 cells was detected in the spinal cord of TMEV-infected mice at all time points. Reduced astrogliosis in Mmp12-/- mice was associated with a reduced density of activated/reactive microglia and a trend for a reduced density of M1 cells. This indicates that MMP12 plays an important role in microglia activation, polarization, and migration as well as astrogliosis and microglia/astrocyte interaction.


Subject(s)
Astrocytes/pathology , Cardiovirus Infections/enzymology , Cardiovirus Infections/virology , Gliosis/enzymology , Gliosis/virology , Matrix Metalloproteinase 12/deficiency , Microglia/pathology , Spinal Cord/pathology , Animals , Cell Movement , Glial Fibrillary Acidic Protein/metabolism , Immunophenotyping , Inflammation/metabolism , Inflammation/pathology , Macrophages/metabolism , Matrix Metalloproteinase 12/metabolism , Mice, Knockout , Phenotype
3.
Curr Alzheimer Res ; 16(3): 219-232, 2019.
Article in English | MEDLINE | ID: mdl-30827242

ABSTRACT

BACKGROUND: Amyloid-ß (Aß) accumulation plays a critical role in the pathogenesis of Alzheimer's disease (AD) lesions. Deficiency of Serotonin signaling recently has been linked to the increased Aß level in transgenic mice and humans. In addition, tryptophan hydroxylase-2 (Tph2), a second tryptophan hydroxylase isoform, controls brain serotonin synthesis. However, it remains to be determined that whether Tph2 deficient APP/PS1mice affect the formation of Aß plaques in vivo. METHODS: Both quantitative and qualitative immunochemistry methods, as well as Congo red staining were used to evaluate the Aß load and astrogliosis in these animals. RESULTS: we studied alterations of cortex and hippocampus in astrocytes and senile plaques by Tph2 conditional knockout (Tph2 CKO) AD mice from 6-10 months of age. Using Congo red staining and immunostained with Aß antibody, we showed that plaques load or plaques numbers significantly increased in Tph2 CKO experimental groups at 8 to 10 months old, compared to wild type (WT) group, respectively. Using GFAP+ astrocytes immunofluorescence method, we found that the density of GFAP+ astrocytes markedly enhanced in Tph2 CKO at 10 months. We showed Aß plaques co-localized autophagic markers LC3 and p62. Nevertheless, we did not observe any co-localization between GFAP+ astrocytes and autophagic markers, but detected the co-localization between ßIII-tubulin+ neurons and autophagic markers. CONCLUSION: Overall, our work provides the preliminary evidence in vivo that Tph2 plays a role in amyloid plaques generation.


Subject(s)
Alzheimer Disease/enzymology , Amyloid beta-Peptides/metabolism , Gliosis/enzymology , Plaque, Amyloid/enzymology , Tryptophan Hydroxylase/metabolism , Aging/metabolism , Aging/pathology , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Astrocytes/enzymology , Astrocytes/pathology , Brain/enzymology , Brain/pathology , Disease Models, Animal , Gliosis/pathology , Mice, Transgenic , Neurons/enzymology , Neurons/pathology , Plaque, Amyloid/pathology , Preliminary Data , Tryptophan Hydroxylase/genetics
4.
CNS Neurosci Ther ; 25(1): 86-100, 2019 01.
Article in English | MEDLINE | ID: mdl-29855151

ABSTRACT

AIMS: Usually, spinal cord injury (SCI) develops into a glial scar containing extracellular matrix molecules including chondroitin sulfate proteoglycans (CSPGs). Chondroitinase ABC (ChABC), from Proteus vulgaris degrading the glycosaminoglycan (GAG) side chains of CSPGs, offers the opportunity to improve the final outcome of SCI. However, ChABC usage is limited by its thermal instability, requiring protein structure modifications, consecutive injections at the lesion site, or implantation of infusion pumps. METHODS: Aiming at more feasible strategy to preserve ChABC catalytic activity, we assessed various stabilizing agents in different solutions and demonstrated, via a spectrophotometric protocol, that the 2.5 mol/L Sucrose solution best stabilized ChABC as far as 14 days in vitro. RESULTS: ChABC activity was improved in both stabilizing and diluted solutions at +37°C, that is, mimicking their usage in vivo. We also verified the safety of the proposed aqueous sucrose solution in terms of viability/cytotoxicity of mouse neural stem cells (NSCs) in both proliferating and differentiating conditions in vitro. Furthermore, we showed that a single intraspinal treatment with ChABC and sucrose reduced reactive gliosis at the injury site in chronic contusive SCI in rats and slightly enhanced their locomotor recovery. CONCLUSION: Usage of aqueous sucrose solutions may be a feasible strategy, in combination with rehabilitation, to ameliorate ChABC-based treatments to promote the regeneration of central nervous system injuries.


