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










Publication year range
1.
BMB Rep ; 55(9): 447-452, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35651331

ABSTRACT

Neurogenic differentiation 1 (NeuroD1) is an essential transcription factor for neuronal differentiation, maturation, and survival, and is associated with inflammation in lipopolysaccharide (LPS)- induced glial cells; however, the concrete mechanisms are still ambiguous. Therefore, we investigated whether NeuroD1-targeting miRNAs affect inflammation and neuronal apoptosis, as well as the underlying mechanism. First, we confirmed that miR-30a-5p and miR-153-3p, which target NeuroD1, reduced NeuroD1 expression in microglia and astrocytes. In LPS-induced microglia, miR-30a-5p and miR-153-3p suppressed pro-inflammatory cytokines, reactive oxygen species, the phosphorylation of c-Jun N-terminal kinase, extracellular-signal-regulated kinase (ERK), and p38, and the expression of cyclooxygenase and inducible nitric oxide synthase (iNOS) via the NF-κB pathway. Moreover, miR-30a-5p and miR-153-3p inhibited the expression of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasomes, NLRP3, cleaved caspase-1, and IL-1ß, which are involved in the innate immune response. In LPS-induced astrocytes, miR-30a-5p and miR-153-3p reduced ERK phosphorylation and iNOS expression via the STAT-3 pathway. Notably, miR-30a-5p exerted greater anti-inflammatory effects than miR-153-3p. Together, these results indicate that miR-30a-5p and miR-153-3p inhibit MAPK/NF-κB pathway in microglia as well as ERK/STAT-3 pathway in astrocytes to reduce LPS-induced neuronal apoptosis. This study highlights the importance of NeuroD1 in microglia and astrocytes neuroinflammation and suggests that it can be regulated by miR-30a-5p and miR-153-3p. [BMB Reports 2022; 55(9): 447-452].


Subject(s)
Lipopolysaccharides , MicroRNAs , Anti-Inflammatory Agents , Apoptosis , Basic Helix-Loop-Helix Transcription Factors , Caspases/metabolism , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Inflammasomes/metabolism , Inflammation/genetics , Inflammation/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Reactive Oxygen Species/metabolism
2.
BMB Rep ; 54(11): 557-562, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34353430

ABSTRACT

Microglial activation is closely associated with neuroinflammatory pathologies. The nucleotide-binding and oligomerization domain-like receptor containing a pyrin domain 3 (NLRP3) inflammasomes are highly organized intracellular sensors of neuronal alarm signaling. NLRP3 inflammasomes activate nuclear factor kappa-B (NF-κB) and reactive oxygen species (ROS), which induce inflammatory responses. Moreover, NLRP3 dysfunction is a common feature of chronic inflammatory diseases. The present study investigated the effect of a novel thiazol derivative, N-cyclooctyl-5-methylthiazol-2-amine hydrobromide (KHG26700), on inflammatory responses in lipopolysaccharide (LPS)-treated BV-2 microglial cells. KHG26700 significantly attenuated the expression of several pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin-1ß, and interleukin-6, in these cells, as well as the LPS-induced increases in NLRP3, NF-κB, and phospho-IkBα levels. KHG26700 also suppressed the LPS-induced increases in protein levels of autophagy protein 5 (ATG5), microtubule- associated protein 1 light chain 3 (LC3), and beclin-1, as well as downregulating the LPS-enhanced levels of ROS, lipid peroxidation, and nitric oxide. These results suggest that the anti-inflammatory effects of KHG26700 may be due, at least in part, to the regulation of the NLRP3-mediated signaling pathway during microglial activation. [BMB Reports 2021; 54(11): 557-562].


