Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(20)2020 Oct 16.
Article in English | MEDLINE | ID: mdl-33081303

ABSTRACT

In ischemic stroke, neutrophils infiltrate damaged brain tissue immediately following the ischemic insult and aggravate inflammation via various mechanisms which include neutrophil extracellular traps (NETs) formation. In the present study, we showed that adenosine triphosphate (ATP), a DAMP molecule, accumulates in the brain and induces NETosis in brain parenchyma and in circulating neutrophils (PMNs) isolated from a murine model of stroke induced by middle cerebral artery occlusion (MCAO). Expression of peptidylarginine deiminase-4 (PAD4), which induces citrullination of histones H3 (CitH3) and initiates NETosis, was significantly enhanced in brain parenchyma and blood PMNs following MCAO. ATP or BzATP (a prototypic P2X7R agonist) significantly enhanced the inductions of PAD4 and CitH3 in a P2X7R-dependent manner and intracellular Ca2+ influx, PKCα activation, and NADPH oxidase-dependent reactive oxygen species (ROS) production play critical roles in this ATP-P2X7R-mediated NETosis. In our MCAO animal model, NETosis was markedly suppressed by treatment with apyrase, an enzyme hydrolyzing ATP, but enhanced by co-treatment of BzATP, confirming ATP-P2X7R-mediated NETosis. Since ATP not only induced NETosis but was also extruded after NETosis, our results indicate that ATP accumulated in the ischemic brain induces NETosis, mediating a cross-talk linking NETosis with neuronal damage that might aggravate inflammation and brain damage.


Subject(s)
Adenosine Triphosphate/metabolism , Extracellular Traps/metabolism , Infarction, Middle Cerebral Artery/metabolism , Neutrophils/metabolism , Animals , Calcium/metabolism , Cells, Cultured , Male , Protein Kinase C/metabolism , Protein-Arginine Deiminase Type 4/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptors, Purinergic P2X7/metabolism
2.
Sci Rep ; 10(1): 16656, 2020 10 07.
Article in English | MEDLINE | ID: mdl-33028854

ABSTRACT

Nerve injury-induced protein 1 (Ninjurin 1, Ninj1) is a cell adhesion molecule responsible for cell-to-cell interactions between immune cells and endothelial cells. In our previous paper, we have shown that Ninj1 plays an important role in the infiltration of neutrophils in the postischemic brain and that the dodecamer peptide harboring the Ninj1 N-terminal adhesion motif (N-NAM, Pro26-Asn37) inhibits infiltration of neutrophils in the postischemic brain and confers robust neuroprotective and anti-inflammatory effects. In the present study, we examinedt the pro-angiogenic effect of N-NAM using human umbilical vein endothelial cells (HUVECs) and rat MCAO (middle cerebral artery occlusion) model of stroke. We found that N-NAM promotes proliferation, migration, and tube formation of HUVECs and demonstrate that the suppression of endogenous Ninj1 is responsible for the N-NAM-mediated pro-angiogenic effects. Importantly, a pull-down assay revealed a direct binding between exogenously delivered N-NAM and endogenous Ninj1 and it is N-terminal adhesion motif dependent. In addition, N-NAM activated the Ang1-Tie2 and AKT signaling pathways in HUVECs, and blocking those signaling pathways with specific inhibitors suppressed N-NAM-induced tube formation, indicating critical roles of those signaling pathways in N-NAM-induced angiogenesis. Moreover, in a rat MCAO model, intranasal administration of N-NAM beginning 4 days post-MCAO (1.5 µg daily for 3 days) augmented angiogenesis in the penumbra of the ipsilateral hemisphere of the brain and significantly enhanced total vessel lengths, vessel densities, and pro-angiogenic marker expression. These results demonstrate that the 12-amino acid Ninj1 peptide, which contains the N-terminal adhesion motif of Ninj1, confers pro-angiogenic effects and suggest that those effects might contribute to its neuroprotective effects in the postischemic brain.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Cell Adhesion Molecules, Neuronal/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Ischemic Stroke/drug therapy , Neovascularization, Physiologic/drug effects , Nerve Growth Factors/pharmacology , Neuroprotective Agents/pharmacology , Angiogenesis Inducing Agents/therapeutic use , Animals , Brain/drug effects , Brain/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Ischemic Stroke/metabolism , Nerve Growth Factors/metabolism , Neuroprotective Agents/therapeutic use , Neutrophils/drug effects , Neutrophils/metabolism , Rats , Signal Transduction/drug effects
3.
Biochem Biophys Res Commun ; 524(2): 371-377, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32005517