Subject(s)
Astrocytes/drug effects , Chondroitin ABC Lyase/pharmacology , Gliosis/drug therapy , Neuroprotective Agents/pharmacology , Spinal Cord Injuries/drug therapy , Animals , Astrocytes/enzymology , Astrocytes/pathology , Cells, Cultured , Chondroitin ABC Lyase/isolation & purification , Chondroitin ABC Lyase/metabolism , Chronic Disease , Disease Models, Animal , Enzyme Stability/drug effects , Gliosis/enzymology , Gliosis/pathology , Mice , Neural Stem Cells/drug effects , Neural Stem Cells/enzymology , Neural Stem Cells/pathology , Neuroprotective Agents/isolation & purification , Neuroprotective Agents/metabolism , Proteus vulgaris/enzymology , Random Allocation , Rats, Sprague-Dawley , Spinal Cord Injuries/enzymology , Spinal Cord Injuries/pathology , Spinal Cord Regeneration/drug effects
5.
Elife ; 72018 07 03.
Article in English | MEDLINE | ID: mdl-29968565

ABSTRACT

Neuropathic pain resulting from nerve injury can become persistent and difficult to treat but the molecular signaling responsible for its development remains poorly described. Here, we identify the neuronal stress sensor dual leucine zipper kinase (DLK; Map3k12) as a key molecule controlling the maladaptive pathways that lead to pain following injury. Genetic or pharmacological inhibition of DLK reduces mechanical hypersensitivity in a mouse model of neuropathic pain. Furthermore, DLK inhibition also prevents the spinal cord microgliosis that results from nerve injury and arises distant from the injury site. These striking phenotypes result from the control by DLK of a transcriptional program in somatosensory neurons regulating the expression of numerous genes implicated in pain pathogenesis, including the immune gene Csf1. Thus, activation of DLK is an early event, or even the master regulator, controlling a wide variety of pathways downstream of nerve injury that ultimately lead to chronic pain.


Subject(s)
Gliosis/genetics , Hyperalgesia/genetics , MAP Kinase Kinase Kinases/genetics , Neuralgia/genetics , Peripheral Nerve Injuries/genetics , Sensory Receptor Cells/enzymology , Animals , Disease Models, Animal , Female , Gene Expression Regulation , Gliosis/enzymology , Gliosis/pathology , Gliosis/prevention & control , Hyperalgesia/enzymology , Hyperalgesia/pathology , Hyperalgesia/prevention & control , MAP Kinase Kinase Kinases/deficiency , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Mice, Transgenic , Microglia/enzymology , Microglia/pathology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuralgia/enzymology , Neuralgia/pathology , Neuralgia/prevention & control , Peripheral Nerve Injuries/enzymology , Peripheral Nerve Injuries/pathology , Sciatic Nerve/enzymology , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Sensory Receptor Cells/pathology , Signal Transduction , Spinal Cord/enzymology , Spinal Cord/pathology , Touch , Transcription, Genetic
6.
Glia ; 66(5): 920-933, 2018 05.
Article in English | MEDLINE | ID: mdl-29350438

ABSTRACT

Human mesial temporal lobe epilepsy (MTLE) features subregion-specific hippocampal neurodegeneration and reactive astrogliosis, including up-regulation of the glial fibrillary acidic protein (GFAP) and down-regulation of glutamine synthetase (GS). However, the regional astrocytic expression pattern of GFAP and GS upon MTLE-associated neurodegeneration still remains elusive. We assessed GFAP and GS expression in strict correlation with the local neuronal number in cortical and hippocampal surgical specimens from 16 MTLE patients using immunohistochemistry, stereology and high-resolution image analysis for digital pathology and whole-slide imaging. In the cortex, GS-positive (GS+) astrocytes are dominant in all neuronal layers, with a neuron to GS+ cell ratio of 2:1. GFAP-positive (GFAP+) cells are widely spaced, with a GS+ to GFAP+ cell ratio of 3:1-5:1. White matter astrocytes, on the contrary, express mainly GFAP and, to a lesser extent, GS. In the hippocampus, the neuron to GS+ cell ratio is approximately 1:1. Hippocampal degeneration is associated with a reduction of GS+ astrocytes, which is proportional to the degree of neuronal loss and primarily present in the hilus. Up-regulation of GFAP as a classical hallmark of reactive astrogliosis does not follow the GS-pattern and is prominent in the CA1. Reactive alterations were proportional to the neuronal loss in the neuronal somatic layers (stratum pyramidale and hilus), while observed to a lesser extent in the axonal/dendritic layers (stratum radiatum, molecular layer). We conclude that astrocytic GS is expressed in the neuronal somatic layers and, upon neurodegeneration, is down-regulated proportionally to the degree of neuronal loss.