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bromides/chemistry , Inflammasomes/drug effects , Inflammation/drug therapy , Lipopolysaccharides/toxicity , Microglia/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/chemistry , Cytokines/metabolism , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lipid Peroxidation , Macrophage Activation , Mice , Microglia/metabolism , Microglia/pathology , NF-kappa B/antagonists & inhibitors , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
3.
Int J Mol Sci ; 22(3)2021 Jan 26.
Article in English | MEDLINE | ID: mdl-33530496

ABSTRACT

S100 calcium-binding protein A8 (S100A8), a danger-associated molecular pattern, has emerged as an important mediator of the pro-inflammatory response. Some S100 proteins play a prominent role in neuroinflammatory disorders and increase the secretion of pro-inflammatory cytokines in microglial cells. The aim of this study was to determine whether S100A8 induced neuronal apoptosis during cerebral hypoxia and elucidate its mechanism of action. In this study, we reported that the S100A8 protein expression was increased in mouse neuronal and microglial cells when exposed to hypoxia, and induced neuroinflammation and neuronal apoptosis. S100A8, secreted from neurons under hypoxia, activated the secretion of tumor necrosis factor (TNF-α) and interleukin-6 (IL-6) through phosphorylation of extracellular-signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) in microglia. Also, phosphorylation of ERK via the TLR4 receptor induced the priming of the NLRP3 inflammasome. The changes in Cyclooxygenase-2 (COX-2) expression, a well-known inflammatory activator, were regulated by the S100A8 expression in microglial cells. Knockdown of S100A8 levels by using shRNA revealed that microglial S100A8 expression activated COX-2 expression, leading to neuronal apoptosis under hypoxia. These results suggested that S100A8 may be an important molecule for bidirectional microglia-neuron communication and a new therapeutic target for neurological disorders caused by microglial inflammation during hypoxia.


Subject(s)
Apoptosis/genetics , Calgranulin A/genetics , Gene Expression Regulation , Hypoxia/genetics , Hypoxia/metabolism , Microglia/metabolism , Neurons/metabolism , Animals , Biomarkers , Calgranulin A/metabolism , Cell Line , Cytokines/metabolism , Disease Susceptibility , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Phosphorylation
4.
Front Immunol ; 11: 1258, 2020.
Article in English | MEDLINE | ID: mdl-32903598

ABSTRACT

S100A8 and S100A9 function as essential factors in inflammation and also exert antitumor or tumorigenic activity depending on the type of cancer. Chronic eosinophilic leukemia (CEL) is a rare hematological malignancy having elevated levels of eosinophils and characterized by the presence of the FIP1L1-PDGFRA fusion gene. In this study, we examined the pro-apoptotic mechanisms of S100A8 and S100A9 in FIP1L1-PDGFRα+ eosinophilic cells and hypereosinophilic patient cells. S100A8 and S100A9 induce apoptosis of the FIP1L1-PDGFRα+ EoL-1 cells via TLR4. The surface TLR4 expression increased after exposure to S100A8 and S100A9 although total TLR4 expression decreased. S100A8 and S100A9 suppressed the FIP1L1-PDGFRα-mediated signaling pathway by downregulating FIP1L1-PDGFRα mRNA and protein expression and triggered cell apoptosis by regulating caspase 9/3 pathway and Bcl family proteins. S100A8 and S100A9 also induced apoptosis of imatinib-resistant EoL-1 cells (EoL-1-IR). S100A8 and S100A9 blocked tumor progression of xenografted EoL-1 and EoL-1-IR cells in NOD-SCID mice and evoked apoptosis of eosinophils derived from hypereosinophilic syndrome as well as chronic eosinophilic leukemia. These findings may contribute to a progressive understanding of S100A8 and S100A9 in the pathogenic and therapeutic mechanism of hematological malignancy.


Subject(s)
Apoptosis , Calgranulin A/metabolism , Calgranulin B/metabolism , Hypereosinophilic Syndrome/etiology , Hypereosinophilic Syndrome/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cells, Cultured , Chronic Disease , Drug Resistance, Neoplasm , Female , Gene Expression , Humans , Hypereosinophilic Syndrome/drug therapy , Hypereosinophilic Syndrome/pathology , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Recombinant Proteins
5.
BMB Rep ; 53(10): 527-532, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32635984