ABSTRACT

Osteopontin (OPN) is a phosphorylated glycoprotein expressed in various tissues, including brain, and mediates a wide range of cellular activities. In our previous studies, we reported recombinant OPN and RGD and SLAY-containing OPN-peptide icosamer (OPNpt20) exhibited robust neuroprotective activities in an animal model of transient focal ischemia, and attributed these effects to the anti-inflammatory, pro-angiogenic, and phagocytic functions of OPNpt20. In the present study, we truncated OPNpt20 to 13 or 7 amino acid peptides containing RGD (R) and/or SLAY (S) motif (OPNpt13RS, OPNpt7R, OPNpt7RS, and OPNpt7S) and their cell motility and migration inducing activities were examined in BV2 cells (a microglia cell line). All four peptides significantly enhanced BV2 cell motility and migration, but OPNpt7R, an RGD-containing 7-amino-acid OPN peptide (VPNGRGD), was found to be most potent and its potency was comparable to OPNpt20. Phagocytic activity and F-actin polymerization were also significantly enhanced in OPNpt7R-treated BV2 cells. Importantly, studies using two mutant peptides (OPNpt7R-RAA and OPNpt7R-RAD, wherein RGD in OPNpt7R was replaced with RAA or RAD, respectively) revealed that all these effects of OPNpt7R, motility, migration, F-actin polymerization, and phagocytosis induction, were RGD-dependent. Furthermore, the Erk, Fak, and Akt signaling pathways appeared to be involved in the induction of phagocytic activity by OPNpt7R. Co-treating cells with OPNpt7R and D98059 or wortmannin (pharmacological inhibitors of Erk and Akt, respectively) significantly suppressed OPNpt7R-mediated phagocytosis induction. These results indicate the RGD-containing OPN heptamer OPNpt7R triggers microglial motility, migration, and phagocytic activity and that the RGD motif plays a critical role in these activities.


Subject(s)
Microglia/drug effects , Oligopeptides/pharmacology , Osteopontin/pharmacology , Phagocytosis/drug effects , Animals , Cell Line , Cell Movement/drug effects , Mice , Microglia/immunology , Oligopeptides/chemistry , Osteopontin/chemistry
4.
Acta Neuropathol Commun ; 7(1): 94, 2019 06 10.
Article in English | MEDLINE | ID: mdl-31177989

ABSTRACT

It has been reported that neutrophil extracellular traps (NETs) play important roles in non-infectious diseases. In ischemic stroke, neutrophils infiltrate damaged brain tissue soon after injury and aggravate inflammation. Using a rat permanent MCAO model, we showed citrullinated histone H3+ (CitH3, a marker of NETosis) induction in neutrophils in leptomeninges and in peripheral blood soon after MCAO. Entry of CitH3+ cells occurred through leptomeninges after 6 h of MCAO and these cells were observed in cerebral cortex from 12 h and subsequently in striatum. It is interesting to note that CitH3+ induction began in circulating neutrophils before they migrated to brain parenchyma and they were detected as intact or lysed form. High mobility group box 1 (HMGB1), a danger associated molecular pattern (DAMP) molecule, was accumulated massively in serum after permanent MCAO and plays a critical role in CitH3 inductions in neutrophils in brain parenchyma and in peripheral blood. Both the all-thiol and disulfide types of HMGB1 induced CitH3 via their specific receptors, CXCR4 and TLR4, respectively. Importantly, HMGB1 not only induced NETosis but was included as a part of the extruded NETs, and contribute to NETosis-mediated neuronal death. Therefore, it would appear a vicious cycle exists between neuronal cell death and NETosis and HMGB1 mediates detrimental effects exerted by this cycle. When NETosis was suppressed by a PAD inhibitor in MCAO animals, delayed immune cell infiltrations were markedly suppressed and damages in blood vessels were significantly mitigated. The study shows NETosis with the involvement of HMGB1 as a mediator in a vicious cycle aggravates inflammation and subsequent damage in the ischemic brain.


Subject(s)
Brain Ischemia/metabolism , Brain/metabolism , Extracellular Traps/metabolism , HMGB1 Protein/administration & dosage , Neutrophils/metabolism , Administration, Intravenous , Animals , Brain/drug effects , Brain/pathology , Brain Ischemia/pathology , Cells, Cultured , Extracellular Traps/drug effects , Male , Neutrophils/drug effects , Rats , Rats, Sprague-Dawley
5.
Neurotherapeutics ; 16(2): 523-537, 2019 04.
Article in English | MEDLINE | ID: mdl-30680637