Subject(s)
Astrocytes/enzymology , Cerebral Cortex/enzymology , Epilepsy, Temporal Lobe/enzymology , Glutamate-Ammonia Ligase/metabolism , Neurons/enzymology , Adult , Astrocytes/pathology , Cell Death/physiology , Cerebral Cortex/pathology , Drug Resistant Epilepsy/enzymology , Drug Resistant Epilepsy/pathology , Drug Resistant Epilepsy/surgery , Epilepsy, Temporal Lobe/pathology , Epilepsy, Temporal Lobe/surgery , Female , Glial Fibrillary Acidic Protein/metabolism , Gliosis/enzymology , Gliosis/pathology , Humans , Immunohistochemistry , Male , Neurodegenerative Diseases/enzymology , Neurodegenerative Diseases/pathology , Neurons/pathology , White Matter/enzymology , White Matter/pathology
7.
Microb Pathog ; 111: 75-80, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28823791

ABSTRACT

Chagas disease is caused by the protozoan parasite Trypanosoma cruzi and causes severe cardiac and brain damage, leading to behavioral alterations in humans and animals. However, the mechanisms involved in memory impairment during T. cruzi infection remain unknown. It has long been recognized that the enzymatic activities of acetylcholinesterase (AChE) and Na+, K+-ATPase are linked with memory dysfunction during other trypanosomiasis. Thus, the aim of this study was to evaluate the involvement of cerebral AChE and Na+, K+-ATPase activities in the memory impairment during T. cruzi (Colombian strain) infection. A significant decrease on latency time during the inhibitory avoidance task was observed in animals infected by T. cruzi compared to uninfected animals, findings compatible to memory dysfunction. Moreover, the cerebral AChE activity increased, while the Na+, K+-ATPase decreased in T. cruzi infected compared to uninfected animals. Histopathology revealed mild to moderate multifocal gliosis in the cerebral cortex and light focal meningeal lymphoplasmacytic infiltrate, which may have contributed to memory loss. Based on these evidences, we can conclude that T. cruzi (Colombian strain) causes memory impairment in mice experimentally infected. Moreover, the changes in AChE and Na+, K+-ATPase activities may be considered a mechanism involved in disease pathogenesis.


Subject(s)
Acetylcholinesterase/metabolism , Central Nervous System Protozoal Infections/enzymology , Cerebral Cortex/enzymology , Memory Disorders/enzymology , Sodium-Potassium-Exchanging ATPase/metabolism , Trypanosoma cruzi/pathogenicity , Animals , Behavior, Animal , Brain/enzymology , Brain/parasitology , Brain/pathology , Central Nervous System Protozoal Infections/parasitology , Central Nervous System Protozoal Infections/pathology , Central Nervous System Protozoal Infections/psychology , Cerebral Cortex/parasitology , Cerebral Cortex/pathology , Chagas Disease , Disease Models, Animal , Female , Gliosis/enzymology , Gliosis/parasitology , Gliosis/pathology , Heart , Humans , Memory Disorders/parasitology , Memory Disorders/pathology , Memory Disorders/psychology , Mice , Trypanosomiasis/parasitology , Trypanosomiasis/psychology
8.
Orphanet J Rare Dis ; 12(1): 145, 2017 08 25.
Article in English | MEDLINE | ID: mdl-28841900

ABSTRACT

BACKGROUND: Niemann-Pick disease Type C1 (NPC1) is a rare progressive neurodegenerative disorder caused by mutations in the NPC1 gene. The pathological mechanisms, underlying NPC1 are not yet completely understood. Especially the contribution of glial cells and gliosis to the progression of NPC1, are controversially discussed. As an analysis of affected cells is unfeasible in NPC1-patients, we recently developed an in vitro model system, based on cells derived from NPC1-patient specific iPSCs. Here, we asked if this model system recapitulates gliosis, observed in non-human model systems and NPC1 patient post mortem biopsies. We determined the amount of reactive astrocytes and the regulation of the intermediate filaments GFAP and vimentin, all indicating gliosis. Furthermore, we were interested in the assembly and phosphorylation of these intermediate filaments and finally the impact of the activation of protein kinase C (PKC), which is described to ameliorate the pathogenic phenotype of NPC1-deficient fibroblasts, including hypo-phosphorylation of vimentin and cholesterol accumulation. METHODS: We analysed glial cells derived from NPC1 patient specific induced pluripotent stem cells, carrying different NPC1 mutations. The amount of reactive astrocytes was determined by means of immuncytochemical stainings and FACS-analysis. Semi-quantitative western blot was used to determine the amount of phosphorylated GFAP and vimentin. Cholesterol accumulation was analysed by Filipin staining and quantified by Amplex Red Assay. U18666A was used to induce NPC1 phenotype in unaffected cells of the control cell line. Phorbol 12-myristate 13-acetate (PMA) was used to activate PKC. RESULTS: Immunocytochemical detection of GFAP, vimentin and Ki67 revealed that NPC1 mutant glial cells undergo gliosis. We found hypo-phosphorylation of the intermediate filaments GFAP and vimentin and alterations in the assembly of these intermediate filaments in NPC1 mutant cells. The application of U18666A induced not only NPC1 phenotypical accumulation of cholesterol, but characteristics of gliosis in glial cells derived from unaffected control cells. The application of phorbol 12-myristate 13-acetate, an activator of protein kinase C resulted in a significantly reduced number of reactive astrocytes and further characteristics of gliosis in NPC1-deficient cells. Furthermore, it triggered a restoration of cholesterol amounts to level of control cells. CONCLUSION: Our data demonstrate that glial cells derived from NPC1-patient specific iPSCs undergo gliosis. The application of U18666A induced comparable characteristics in un-affected control cells, suggesting that gliosis is triggered by hampered function of NPC1 protein. The activation of protein kinase C induced an amelioration of gliosis, as well as a reduction of cholesterol amount. These results provide further support for the line of evidence that gliosis might not be only a secondary reaction to the loss of neurons, but might be a direct consequence of a reduced PKC activity due to the phenotypical cholesterol accumulation observed in NPC1. In addition, our data support the involvement of PKCs in NPC1 disease pathogenesis and suggest that PKCs may be targeted in future efforts to develop therapeutics for NPC1 disease.