ABSTRACT

We recently reported that N-adamantyl-4-methylthiazol-2-amine (KHG26693) attenuates glutamate-induced oxidative stress and inflammation in the brain. In this study, we investigated KHG 26693 as a therapeutic agent against glutamate-induced autophagic death of cortical neurons. Treatment with KHG26693 alone did not affect the viability of cultured cortical neurons but was protective against glutamate-induced cytotoxicity in a concentration-dependent manner. KHG26693 attenuated the glutamate-induced increase in protein levels of LC3, beclin-1, and p62. Whereas glutamate decreased the phosphorylation of PI3K, Akt, and mTOR, these levels were restored by treatment with KHG26693. These results suggest that KHG26693 inhibits glutamate-induced autophagy by regulating PI3K/Akt/mTOR signaling. Finally, KHG26693 treatment also attenuated glutamateinduced increases in reactive oxygen species, glutathione, glutathione peroxidase, and superoxide dismutase levels in cortical neurons, indicating that KHG26693 also protects cortical neurons against glutamate-induced autophagy by regulating the reactive oxygen species scavenging system. [BMB Reports 2020; 53(10): 527-532].


Subject(s)
Adamantane/analogs & derivatives , Autophagy/drug effects , Neurons/metabolism , Thiazoles/pharmacology , Adamantane/metabolism , Adamantane/pharmacology , Animals , Antioxidants/pharmacology , Autophagic Cell Death , Autophagy/physiology , Cerebral Cortex/metabolism , Glutamic Acid/adverse effects , Glutamic Acid/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Neurons/drug effects , Oxidative Stress/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , TOR Serine-Threonine Kinases/metabolism , Thiazoles/metabolism
6.
Eur J Pharmacol ; 856: 172412, 2019 Aug 05.
Article in English | MEDLINE | ID: mdl-31129157

ABSTRACT

New compounds were screened to develop effective drugs against glutamate-induced toxicity. The present study assessed the effects of the novel thiazole derivative KHG21834 against glutamate-induced toxicity in human neuroblastoma SH-SY5Y cell cultures. Treatment of SH-SY5Y cells with KHG21834 significantly protected cells against glutamate-induced toxicity in a dose-dependent manner, with an optimum concentration of 50 µM. KHG21834 protected SH-SY5Y cells against glutamate toxicity by suppressing glutamate-induced oxidative stress by 50%. KHG21834 also attenuated glutamate-induced mitochondrial membrane potential, ATP level reductions, and intracellular Ca2+ influx. Furthermore, KHG21834 efficiently reduced glutamate-induced ER stress and NLRP3 inflammasome activation (59% and 65% of glutamate group, respectively). In addition, KHG21834 effectively attenuated glutamate-induced levels of Bax, Bcl-2, cleaved caspase-3, p-p38, p-JNK proteins, and TUNEL positive cells. To our knowledge, this is the first study showing that KHG21834 can effectively protect SH-SY5Y cells against glutamate toxicity, suggesting that this compound may be a valuable therapeutic agent for the treatment of glutamate toxicity.


Subject(s)
Apoptosis/drug effects , Benzothiazoles/pharmacology , Glutamic Acid/adverse effects , Inflammasomes/metabolism , Mitochondria/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroblastoma/pathology , Adenosine Triphosphate/metabolism , Calcium/metabolism , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/pathology , Oxidative Stress/drug effects
7.
BMB Rep ; 52(10): 613-618, 2019 Oct.
Article in English | MEDLINE | ID: mdl-30940325

ABSTRACT

Microglial cells are known as the main immune cells in the central nervous system, both regulating its immune response and maintaining its homeostasis. Furthermore, the antioxidant α-lipoic acid (LA) is a recognized therapeutic drug for diabetes because it can easily invade the blood-brain barrier. This study investigated the effect of α-LA on the inflammatory response in lipopolysaccharide (LPS)-treated BV-2 microglial cells. Our results revealed that α-LA significantly attenuated several inflammatory responses in BV-2 microglial cells, including pro-inflammatory cytokines, such as tumor necrosis factor-α and interleukin (IL)-6, and other cytotoxic molecules, such as nitric oxide and reactive oxygen species. In addition, α-LA inhibited the LPS-induced phosphorylation of ERK and p38 and its pharmacological properties were facilitated via the inhibition of the nuclear factor kappa B signaling pathway. Moreover, α-LA suppressed the activation of NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasomes, multiprotein complexes consisting of NLRP3 and caspase-1, which are involved in the innate immune response. Finally, α-LA decreased the genes accountable for the M1 phenotype, IL-1ß and ICAM1, whereas it increased the genes responsible for the M2 phenotype, MRC1 and ARG1. These findings suggest that α-LA alleviates the neuroinflammatory response by regulating microglial polarization. [BMB Reports 2019; 52(10): 613-618].