ABSTRACT

Ethyl pyruvate (EP) is a simple aliphatic ester of pyruvic acid and has been shown to have protective properties, which have been attributed to its anti-inflammatory, anti-oxidative, and anti-apoptotic functions. In an effort to develop better derivatives of EP, we previously synthesized DEOPA (N,N-diethyl-2-oxopropanamide, a novel isoster of EP) which has greater neuroprotective effects than EP, probably due to its anti-inflammatory and anti-excitotoxic effects. In the present study, we synthesized 3 DEOPA derivatives, in which its diethylamino group was substituted with diisopropylamino, dipropylamino, or diisobutylamino groups. Among them, DIPOPA (N,N-diisopropyl-2-oxopropanamide) containing diisopropylamino group had a greater neuroprotective effect than DEOPA or EP when administered intravenously to a rat middle cerebral artery occlusion (MCAO) model at 9 h after MCAO. Furthermore, DIPOPA had a wider therapeutic window than DEOPA and a marked reduction of infarct volume was accompanied by greater neurological and behavioral improvements. In particular, DIPOPA exerted robust anti-inflammatory effects, as evidenced by marked suppressions of microglia activation and neutrophil infiltration in the MCAO model, in microglial cells, and in neutrophil-endothelial cocultures at lower concentration, and did so more effectively than DEOPA. In particular, DIPOPA remarkably suppressed neutrophil infiltration into brain parenchyma, and this effect was attributed to the expressional inhibitions of cell adhesion molecules in neutrophils of brain parenchyma and in circulating neutrophils via NF-κB inhibition. Together, these results indicate the robust neuroprotective effects of DIPOPA are attributable to its anti-inflammatory effects and suggest that DIPOPA offers a potential therapeutic means of ameliorating cerebral ischemic injury and other inflammation-related pathologies.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Brain Ischemia/drug therapy , Infarction, Middle Cerebral Artery/drug therapy , Neuroprotective Agents/therapeutic use , Pyruvates/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacology , Male , Neuroprotective Agents/pharmacology , Pyruvates/pharmacology , Rats , Rats, Sprague-Dawley
6.
Anim Cells Syst (Seoul) ; 22(6): 429-437, 2018.
Article in English | MEDLINE | ID: mdl-30533266

ABSTRACT

Gastrodin (GAS) is a predominant bioactive constituent of the Chinese herbal medicine Tianma (Gastrodia elata Blume). Many authors have reported GAS has the beneficial effect on diverse diseases of the CNS, including epilepsy, Alzheimer's disease, Parkinson's disease, and cerebral ischemia. Here, we report GAS exhibited a robust neuroprotective effect in an Sprague-Dawley rat model of stroke (transient middle cerebral artery occlusion, tMCAO), and show that the underlying molecular mechanism involves its protective effect against Zn2+-toxicity and its anti-oxidative effects in astrocytes. Intraperitoneal administration of GAS (40 mg/kg) after MCAO reduced mean infarct volume to 30.1 ± 5.9% of that of MCAO controls and this neuroprotective effect was accompanied by neurological function recoveries which was measured using modified neurological severity score (mNSS). Interestingly, GAS induced up-regulation and nuclear translocation of Nrf2, and subsequently increased the expressions of anti-oxidative genes, such as, HO-1 and GCLM, in astrocytes. Furthermore, GAS co- or pre-treatment markedly suppressed Zn2+-induced cell death caused by excessive ROS production and PARP-1 induction. We found that GAS suppressed p67 expression and PAR formation in astrocytes, which might underlie the anti- Zn2+-toxicity and anti-oxidative effects of GAS in astrocytes. These findings indicate GAS protects astrocytes from Zn2+-induced toxicity and oxidative stress and these effects contribute to its neuroprotective effects in the postischemic brain.

7.
Cell Death Dis ; 9(4): 426, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29555931

ABSTRACT

Post-stroke infection (PSI) is known to worsen functional outcomes of stroke patients and accounts to one-third of stroke-related deaths in hospital. In our previous reports, we demonstrated that massive release of high-mobility group box protein 1 (HMGB1), an endogenous danger signal molecule, is promoted by N-methyl-D-aspartic acid-induced acute damage in the postischemic brain, exacerbating neuronal damage by triggering delayed inflammatory processes. Moreover, augmentation of proinflammatory function of lipopolysaccharides (LPS) by HMGB1 via direct interaction has been reported. The aim of this study was to investigate the role of HMGB1 in aggravating inflammation in the PSI by exacerbating the function of LPS. PSI animal model was produced by administrating a low-dose LPS at 24 h post-middle cerebral artery occlusion (MCAO). Profound aggravations of inflammation, deterioration of behavioral outcomes, and infarct expansion were observed in LPS-injected MCAO animals, in which serum HMGB1 surge, especially disulfide type, occurred immediately after LPS administration and aggravated brain and systemic inflammations probably by acting in synergy with LPS. Importantly, blockage of HMGB1 function by delayed administrations of therapeutic peptides known to inhibit HMGB1 (HMGB1 A box, HPep1) or by treatment with LPS after preincubation with HMGB1 A box significantly ameliorated damages observed in the rat PSI model, demonstrating that HMGB1 plays a crucial role. Furthermore, administration of Rhodobacter sphaeroides LPS, a selective toll-like receptor 4 antagonist not only failed to exert these effects but blocked the effects of LPS, indicating its TLR4 dependence. Together, these results indicated that alarmin HMGB1 mediates potentiation of LPS function, exacerbating TLR4-dependent systemic and brain inflammation in a rat PSI model and there is a positive-feedback loop between augmentation of LPS function by HMGB1 and subsequent HMGB1 release/serum. Therefore, HMGB1 might be a valuable therapeutic target for preventing post-stroke infection.