Subject(s)
Gliosis/metabolism , Induced Pluripotent Stem Cells/metabolism , Neuroglia/metabolism , Niemann-Pick Disease, Type C/enzymology , Niemann-Pick Disease, Type C/metabolism , Protein Kinase C/metabolism , Cell Line , Glial Fibrillary Acidic Protein/metabolism , Gliosis/enzymology , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/enzymology , Neuroglia/enzymology , Niemann-Pick Disease, Type C/genetics , Protein Kinase C/genetics , Stem Cells/metabolism , Vimentin/metabolism
9.
Biochem Biophys Res Commun ; 487(1): 134-139, 2017 05 20.
Article in English | MEDLINE | ID: mdl-28400047

ABSTRACT

Citrullination is an important posttranslational modification that occurs during retinal gliosis. We examined the expression of peptidyl arginine deiminases (PADs) to identify the PADs that mediate citrullination in a model of alkali-induced retinal gliosis. Mouse corneas were exposed to 1.0 N NaOH and posterior eye tissue from injured and control uninjured eyes was evaluated for transcript levels of various PADs by reverse-transcription polymerase chain reaction (RT-PCR), and quantitative RT-PCR (qPCR). Retinas were also subjected to immunohistochemistry (IHC) for glial fibrillary acidic protein (GFAP), citrullinated species, PAD2, and PAD4 and tissue levels of GFAP, citrullinated species, and PAD4 were measured by western blots. In other experiments, the PAD4 inhibitor streptonigrin was injected intravitreally into injured eyes ex vivo to test inhibitory activity in an organ culture system. We found that uninjured retina and choroid expressed Pad2 and Pad4 transcripts. Pad4 transcript levels increased by day 7 post-injury (p < 0.05), whereas Pad2 levels did not change significantly (p > 0.05) by qPCR. By IHC, PAD2 was expressed in uninjured eyes along ganglion cell astrocytes, but in injured retina PAD2 was downregulated at 7 days. On the other hand, PAD4 showed increased staining in the retina upon injury revealing a pattern that overlapped with filamentous GFAP staining in Müller glial processes by 7 days. Injury-induced citrullination and soluble GFAP protein levels were reduced by PAD4 inhibition in western blot experiments of organ cultures. Together, our findings for the first time identify PAD4 as a novel injury-inducible druggable target for retinal gliosis.


Subject(s)
Eye Burns/enzymology , Gliosis/enzymology , Hydrolases/metabolism , Retina/enzymology , Retina/injuries , Retinal Diseases/enzymology , Animals , Arginine/metabolism , Burns, Chemical/enzymology , Citrulline/metabolism , Eye Burns/chemically induced , Eye Burns/complications , Female , Gliosis/chemically induced , Male , Mice , Protein-Arginine Deiminase Type 4 , Retinal Diseases/chemically induced , Retinal Diseases/etiology , Sodium Hydroxide
10.
PLoS One ; 10(7): e0132604, 2015.
Article in English | MEDLINE | ID: mdl-26161952

ABSTRACT

Microgliosis is a major hallmark of Alzheimer's disease (AD) brain pathology. Aß peptide is hypothesized to act as a stimulus for microglia leading to activation of non-receptor tyrosine kinases and subsequent secretion of pro-inflammatory cytokines. Therefore, the signaling pathways mediating microglial activation may be important therapeutic targets of anti-inflammatory therapy for AD. Four novel compounds were chosen after high throughput screening kinase activity assays determined them as potential Lyn kinase inhibitors. Their kinase inhibitory and anti-inflammatory effect on Aß-stimulated activation was assessed using the murine microglial cell line, BV2. Cells were treated with the compounds to determine effects on active, phosphorylated levels of Src family kinases, Src and Lyn, as well as MAP kinases ERK, JNK and p38. Only one compound, LDDN-0003499, produced a dose dependent decrease in basal levels of active, phosphorylated Src and Lyn in the BV2 cells. LDDN-0003499 treatment also attenuated the Aß-stimulated increase in active, phosphorylated levels of Lyn/Src and TNFα and IL-6 secretion. This study identifies a novel small molecule Src family tyrosine kinase inhibitor with anti-inflammatory effects in response to Aß stimulation of microglia. Further in vitro/in vivo characterization of LDDN-0003499 as well as structural modification may provide a new tool for attenuating microglial-mediated brain inflammatory conditions such as that occurring in AD.