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammasomes/metabolism , Microglia/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Thioctic Acid/pharmacology , Animals , Caspase 1/metabolism , Cell Line , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Inflammation/metabolism , Intercellular Adhesion Molecule-1/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation , Membrane Glycoproteins/metabolism , Mice , Microglia/metabolism , NF-kappa B/metabolism , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , Receptors, Immunologic/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
8.
BMB Rep ; 51(11): 590-595, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29966582

ABSTRACT

Parkinson's disease (PD) is a common chronic neurodegenerative disease mainly caused by the death of dopaminergic neurons. However, no complete pharmacotherapeutic approaches are currently available for PD therapies. 1-methyl-4- phenylpyridinium (MPP+)-induced SH-SY5Y neurotoxicity has been broadly utilized to create cellular models and study the mechanisms and critical aspects of PD. In the present study, we examined the role of a novel azetidine derivative, 3-(naphthalen-2-yl(propoxy)methyl)azetidine hydrochloride (KHG26792), against MPP+-induced neurotoxicity in SH-SY5Y cells. Treatment of KHG26792 significantly attenuated MPP+-induced changes in the protein levels of Bcl-2 and Bax together with efficient suppression of MPP+-induced activation of caspase-3 activity. KHG26792 also attenuated mitochondrial potential and levels of ROS, Ca2+, and ATP in MPP+-treated SH-SY5Y cells. Additionally, KHG26792 inhibited the induced production of nitric oxide and malondialdehyde. Moreover, the protective effect of KHG26792 is mediated through regulation of glutathione peroxidase and GDNF levels. Our results suggest a possibility that KHG26792 treatment significantly protects against MPP+-induced neurotoxicity in SH-SY5Y cells and KHG26792 may be a valuable therapeutic agent for the treatment of PD induced by an environmental toxin. [BMB Reports 2018; 51(11): 590-595].


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Azetidines/pharmacology , Cytoprotection/drug effects , Mitochondria/drug effects , Naphthalenes/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Humans , Microglia/drug effects , Microglia/physiology , Mitochondria/metabolism , Neurons/physiology , Neurotoxicity Syndromes/pathology , Neurotoxicity Syndromes/prevention & control , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
9.
ACS Comb Sci ; 20(5): 282-291, 2018 05 14.
Article in English | MEDLINE | ID: mdl-29557646

ABSTRACT

A 2-amido benzo[ d]imidazole library has been constructed by solid-phase synthesis. The key step of this solid-phase synthesis involves the preparation of polymer-bound 2-amino benzo[ d]imidazole resin through desulfurative cyclization of thiourea resin using 2-chloro-1,3-dimethylimidazolinium chloride and N, N-diisopropylethylamine in dichloromethane (DCM), and the resin is then functionalized by acylation at the 2-amine position to afford 2-amidobenzo[ d]imidazole resin. In the case of 2-amidobenzo[ d]imidazole resin having a p-I or m-NO2, the resin was further functionalized by Suzuki/Sonogashira-coupling ( p-I) and reduction to the primary amine ( m-NO2) followed by acylation. Finally, the functionalized 2-amido-benzo[ d]imidazole resin was cleaved from the polymer support by treatment with a cocktail of trifluoroacetic acid and DCM. As a result, we obtained 2-amidobenzo[ d]imidazole analogues in high yield and good purities.