Subject(s)
Bacterial Infections/etiology , Brain/metabolism , HMGB1 Protein/metabolism , Infarction, Middle Cerebral Artery/pathology , Animals , Bacterial Infections/metabolism , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Cyclooxygenase 2/metabolism , Disease Models, Animal , HMGB1 Protein/antagonists & inhibitors , HMGB1 Protein/blood , Infarction, Middle Cerebral Artery/metabolism , Inflammation/metabolism , Inflammation/pathology , Ketamine/pharmacology , Lipopolysaccharides/toxicity , Male , Nitric Oxide Synthase Type II/metabolism , Peptides/pharmacology , Rats , Rats, Sprague-Dawley , Toll-Like Receptor 4/antagonists & inhibitors , Toll-Like Receptor 4/metabolism
8.
Exp Mol Med ; 50(1): e430, 2018 01 19.
Article in English | MEDLINE | ID: mdl-29350679

ABSTRACT

Osteopontin (OPN) is a phosphorylated glycoprotein secreted into body fluids by various cell types. OPN contains arginine-glycine-aspartate (RGD) and serine-leucine-alanine-tyrosine (SLAY) motifs that bind to several integrins and mediate a wide range of cellular processes. In the present study, the proangiogenic effects of a 20-amino-acid OPN peptide (OPNpt20) containing RGD and SLAY motifs were examined in human umbilical vein endothelial cells (HUVECs) and in a rat focal cerebral ischemia model. OPNpt20 exerted robust proangiogenic effects in HUVECs by promoting proliferation, migration and tube formation. These effects were significantly reduced in OPNpt20-RAA (RGD->RAA)-treated cells, but only slightly reduced in OPNpt20-SLAA (SLAY->SLAA)-treated cells. Interestingly, a mutant peptide without both motifs failed to induce these proangiogenic processes, indicating that the RGD motif is crucial and that SLAY also has a role. In OPNpt20-treated HUVEC cultures, AKT and ERK signaling pathways were activated, but activation of these pathways and tube formation were suppressed by anti-αvß3 antibody, indicating that OPNpt20 stimulates angiogenesis via the αvß3-integrin/AKT and ERK pathways. The proangiogenic function of OPNpt20 was further confirmed in a rat middle cerebral artery occlusion model. Total vessel length and vessel densities were markedly greater in OPNpt20-treated ischemic brains, accompanied by induction of proangiogenic markers. Together, these results demonstrate that the 20-amino-acid OPN peptide containing RGD and SLAY motifs exerts proangiogenic effects, wherein both motifs have important roles, and these effects appear to contribute to the neuroprotective effects of this peptide in the postischemic brain.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Infarction, Middle Cerebral Artery/drug therapy , Osteopontin/chemistry , Peptide Fragments/pharmacology , Amino Acid Motifs , Angiogenesis Inducing Agents/chemistry , Animals , Brain/blood supply , Brain/drug effects , Brain/physiopathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Integrin alphaVbeta3/metabolism , MAP Kinase Signaling System/drug effects , Male , Neuroprotective Agents/pharmacology , Oligopeptides/chemistry , Peptide Fragments/chemistry , Rats, Sprague-Dawley
9.
Mol Neurobiol ; 55(7): 6094-6111, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29177697

ABSTRACT

It has been reported that the innate immune response plays important roles in brain ischemia and that the infiltration of blood-derived immune cells is a key initiator of this response. Nerve injury-induced protein 1 (Ninjurin 1, Ninj1) is a cell adhesion molecule responsible for cell-to-cell interactions between immune cells and endothelial cells. In the present study, we investigated the proinflammatory and neuroprotective effects of Ninj1 and a dodecamer peptide harboring Ninj1 N-terminal adhesion motif (N-NAM, Pro26~Asn37) in a rat middle cerebral artery occlusion (MCAO) model of stroke. Ninj1 was predominantly induced in neutrophils and endothelial cells in the ischemic hemispheres around 12 h to 1 day post-MCAO, which coincided with a massive neutrophil influx. We demonstrated that intranasal administration of Ninj1 small interfering RNA (siRNA) or N-NAM significantly blocked neutrophil infiltration in postischemic brains. In addition, intranasal administration of Ninj1 siRNA or N-NAM reduced the mean infarct volume to 46.5 ± 9.2 or 30.6 ± 11.7% of that of the PBS-treated MCAO controls, respectively, which was accompanied by significant amelioration of neurological and motor deficits. We showed that N-NAM or Ninj1 siRNA effectively blocked the adhesion and transendothelial migration of TNF-α-stimulated human myelocytic leukemia cells to human umbilical vein endothelial cells and similarly suppressed adhesion and migration of monocytes. Activations of phosphoinositide 3-kinase and Ras-related C3 botulinum toxin substrate 1 are involved in these Ninj1-mediated processes and can be inhibited by N-NAM or Ninj1 siRNA. These results indicate that Ninj1 plays an important role in neutrophil infiltration in the postischemic brain and N-NAM confers robust neuroprotective and anti-inflammatory effects by inhibiting Ninj1-mediated infiltration of neutrophils.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Brain Ischemia/drug therapy , Brain/pathology , Cell Adhesion Molecules, Neuronal/chemistry , Nerve Growth Factors/chemistry , Neuroprotective Agents/therapeutic use , Peptides/therapeutic use , Amino Acid Motifs , Amino Acid Sequence , Animals , Anti-Inflammatory Agents/pharmacology , Brain/physiopathology , Brain Ischemia/pathology , Cell Adhesion , Cell Adhesion Molecules, Neuronal/metabolism , Cell Communication/drug effects , Cell Movement/drug effects , Disease Models, Animal , Gene Knockdown Techniques , HL-60 Cells , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Inflammation/pathology , Male , Motor Activity/drug effects , Nerve Growth Factors/metabolism , Neuroprotective Agents/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Peptides/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Rats, Sprague-Dawley , Recovery of Function/drug effects , Tumor Necrosis Factor-alpha/pharmacology , rac1 GTP-Binding Protein/metabolism
10.
Cell Mol Neurobiol ; 38(3): 615-626, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28608001