Subject(s)
Gliosis/pathology , src-Family Kinases/antagonists & inhibitors , Administration, Oral , Amyloid beta-Peptides/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Caco-2 Cells , Cell Line , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/metabolism , Gliosis/enzymology , Humans , Interleukin-6/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Microsomes/drug effects , Microsomes/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Kinase Inhibitors/pharmacology , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , src-Family Kinases/metabolism
11.
J Mol Neurosci ; 57(3): 452-62, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26080748

ABSTRACT

Extracellular ATP (eATP) acts as a danger-associated molecular pattern which induces reactive response of astrocytes after brain insult, including morphological remodeling of astrocytes, proliferation, chemotaxis, and release of proinflammatory cytokines. The responses induced by eATP are under control of ecto-nucleotidases, which catalyze sequential hydrolysis of ATP to adenosine. In the mammalian brain, ecto-nucleotidases comprise three enzyme families: ecto-nucleoside triphosphate diphosphohydrolases 1-3 (NTPDase1-3), ecto-nucleotide pyrophosphatase/phospodiesterases 1-3 (NPP1-3), and ecto-5'-nucleotidase (eN), which crucially determine ATP/adenosine ratio in the pericellular milieu. Altered expression of ecto-nucleotidases has been demonstrated in several experimental models of human brain dysfunctions. In the present study, we have explored the pattern of NTPDase1-3, NPP1-3, and eN expression by cultured cortical astrocytes challenged with 1 mmol/L ATP (eATP). At the transcriptional level, eATP upregulated expression of NTPDase1, NTPDase2, NPP2, and eN, while, at translational and functional levels, these were paralleled only by the induction of NTPDase2 and eN. Additionally, eATP altered membrane topology of eN, from clusters localized in membrane domains to continuous distribution along the cell membrane. Our results suggest that eATP, by upregulating NTPDase2 and eN and altering the enzyme membrane topology, affects local kinetics of ATP metabolism and signal transduction that may have important roles in the process related to inflammation and reactive gliosis.


Subject(s)
5'-Nucleotidase/biosynthesis , Adenosine Triphosphate/pharmacology , Astrocytes/drug effects , Cell Membrane/enzymology , Nerve Tissue Proteins/biosynthesis , Phosphoric Diester Hydrolases/biosynthesis , Pyrophosphatases/biosynthesis , 5'-Nucleotidase/genetics , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Animals , Astrocytes/metabolism , Cell Division , Cell Membrane/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Enzyme Induction/drug effects , Gliosis/enzymology , Nerve Tissue Proteins/genetics , Phosphoric Diester Hydrolases/genetics , Primary Cell Culture , Protein Biosynthesis/drug effects , Pyrophosphatases/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription, Genetic/drug effects , Up-Regulation/drug effects
12.
Graefes Arch Clin Exp Ophthalmol ; 253(9): 1503-13, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25921391

ABSTRACT

Retinopathy of prematurity (ROP) is a leading cause of childhood blindness where vascular abnormality and retinal dysfunction are reported. We showed earlier that genetic deletion of aldose reductase (AR), the rate-limiting enzyme in the polyol pathway, reduced the neovascularization through attenuating oxidative stress induction in the mouse oxygen-induced retinopathy (OIR) modeling ROP. In this study, we further investigated the effects of AR deficiency on retinal neurons in the mouse OIR. Seven-day-old wild-type and AR-deficient mice were exposed to 75 % oxygen for 5 days and then returned to room air. Electroretinography was used to assess the neuronal function at postnatal day (P) 30. On P17 and P30, retinal cytoarchitecture was examined by morphometric analysis and immunohistochemistry for calbindin, protein kinase C alpha, calretinin, Tuj1, and glial fibrillary acidic protein. In OIR, attenuated amplitudes and delayed implicit time of a-wave, b-wave, and oscillatory potentials were observed in wild-type mice, but they were not significantly changed in AR-deficient mice. The morphological changes of horizontal, rod bipolar, and amacrine cells were shown in wild-type mice and these changes were partly preserved with AR deficiency. AR deficiency attenuated the Müller cell gliosis induced in OIR. Our observations demonstrated AR deficiency preserved retinal functions in OIR and AR deficiency could partly reduce the extent of retinal neuronal histopathology. These findings suggested a therapeutic potential of AR inhibition in ROP treatment with beneficial effects on the retinal neurons.


Subject(s)
Aldehyde Reductase/deficiency , Disease Models, Animal , Gliosis/prevention & control , Retinal Neurons/enzymology , Retinopathy of Prematurity/prevention & control , Animals , Animals, Newborn , Calbindin 2/metabolism , Calbindins/metabolism , Electroretinography , Glial Fibrillary Acidic Protein , Gliosis/enzymology , Immunohistochemistry , Mice , Nerve Tissue Proteins/metabolism , Protein Kinase C-alpha/metabolism , Retina/physiopathology , Retinopathy of Prematurity/enzymology , Tubulin/metabolism
13.
Glia ; 63(3): 497-511, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25421817