Subject(s)
Benzimidazoles/chemical synthesis , Resins, Synthetic/chemistry , Solid-Phase Synthesis Techniques/methods , Thiourea/chemistry , Acylation , Amines/chemistry , Cyclization , Small Molecule Libraries/chemistry
10.
ACS Comb Sci ; 20(2): 82-97, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29309123

ABSTRACT

An efficient solid-phase synthetic route for the construction of 1,3,4-oxadiazole and 1,3,4-thiadiazole libraries based on branching diversity-oriented synthesis (DOS) has been developed in this study. The core skeleton resins, 1,3,4-oxadiazole and 1,3,4-thiadiazole, were obtained through desulfurative and dehydrative cyclizations of thiosemicarbazide resin, respectively. Various functional groups have been introduced to the core skeleton resins, such as aryl, amine, amide, urea, thiourea, and an amino acid. Most of the libraries were purified by simple trituration without extraction or column chromatography after cleavage of the products from the solid-supported resin. As a result, we obtained high yields of pure 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives (total numbers = 128). Finally, we confirmed the drug-like properties of our library by calculation of physicochemical properties, displays of the skeletal diversities of the library in 3D-space, and occupation of a broad range of areas by their functional groups.


Subject(s)
Oxadiazoles/chemical synthesis , Small Molecule Libraries/chemical synthesis , Solid-Phase Synthesis Techniques/methods , Thiadiazoles/chemical synthesis , Amides/chemistry , Amines/chemistry , Amino Acids/chemistry , Combinatorial Chemistry Techniques/methods , Cyclization , Molecular Structure , Semicarbazides/chemistry , Structure-Activity Relationship , Thiourea/chemistry
11.
Biochem Biophys Res Commun ; 495(1): 151-156, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29111328

ABSTRACT

The nucleotide-binding and oligomerization domain-like receptor containing a pyrin domain 3 (NLRP3) inflammasome is a multiprotein complex with a role in innate immune responses. NLRP3 inflammasome dysfunction is a common feature of chronic inflammatory diseases. Microglia activation is also associated with neuroinflammatory pathologies. We previously reported that 3-(naphthalen-2-yl(propoxy)methyl)azetidine hydrochloride (KHG26792) reduced hypoxia-induced toxicity by modulating inflammation. However, no studies have elucidated the precise mechanisms for the anti-inflammatory action of KHG26792, in particular via inflammasome mediation. This study investigated the effects of KHG26792 on the inflammasome-mediated signaling pathway in lipopolysaccharide (LPS)-stimulated BV2 microglial cells. KHG26792 significantly attenuated several inflammatory responses including tumor necrosis factor-α, interleukin-1ß, interleukin-6, reactive oxygen species, and mitochondrial potential in these cells. KHG26792 also suppressed LPS-induced increase NLRP3, activated caspase-1, and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) levels. Furthermore, KHG26792 successfully blocked LPS-activated adenosine triphosphate (ATP) level, likely through the purinergic receptor P2X ligand-gated ion channel 7 (P2X7) receptor. Our results suggest that the anti-inflammatory functions of KHG26792 may be, at least in part, due to regulation of the P2X7R/NLRP3-mediated signaling pathway during microglial activation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Azetidines/pharmacology , Inflammasomes/immunology , Microglia/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Naphthalenes/pharmacology , Animals , Cell Line , Interleukin-1beta/immunology , Interleukin-6/immunology , Lipopolysaccharides/immunology , Mice , Microglia/immunology , Reactive Oxygen Species/immunology , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/immunology
12.
ACS Comb Sci ; 19(12): 738-747, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29095593

ABSTRACT

2-Alkoxy/thioalkoxy benzo-[d]-imidazole and 2-thione benzo-[d]-imidazole libraries were constructed in solution phase and on solid phase, respectively. The key step in this work is the phase-based chemoselective reaction of the 2-mercaptobenzo-[d]-imidazole intermediate with benzyl chloride (solution phase) and Merrifield resin (solid phase). In the solution-phase case, benzyl chloride reacted with the thiol group of 2-mercaptobenzo-[d]-imidazole, whereas in the solid-phase case, Merrifield resin was introduced at an internal amine group of benzo-[d]-imidazole. To afford the desired 2-alkoxy/thioalkoxy benzo-[d]-imidazole analogues, we used various alkyl halides, alcohols, and thiols in solution phase, and to obtain 2-thione benzo-[d]-imidazole derivatives on solid phase, we used diverse alkyl halides and boronic acids. Finally, to measure the drug potential to be orally active and the molecular diversity in three-dimensional (3D) space, we calculated physicochemical properties and displayed energy-minimized 3D structures. As a result, the libraries from solution phase and solid phase show distinct features in physicochemical properties and skeletal diversities in 3D space.