ABSTRACT

4-Hydroxybenzyl alcohol (4-HBA) is an important phenolic constituent of Gastrodia elata (GE) Blume, which is used as a traditional herbal medicine in East Asia. Many activities have been reported to underlie the beneficial effects of 4-HBA in brain, such as, anti-oxidative, anti-inflammatory, anti-excitotoxic, and anti-apoptotic effects in neurons and microglia. Here, the authors demonstrate the robust neuroprotective effects of 4-HBA in rat middle cerebral artery occlusion (MCAO) model of stroke, and showed anti-Zn2+ toxicity in neurons and astrocytes as a molecular mechanism contributing to these effects. Intraperitoneal administration of 4-HBA (20 mg/kg) in Sprague-Dawley rats 1 h after MCAO reduced infarct volumes to 27.1 ± 9.2% of that of MCAO controls and significantly ameliorated motor impairments and neurological deficits. Significant suppressions of Zn2+-induced cell death, ROS generation, and PARP-1 induction by 4-HBA were observed in primary cortical cultures. 4-HBA also protected astrocytes from Zn2+-induced toxicity and suppressing ROS generation by employing slightly different molecular mechanisms, i.e., suppressing PARP-1 induction and NAD depletion under acute Zn2+-treatment and suppressing p67 NADPH oxidase subunit induction under chronic Zn2+-treatment. Results indicate that the protective effects of 4-HBA against Zn2+-toxicity in neurons and astrocytes contribute to its robust neuroprotective effects in the postischemic brain. Considering the pleiotropic effects of 4-HBA, which have been reported in previous reports and added in the present study, it has therapeutic potential for the amelioration of ischemic brain damage.


Subject(s)
Astrocytes/drug effects , Benzyl Alcohols/pharmacology , Brain Ischemia/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Astrocytes/metabolism , Brain/drug effects , Brain/metabolism , Brain Ischemia/metabolism , Cell Death/drug effects , Infarction, Middle Cerebral Artery/drug therapy , Male , Neurons/metabolism , Rats, Sprague-Dawley
11.
PLoS One ; 12(5): e0177322, 2017.
Article in English | MEDLINE | ID: mdl-28489907

ABSTRACT

4-Hydroxybenzyl alcohol (4-HBA) is an important phenolic constituent of Gastrodia elata Blume (GEB), a traditional herbal medicine used in East Asia. Many activities have been reported to underlie the beneficial effects of 4-HBA in the brain, and in particular, its anti-inflammatory, anti-oxidative, and anti-zinc-toxic effects have been implicated in the postischemic brain. Here, the authors investigated the anti-oxidative effect of 4-HBA on astrocytes and sought to identify the underlying molecular mechanisms involved. 4-HBA dose-dependently suppressed H2O2-induced astrocyte cell death. More specifically, pre-incubation of C6 cells (an astrocyte cell line) with 100 µM 4-HBA for 6 hrs increased survival when cells were treated with H2O2 (100 µM, 1 hr) from 54.2±0.7% to 85.9±1.5%. In addition, 4-HBA was found to up-regulate and activate Nrf2, and subsequently, to induce the expressions of several anti-oxidative genes, such as, HO-1, NQO1, and GCLM. Notably, HO-1 was induced by 3.4-fold in 4-HBA-treated C6 cells, and siRNA-mediated HO-1 knockdown demonstrated that Nrf2 activation and HO-1 induction were responsible for the observed cytoprotective effect of 4-HBA. ERK and Akt signaling pathways were activated by 4-HBA in C6 cells, suggesting their involvements in protective effect of 4-HBA. In addition, 4-HBA-conditioned astrocyte culture medium was found to have neuroprotective effects on primary neuronal cultures or fresh C6 cells exposed to oxidative stress, and these effects seemed to be mediated by glial cell line-derived neurotrophic factor (GDNF) and vascular endothelial growth factor (VEGF), which both accumulated in 4-HBA-treated astrocyte culture media. Thus, the 4-HBA-mediated activation of Nrf2 and induction of HO-1 in astrocytes were found to act via autocrine and paracrine mechanisms to confer protective effects. Furthermore, given the pleiotropic effects of 4-HBA with respect to its targeting of various brain cell types and functions, it would appear that 4-HBA has therapeutic potential for the prevention and amelioration of various brain diseases.