ABSTRACT

Microglial cells are the resident macrophages of the central nervous system. Their function is essential for neuronal tissue homeostasis. After inflammatory stimuli, microglial cells become activated changing from a resting and highly ramified cell shape to an amoeboid-like morphology. These morphological changes are associated with the release of proinflammatory cytokines and glutamate, as well as with high phagocytic activity. The acquisition of such phenotype has been associated with activation of cytoplasmic tyrosine kinases, including those of the Src family (SFKs). In this study, using both in vivo and in vitro inflammation models coupled to FRET-based time-lapse microscopy, lentiviruses-mediated shRNA delivery and genetic gain-of-function experiments, we demonstrate that among SFKs c-Src function is necessary and sufficient for triggering microglia proinflammatory signature, glutamate release, microglia-induced neuronal loss, and phagocytosis. c-Src inhibition in retinal neuroinflammation experimental paradigms consisting of intravitreal injection of LPS or ischemia-reperfusion injury significantly reduced microglia activation changing their morphology to a more resting phenotype and prevented neuronal apoptosis. Our data demonstrate an essential role for c-Src in microglial cell activation.


Subject(s)
Microglia/enzymology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Animals , Apoptosis/physiology , CSK Tyrosine-Protein Kinase , Cell Line , Cells, Cultured , Chickens , Gliosis/enzymology , Gliosis/pathology , Glutamic Acid/metabolism , HEK293 Cells , Humans , Inflammation/enzymology , Inflammation/pathology , Ischemia/enzymology , Ischemia/pathology , Lipopolysaccharides , Male , Mice , Microglia/pathology , Neurons/physiology , Phagocytosis/physiology , Rats, Wistar , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Retinal Neurons/pathology , Retinal Neurons/physiology , Tumor Necrosis Factor-alpha/metabolism , src-Family Kinases/metabolism
14.
Brain Res ; 1551: 45-58, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24440774

ABSTRACT

Reactive astrogliosis is an essential feature of astrocytic response to all forms of central nervous system (CNS) injury and disease, which may benefit or harm surrounding neural and non-neural cells. Despite extensive study, its molecular triggers remain largely unknown in term of ischemic stroke. In the current study we investigated the role p38 mitogen-activated protein kinase (MAPK) in astrogliosis both in vitro and in vivo. In a mouse model of middle cerebral artery occlusion (MCAO), p38 MAPK activation was observed in the glia scar area, along with increased glial fibrillary acidic protein (GFAP) expression. In primary astrocyte cultures, hypoxia and scratch injury-induced astrogliosis was attenuated by both p38 inhibition and knockout of p38 MAPK. In addition, both knockout and inhibition of p38 MAPK also reduced astrocyte migration, but did not affect astrocyte proliferation. In a mouse model of permanent MCAO, no significant difference in motor function recovery and lesion volume was observed between conditional GFAP/p38 MAPK knockout mice and littermates. While a significant reduction of astrogliosis was observed in the GFAP/p38 knockout mice compared with the littermates. Our findings suggest that p38 MAPK signaling pathway plays an important role in the ischemic stroke-induced astrogliosis and thus may serve as a novel target to control glial scar formation.


Subject(s)
Astrocytes/enzymology , Brain Ischemia/enzymology , Gliosis/enzymology , Stroke/enzymology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Astrocytes/metabolism , Brain Ischemia/complications , Brain Ischemia/metabolism , Cerebral Cortex/enzymology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Gliosis/etiology , Gliosis/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Stroke/complications , Stroke/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
J Neuropathol Exp Neurol ; 72(11): 1000-8, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24128682

ABSTRACT

Rasmussen encephalitis (RE) is a rare neurologic disorder of childhood characterized by unihemispheric inflammation, progressive neurologic deficits, and intractable focal epilepsy. The pathogenesis of RE is still enigmatic. Adenosine is a key endogenous signaling molecule with anticonvulsive and anti-inflammatory effects, and our previous work demonstrated that dysfunction of the adenosine kinase (ADK)-adenosine system and astrogliosis are the hallmarks of epilepsy. We hypothesized that the epileptogenic mechanisms underlying RE are related to changes in ADK expression and that those changes might be associated with the development of epilepsy in RE patients. Immunohistochemistry was used to examine the expression of ADK and glial fibrillary acidic protein in surgically resected human epileptic cortical specimens from RE patients (n = 12) and compared with control cortical tissues (n = 6). Adenosine kinase expression using Western blot and enzymatic activity for ADK were assessed in RE versus control samples. Focal astrogliosis and marked expression of ADK were observed in the lesions of RE. Significantly greater ADK expression in RE versus controls was demonstrated by Western blot, and greater enzymatic activity for ADK was demonstrated using an enzyme-coupled bioluminescent assay. These results suggest that upregulation of ADK is a common pathologic hallmark of RE and that ADK might be a target in the treatment of epilepsy associated with RE.