Subject(s)
Imidazoles/chemistry , Small Molecule Libraries/chemistry , Sulfhydryl Compounds/chemistry , Thiones/chemistry , Cyclization , Imidazoles/chemical synthesis , Molecular Structure , Small Molecule Libraries/chemical synthesis , Solid-Phase Synthesis Techniques , Sulfhydryl Compounds/chemical synthesis , Thermodynamics , Thiones/chemical synthesis
13.
BMB Rep ; 50(12): 634-639, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29065971

ABSTRACT

We aimed to assess the anti-inflammatory and antioxidative properties of KHG26792, a novel azetidine derivative, in amyloid ß (Aß)-treated primary microglial cells. KHG26792 attenuated the Aß-induced production of inflammatory mediators such as IL-6, IL-1ß, TNF-α, and nitric oxide. The levels of protein oxidation, lipid peroxidation, ROS, and NADHP oxidase enhanced by Aß were also downregulated by KHG26792 treatment. The effects of KHG26792 against the Aß-induced increases in inflammatory cytokine levels and oxidative stress were achieved by increasing the phosphorylation of Akt/ GSK-3ß signaling and by decreasing the Aß-induced translocation of NF-κB. Our results provide novel insights into the use of KHG26792 as a potential agent against Aß toxicity, including its role in the reduction of inflammation and oxidative stress. Nevertheless, further investigations of cellular signaling are required to clarify the in vivo effects of KHG26792 against Aß-induced toxicity. [BMB Reports 2017; 50(12): 634-639].


Subject(s)
Amyloid beta-Peptides/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Azetidines/pharmacology , Microglia/drug effects , Naphthalenes/pharmacology , Amyloid beta-Peptides/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Antioxidants/chemistry , Azetidines/chemistry , Cells, Cultured , Microglia/cytology , Microglia/metabolism , Naphthalenes/chemistry , Rats , Rats, Sprague-Dawley
14.
Neurotox Res ; 32(1): 107-120, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28285348

ABSTRACT

In this study, we explored the possible mechanisms underlying the neuroprotective and anti-oxidative effects of N-adamantyl-4-methylthiazol-2-amine (KHG26693) against in vivo glutamate-induced toxicity in the rat cerebral cortex. Our results showed that pretreatment with KHG26693 significantly attenuated glutamate-induced elevation of lipid peroxidation, tumor necrosis factor-α, interferon gamma, IFN-γ, interleukin-1ß, nitric oxide, reactive oxygen species, NADPH oxidase, caspase-3, calpain activity, and Bax. Furthermore, KHG26693 pretreatment attenuated key antioxidant parameters such as levels of superoxide dismutase, catalase, glutathione, and glutathione reductase. KHG26693 also attenuated the protein levels of inducible nitric oxide synthase, neuronal nitric oxide synthase, nuclear factor erythroid 2-related factor 2, heme oxygenase-1, and glutamate cysteine ligase catalytic subunit caused by glutamate toxicity. Finally, KHG26693 mitigated glutamate-induced changes in mitochondrial ATP level and cytochrome oxidase c. Thus, KHG26693 functions as neuroprotective and anti-oxidative agent against glutamate-induced toxicity through its antioxidant and anti-inflammatory activities in rat brain at least in part.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Azetidines/therapeutic use , Encephalitis , Excitatory Amino Acid Agonists/toxicity , Glutamic Acid/toxicity , Naphthalenes/therapeutic use , Oxidative Stress/drug effects , Adenosine Triphosphate/metabolism , Animals , Cytokines/metabolism , Disease Models, Animal , Electron Transport Complex IV/metabolism , Encephalitis/chemically induced , Encephalitis/drug therapy , Encephalitis/physiopathology , Lipid Peroxidation/drug effects , Male , Mitochondria/drug effects , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sodium-Potassium-Exchanging ATPase/metabolism , Statistics, Nonparametric
15.
ACS Comb Sci ; 18(12): 702-709, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27783481