Subject(s)
Antioxidants/pharmacology , Astrocytes/drug effects , Benzyl Alcohols/pharmacology , Cell Death/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Antioxidants/chemistry , Astrocytes/cytology , Astrocytes/metabolism , Benzyl Alcohols/chemistry , Cells, Cultured , Female , Gastrodia/chemistry , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Heme Oxygenase-1/metabolism , Hydrogen Peroxide/metabolism , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred ICR , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism
12.
Sci Rep ; 7: 42891, 2017 02 21.
Article in English | MEDLINE | ID: mdl-28220827

ABSTRACT

Ethyl pyruvate (EP) is a simple aliphatic ester of pyruvic acid and has been shown to have robust neuroprotective effects via its anti-inflammatory, anti-oxidative, and anti-apoptotic functions. In an effort to develop novel EP derivatives with greater protective potencies than EP, we generated four EP isosteres, among them the neuroprotective potency of N,N-diethyl-2-oxopropanamide (DEOPA), in which the ethoxy group of EP was replaced with diethylamine, was far greater than that of EP. When DEOPA was administered intravenously (5 mg/kg) to rat middle cerebral artery occlusion (MCAO) model at 6 hrs post-surgery, it suppressed infarct formation, ameliorated neurological and sensory/motor deficits, and inhibited microglial activation and neutrophil infiltrations in the postischemic brain more effectively than EP. In particular, DEOPA markedly suppressed LPS-induced nitrite production and cytokine/chemokine inductions in microglia, neutrophils, and endothelial cells and these effects are attributable to inhibition of the activity of NF-κB by suppressing IκB-α degradation and p65 to DNA binding. In addition, DEOPA suppressed NMDA-induced neuronal cell death in primary cortical neuron cultures by NAD replenishment and suppression of NF-κB activity. Together, these results indicate DEOPA has multi-modal protective effects against ischemic brain damage targeting numerous cell types in the brain and also against other inflammation-related diseases.


Subject(s)
Amides/pharmacology , Anti-Inflammatory Agents/pharmacology , Brain/drug effects , Neuroprotective Agents/pharmacology , Amides/therapeutic use , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Brain/metabolism , Brain Ischemia/drug therapy , Brain Ischemia/etiology , Cell Adhesion/drug effects , Cell Movement/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Infarction, Middle Cerebral Artery/complications , Male , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , NF-KappaB Inhibitor alpha/antagonists & inhibitors , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/chemistry , Neuroprotective Agents/therapeutic use , Neutrophils/cytology , Neutrophils/metabolism , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Transcription Factor RelA/chemistry , Transcription Factor RelA/metabolism
13.
Exp Neurobiol ; 26(6): 339-349, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29302201

ABSTRACT

Osteopontin (OPN) is a secreted glycoprotein that is expressed in various tissues, including brain, and mediates a wide range of cellular activities. In a previous study, the authors observed the robust neuroprotective effects of recombinant OPN and of RGD and SLAYGLR-containing OPN-peptide icosamer (OPNpt20) in an animal model of transient focal ischemia, and demonstrated anti-inflammatory and pro-angiogenic effects of OPNpt20 in the postischemic brain. In the present study, we investigated the effects of OPNpt20 on the motility and phagocytic activity of BV2 cells (a microglia cell line). F-actin polymerization and cell motility were significantly enhanced in OPNpt20-treated BV2 cells, and numbers of filopodia-like processes increased and lamellipodia-like structures enlarged and thickened. In addition, treatment of cells with either of three mutant OPN icosamers containing mutation within RGD, SLAY, or RGDSLAY showed that the RGD and SLAY motifs of OPNpt20 play critical roles in the enhancement of cell motility, and the interaction between exogenous OPNpt20 and endogenous αv and α4 integrin and the activations of FAK, Erk, and Akt signaling pathways were found to be involved in the OPNpt20-mediated induction of cell motility. Furthermore, phagocytic activity of microglia was also significantly enhanced by OPNpt20 in a RGD and SLAY dependent manner. These results indicate OPNpt20 containing RGD and SLAY motifs triggers microglial motility and phagocytic activity and OPNpt20-integrin mediated signaling plays a critical role in these activities.