Subject(s)
Adenosine Kinase/metabolism , Cerebral Cortex/enzymology , Encephalitis/enzymology , Up-Regulation , Adolescent , Astrocytes/metabolism , Astrocytes/pathology , Cerebral Cortex/pathology , Child , Child, Preschool , Encephalitis/complications , Encephalitis/pathology , Female , Gliosis/complications , Gliosis/enzymology , Gliosis/pathology , Humans , Male
16.
Neurosci Lett ; 548: 84-9, 2013 Aug 26.
Article in English | MEDLINE | ID: mdl-23727388

ABSTRACT

The aim of this study was to determine the role of NADPH-cytochrome P450 reductase (CPR) and CPR-dependent enzymes in neural stem cell (NSC) genesis in the brain. A mouse model with globally suppressed Cpr gene expression (Cpr-low mouse) was studied for this purpose. Cpr-low and wild-type (WT) mice were compared immunohistochemically for the expression of markers of cell proliferation (Ki67), immature neurons (doublecortin, DCX), oligodendrocytes (oligodendrocyte transcription factor 2, OLIG2), and astrocytes (glial fibrillary acidic protein, GFAP) in the SVZ, and for the in vitro capability of their SVZ cells to form neurospheres and differentiate into astrocytes. We found that the abundance of SVZ cells that are positive for Ki67 or GFAP expression, but not the abundance of SVZ cells that are positive for DCX and OLIG2 expression, was significantly increased in Cpr-low mice, at various ages, compared with WT mice. Furthermore, extents of astrocyte differentiation and growth, but not neurosphere formation, from SVZ cells of the Cpr-low mice were significantly increased, compared with WT mice. These results suggest that CPR and CPR-dependent enzymes play a role in suppressing astrocytosis in the SVZ of adult mice.


Subject(s)
Cerebral Ventricles/enzymology , Cerebral Ventricles/pathology , Cytochrome P-450 Enzyme System/metabolism , Gene Expression Regulation, Enzymologic , Gliosis/enzymology , Gliosis/pathology , Neural Stem Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Cytochrome P-450 Enzyme System/genetics , Doublecortin Protein , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/pathology
17.
Acta Ophthalmol ; 90(7): e560-7, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22998629

ABSTRACT

PURPOSE: Our previous studies suggested that CYP46A1 and 24S-hydroxycholesterol (24SOH) may be associated with glaucoma. Loss of CYP46A1-expressing retinal ganglion cells is involved in the activation of glia, and therefore possibly in the disbalance of cholesterol. In this context, the purpose of our present work was to emphasize the glial and longitudinal CYP46A1 expression after an interventional glaucoma-related stress triggered by elevated intraocular pressure (IOP). METHODS: Sprague-Dawley rats were submitted to laser photocoagulation of the trabecular meshwork, limbus and episcleral veins in one eye to induce elevated IOP. Rats were euthanized at days 3, 14, 30 and 60 (n = 10 per time-point), and 24SOH was measured in plasma and retina by gas chromatography-mass spectrometry. CYP46A1 was quantified by Western blotting. Glial activation was assessed by glial fibrillary acidic protein immunoreactivity in Western blots and retinal cryosections. RESULTS: Sustained high IOP was observed in experimental eyes from day 1 to day 21. Plasma MCP-1 and ICAM-1, quantified using multiplex assay kits, were increased at day 3. Glial activation was observed bilaterally at all time-points, at lower levels in contralateral eyes than in experimental eyes. In experimental retinas, CYP46A1 expression showed a transient increase at day 3 and then returned to baseline. Plasma and retinal 24SOH peaked at day 14 and 30, respectively. CONCLUSIONS: These data show that CYP46A1 expression was induced early in response to retinal stress but remained constant at late time-points, reinforcing the constitutive role of CYP46A1 in maintaining cholesterol balance in neuronal tissues.


Subject(s)
Gliosis/blood , Hydroxycholesterols/blood , Intraocular Pressure , Neuroglia/metabolism , Ocular Hypertension/blood , Animals , Blotting, Western , Cholesterol 24-Hydroxylase , Cytokines/blood , Disease Models, Animal , Gas Chromatography-Mass Spectrometry , Glial Fibrillary Acidic Protein/metabolism , Gliosis/enzymology , Homeostasis , Intercellular Signaling Peptides and Proteins/blood , Ocular Hypertension/enzymology , Rats , Rats, Sprague-Dawley , Retina/metabolism , Steroid Hydroxylases/metabolism
18.
Biol Chem ; 393(5): 355-67, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22505518

ABSTRACT

Kallikrein-related peptidase 6 (KLK6) is a trypsin-like serine protease upregulated at sites of central nervous system (CNS) injury, including de novo expression by reactive astrocytes, yet its physiological actions are largely undefined. Taken with recent evidence that KLK6 activates G-protein-coupled protease-activated receptors (PARs), we hypothesized that injury-induced elevations in KLK6 contribute to the development of astrogliosis and that this occurs in a PAR-dependent fashion. Using primary murine astrocytes and the Neu7 astrocyte cell line, we show that KLK6 causes astrocytes to transform from an epitheliod to a stellate morphology and to secrete interleukin 6 (IL-6). By contrast, KLK6 reduced expression of glial fibrillary acidic protein (GFAP). The stellation-promoting activities of KLK6 were shown to be dependent on activation of the thrombin receptor, PAR1, as a PAR1-specific inhibitor, SCH79797, blocked KLK6-induced morphological changes. The ability of KLK6 to promote astrocyte stellation was also shown to be linked to activation of protein kinase C (PKC). These studies indicate that KLK6 is positioned to serve as a molecular trigger of select physiological processes involved in the development of astrogliosis and that this is likely to occur at least in part by activation of the G-protein-coupled receptor, PAR1.