14.
Sci Rep ; 6: 31843, 2016 08 22.
Article in English | MEDLINE | ID: mdl-27545301

ABSTRACT

Postischemic brain damage in stroke is proceded with complicated pathological events, and so multimodal drug treatments may offer better therapeutic means for improving clinical outcomes. Here, we report robust neuroprotective effects of a novel compound, 2-((2-oxopropanoyl)oxy)-4-(trifluoromethyl)benzoic acid (OPTBA), a 2-hydroxy-4-trifluoromethyl benzoic acid (HTB, a metabolite of triflusal)-pyruvate ester. Intravenous administration of OPTBA (5 mg/kg) 3 or 6 h after middle cerebral artery occlusion (MCAO) in Sprague-Dawley rats reduced infarct volumes to 38.5 ± 11.4% and 46.5 ± 15.3%, respectively, of that of MCAO controls, and ameliorated motor impairment and neurological deficits. Importantly, neuroprotective effects of OPTBA were far greater than those afforded by combined treatment of HTB and pyruvate. Furthermore, OPTBA suppressed microglial activation and proinflammatory cytokine inductions more effectively than HTB/pyruvate co-treatment in the postischemic brain and LPS-treated cortical slice cultures and also attenuated NMDA-induced neuronal death in hippocampal slice cultures. LC-MS analysis demonstrated that OPTBA was hydrolyzed to HTB and pyruvate with a t1/2 of 38.6 min in blood and 7.2 and 2.4 h in cortex and striatum, respectively, and HTB was maintained for more than 24 h both in blood and brain tissue. Together these results indicate OPTBA acts directly and via its hydrolysis products, thus acting as a multimodal neuroprotectant in the postischemic brain.


Subject(s)
Benzoates/administration & dosage , Brain Ischemia/drug therapy , Neuroprotective Agents/administration & dosage , Stroke/prevention & control , Animals , Benzoates/chemical synthesis , Benzoates/chemistry , Blood-Brain Barrier/chemistry , Brain Ischemia/immunology , Cytokines/metabolism , Disease Models, Animal , Hydrolysis , Male , Neuroprotective Agents/chemical synthesis , Neuroprotective Agents/chemistry , Rats , Rats, Sprague-Dawley , Salicylates/chemistry , Stroke/immunology
15.
Exp Neurobiol ; 25(2): 64-74, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27122992

ABSTRACT

Nerve injury-induced protein-1 (Ninjurin-1, Ninj1) was initially identified as a novel adhesion molecule in rat sciatic nerve and to be up-regulated in neurons and Schwann cells of distal nerve segments after nerve transection or crush injury. Recently, Ninj1 was found to act as a modulator of cell migration, angiogenesis, and apoptosis. Accumulating evidence indicates that innate immune response plays beneficial and deleterious roles in brain ischemia, and the trans-endothelial migration of blood-derived immune cells is key initiator of this response. In the present study, we examined the expression profile and cellular distribution of Ninj1 in rat brain after transient focal cerebral ischemia. Ninj1 expression was found to be significantly induced in cortical penumbras 1 day after 60 min of middle cerebral artery occlusion (MCAO) and to increase gradually for 8 days and then declined. In infarction cores of cortices, patterns of Ninj1 expression were similar to those observed in cortical penumbras, except induction was maintained for 10 days. At 1 day post-MCAO, Ninj1 inductions were detected mainly in neutrophils and endothelial cells in both infarction cores and penumbras, but reactive macrophages were the major cellular expressers of Ninj1 at 4 days post-MCAO. Expressional induction in reactive macrophages was maintained in infarction cores after 12 days post-MCAO but not in penumbras. These dynamic expressions of Ninj1 in different immune cells at different times suggest that this protein performs various, critical roles in the modulation of acute and delayed immune responses in the postischemic brain.

16.
Mol Neurobiol ; 53(8): 5652-63, 2016 10.
Article in English | MEDLINE | ID: mdl-26482372

ABSTRACT

Osteopontin (OPN) is a phosphorylated glycoprotein possessing an arginine-glycine-aspartate (RGD)-motif, which binds to several cell surface integrins and mediates a wide range of cellular processes. Inductions of OPN have been reported in the postischemic brain, and the neuroprotective effects of OPN have been demonstrated in animal models of stroke. In the present study, we showed a robust neuroprotective effect of RGD-containing icosamer OPN peptide (OPNpt20) in a rat model of focal cerebral ischemia (middle cerebral artery occlusion, MCAO). Intranasally administered OPNpt20 reduced mean infarct volume by 79.7 % compared to the treatment-naïve MCAO control animals and markedly ameliorated neurological deficits. In addition, OPNpt20 significantly suppressed the inductions of iNOS and of inflammatory markers in postischemic brains and in primary microglial cultures, demonstrating anti-inflammatory effects. Administration of a mutant peptide, in which RGD was replaced by arginine-alanine-alanine (RAA), failed to suppress infarct volumes in MCAO animals and co-administration of OPNpt20 with anti-αvß3 integrin antibody failed to suppress iNOS induction in primary microglia culture, indicating that the RGD motif in OPNpt20 and endogenous αvß3 integrin play critical roles. Furthermore, pull-down assay revealed a direct binding between OPNpt20 and αvß3 integrin in primary microglia culture. Together, these results indicate that RGD-containing OPN icosamer has therapeutic potential in the postischemic brain and αvß3 integrin-mediated anti-inflammatory effect might be an underlying mechanism.