Subject(s)
Gliosis/enzymology , Gliosis/pathology , Kallikreins/metabolism , Animals , Astrocytes/enzymology , Astrocytes/metabolism , Astrocytes/pathology , Brain/enzymology , Brain/metabolism , Brain/pathology , Carrier Proteins/metabolism , Cell Proliferation , DNA-Binding Proteins , Gene Expression Regulation , Glial Fibrillary Acidic Protein/metabolism , Gliosis/metabolism , Humans , Hypertrophy , Interleukin-6/metabolism , Mice , Protein Kinase C/metabolism , Receptor, PAR-1/metabolism , Spinal Cord/enzymology , Spinal Cord/metabolism , Spinal Cord/pathology
19.
Brain Res ; 1427: 1-9, 2012 Jan 03.
Article in English | MEDLINE | ID: mdl-22050961

ABSTRACT

Activation of glial cells and the intracellular ERK signaling pathway plays an important role in the development and maintenance of neuropathic pain. As well as neurons, glial cell membranes also express α2-adrenergic receptors, but the effects of selective activation of these receptors on glial cell activation induced by neuropathic pain have yet to be clarified. We investigated the effects of intraperitoneal (IP) injections of tolerable doses of dexmedetomidine (DEX), a highly selective agonist of α2-adrenergic receptors, on activation of spinal dorsal root glial cells and the intracellular ERK signaling pathway induced by neuropathic pain. Adult rats that underwent partial sciatic nerve ligation (PNSL) were treated with repeated IP injections of DEX 20 µg/kg or 40 µg/kg, and their thermal and mechanical hyperalgesia thresholds were measured. The distribution and morphological changes of microglias and astrocytes were observed by immunofluorescence. Western blot was used to detect changes of glial fibrillary acid protein (GFAP) and pERK expression. Repeated IP injections of DEX 40 µg/kg for 7 or 14 days markedly reduced the thermal and mechanical hyperalgesia induced by PSNL. In addition, DEX 20 µg/kg for 14 days and 40 µg/kg for 7 days also significantly inhibited PSNL-induced activation of pERK in the spinal dorsal horn. Thus, repeated IP injections of DEX can markedly relieve the hyperalgesia of neuropathic pain in rats. The analgesic effect of DEX may be attributed to its inhibition of glial cell hypertrophy in the spinal dorsal horn and activation of the intracellular ERK signaling pathway.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Dexmedetomidine/pharmacology , Gliosis/drug therapy , Gliosis/enzymology , MAP Kinase Signaling System/drug effects , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/enzymology , Animals , Disease Models, Animal , Gliosis/pathology , MAP Kinase Signaling System/physiology , Neuroglia/drug effects , Neuroglia/enzymology , Neuroglia/pathology , Peripheral Nervous System Diseases/pathology , Rats , Rats, Sprague-Dawley
20.
Neurosci Res ; 71(3): 210-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21802455

ABSTRACT

In our previous study, peripheral inflammatory stimulation evoked production of macrophage migration inhibitory factor (MIF) in the spinal cord and found spinal microglia are the major source of MIF in this context. Given the contribution of the activated-microglia to the inflammatory neuropathy plus the role for upregulated COX 2 expression and PGE(2) production in the severity of clinical manifestations of these neuroinflammatory conditions, we herein tested the hypothesis that in vitro MIF stimulation to spinal microglia could result in an activation of COX 2-PGE(2) system by MIF-CD74 interaction. We found MIF played roles in evoking COX 2 mRNA and protein expression in a dose-dependent manner correspondingly in changes in PGE(2) level in the cultured rat microglia, but these changes could be inhibited by genetic deletion of CD74. Finally, MIF-induced COX 2-PGE(2) activation could be blocked by selective inhibitors of p44/p42 and p38 MAPKs. These data highlight MIF/CD74 interaction induces upregulation of COX 2 expression and PGE(2) secretion in primary rodent microglia, and further this effect is associated with downstream activation of p38 and p44/p42 signaling cascades, and favors the role of MIF as a novel pathway for microglia-associated neuroinflammation.


Subject(s)
Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Gliosis/metabolism , Intramolecular Oxidoreductases/physiology , Macrophage Migration-Inhibitory Factors/physiology , Microglia/metabolism , Myelitis/metabolism , Animals , Animals, Newborn , Antigens, Differentiation, B-Lymphocyte/metabolism , Antigens, Differentiation, B-Lymphocyte/physiology , Cyclooxygenase 2/genetics , Dinoprostone/physiology , Enzyme Activation/genetics , Gliosis/enzymology , Gliosis/pathology , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class II/physiology , Microglia/enzymology , Microglia/pathology , Myelitis/enzymology , Myelitis/pathology , Neural Pathways/pathology , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...