Subject(s)
Brain Ischemia/drug therapy , Drug Delivery Systems , Integrin alphaVbeta3/metabolism , Neuroprotection , Oligopeptides/administration & dosage , Osteopontin/administration & dosage , Osteopontin/therapeutic use , Administration, Intranasal , Amino Acid Sequence , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cells, Cultured , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Microglia/drug effects , Microglia/metabolism , Motor Activity , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/metabolism , Nitrites/metabolism , Osteopontin/chemistry , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Isoforms/administration & dosage , Protein Isoforms/chemistry , Protein Isoforms/therapeutic use , Rats, Sprague-Dawley , STAT1 Transcription Factor/metabolism
17.
Neurosci Lett ; 593: 40-4, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25769801

ABSTRACT

High mobility group box 1 (HMGB1) is an endogenous danger signal molecule. In the postischemic brain, HMGB1 is massively released during acute damaging process and triggers inflammatory processes. Moreover, it has been reported HMGB1 augments the proinflammatory effect of LPS by direct interaction. In previous studies, the authors showed intranasally delivered a HMGB1 binding heptamer peptide (HBHP; HMSKPVQ) has robust neuroprotective effects in the ischemic brain after middle cerebral artery occlusion and that it exerts an anti-inflammatory effect. In the present study, the authors investigated whether HBHP suppresses the augmentation of the proinflammatory effect of LPS by HMGB1. In primary microglial cultures, low doses of LPS (5 ng/ml) and recombinant HMGB1 (rHMGB1, 20 ng/ml) synergistically activated microglial cells, and HMGB1-LPS binding was detected. In addition, synergistic NO accumulation along with direct HMGB1-LPS binding was also observed when primary microglial cultures were treated with LPS (5 ng/ml) and HMGB1 accumulated in NMDA-conditioned medium (NCM). Co-treatment of microglial cells with HBHP and LPS or rHMGB1 (NCM), or treatment with rHMGB1 or NCM and LPS after pre-incubating rHMGB1 (or NCM) with HBHP markedly suppressed their synergistic activation. Furthermore, interactions between rHMGB1 and LPS or between HMGB1 in NCM and LPS were suppressed dose-dependently by HBHP, indicating that HBHP suppressed the synergism between HMGB1 and LPS and the underlying mechanism involved inhibition of HMGB1-LPS binding. Together these results show HBHP has anti-inflammatory effects, and that it inhibits synergism caused by the binding of HMGB1 and LPS.


Subject(s)
Anti-Inflammatory Agents/pharmacology , HMGB1 Protein/metabolism , Lipopolysaccharides/pharmacology , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Animals , Brain/cytology , Cells, Cultured , Culture Media, Conditioned , HMGB1 Protein/pharmacology , Mice , Microglia/drug effects , Microglia/metabolism , N-Methylaspartate/pharmacology , Nitric Oxide/metabolism , Oligopeptides/metabolism , Protein Binding , Rats, Sprague-Dawley , Recombinant Proteins/pharmacology
18.
Neurosci Lett ; 558: 159-63, 2014 Jan 13.
Article in English | MEDLINE | ID: mdl-24246904

ABSTRACT

Ethyl pyruvate (EP) has been shown to have anti-inflammatory effects and confer protective effects in various pathological conditions. For example, EP inhibits secretion of high mobility group box 1 (HMGB1), which is known to be released from activated or dying cells and aggravate inflammatory pathways. In the present study, we investigated whether EP reduces HMGB1 phosphorylation and release in ischemic brain and in cultured microglia. In the postischemic brains (60 min middle cerebral artery occlusion (MCAO)), HMGB1 was released extracellularly, generating dual peaks in cerebrospinal fluid (CSF) around 1 and 7 days after ischemic insult, which were probably generated from damaged neurons and activated inflammatory cells, respectively. We showed that treatment with EP 30 min post-MCAO (5 mg/kg, i.v.), which has been shown to confer a robust neuroprotective effect in the postischemic brain, reduced both peaks. In addition, delayed EP treatment from 4 days post-MCAO reduced HMGB1 accumulation in CSF at 7 day post-MCAO in the absence of accompanying amelioration of ischemic brain damage, indicating that the suppression of HMGB1 release is a direct effect. We also found that EP markedly suppressed the LPS-induced nuclear translocations of protein kinase C alpha and calcium/calmodulin-dependent protein kinase IV, HMGB1 phosphorylation, and subsequent secretion of HMGB1 induced by LPS in BV2 cells and EP-mediated above-mentioned effects were also independent of cell death or survival. These results indicate that EP inhibits HMGB1 phosphorylation and release in activated microglia, which might be responsible for EP-mediated suppression of HMGB1 release in the postischemic brain.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Brain Ischemia/metabolism , Brain/drug effects , HMGB1 Protein/metabolism , Microglia/drug effects , Neuroprotective Agents/pharmacology , Pyruvates/pharmacology , Animals , Brain/metabolism , Brain Ischemia/physiopathology , HMGB1 Protein/cerebrospinal fluid , Male , Microglia/metabolism , Phosphorylation , Protein Transport , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...