ABSTRACT

A novel solid-phase synthesis methodology of N-substituted-2-aminothiazolo[4,5-b]pyrazine derivatives was developed. The key step in this synthesis strategy is the tandem reaction of isothiocyanate terminated resin 2 with o-bromo-2-aminopyrazine, affording cyclized 2-aminothiazolo[4,5-b]pyrazine resin 4. To increase the diversity of our library, Suzuki coupling reaction was performed at the position C6. Further functionalization of 2-aminothiazolo[4,5-b]pyrazine core skeleton with various electrophiles such as alkyl halides, acyl chlorides, and sulfonyl chlorides and cleavage from the resin with TFA in DCM generated N-alkyl-, N-acyl-, and N-sulfonyl-2-aminothiazolo[4,5-b]pyrazine derivatives. The physicochemical properties and the polar surface areas of synthesized compounds were evaluated.


Subject(s)
Isothiocyanates/chemistry , Pyrazines/chemistry , Pyrazines/chemical synthesis , Solid-Phase Synthesis Techniques/methods , Thiazoles/chemical synthesis , Cyclization , Molecular Docking Simulation , Molecular Structure , Pyrazines/metabolism , Thiazoles/chemistry
16.
BMB Rep ; 49(12): 687-692, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27756444

ABSTRACT

We recently reported the anti-inflammatory effects of 3-(naphthalen-2-yl(propoxy)methyl)azetidine hydrochloride (KHG26792) on the ATP-induced activation of the NFAT and MAPK pathways through the P2X7 receptor in microglia. To further investigate the underlying mechanism of KHG26792, we studied its protective effects on hypoxia-induced toxicity in microglia. The administration of KHG26792 significantly reduced the hypoxia-induced expression and activity of caspase-3 in BV-2 microglial cells. KHG26792 also reduced hypoxia-induced inducible nitric oxide synthase protein expression, which correlated with reduced nitric oxide accumulation. In addition, KHG26792 attenuated hypoxiainduced protein nitration, reactive oxygen species production, and NADPH oxidase activity. These effects were accompanied by the suppression of hypoxia-induced protein expression of hypoxia-inducible factor 1-alpha and NADPH oxidase-2. Although the clinical relevance of our findings remains to be determined, these data results suggest that KHG26792 prevents hypoxia-induced toxicity by suppressing microglial activation. [BMB Reports 2016; 49(12): 687-692].


Subject(s)
Azetidines/pharmacology , Cell Hypoxia , Microglia/drug effects , Naphthalenes/pharmacology , Protective Agents/pharmacology , Animals , Azetidines/chemical synthesis , Azetidines/chemistry , Blotting, Western , Caspase 3/metabolism , Cell Line , Cell Survival/drug effects , Gene Expression/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microglia/cytology , Microglia/metabolism , Microscopy, Fluorescence , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Naphthalenes/chemical synthesis , Naphthalenes/chemistry , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Protective Agents/chemical synthesis , Protective Agents/chemistry , Reactive Oxygen Species/metabolism
17.
ACS Comb Sci ; 18(8): 499-506, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27362292

ABSTRACT

A 1,3,4-thiadiazole library was constructed by solid-phase organic synthesis. The key step of this solid-phase synthesis involves the preparation of polymer-bound 2-amido-5-amino-1,3,4-thiadiazole resin by the cyclization of thiosemicarbazide resin using p-TsCl as the desulfurative agent, followed by the functionalization of the resin by alkylation, acylation, alkylation/acylation, and Suzuki coupling reactions. Both the alkylation and acylation reactions chemoselectively occurred at the 2-amide position of 2-amido-5-amino-1,3,4-thiadiazole resin and the 5-amine position of 2-amido-5-amino-1,3,4-thiadiazole resin, respectively. Finally, these functionalized 1,3,4-thiadiazole resins were treated with trifluoroacetic acid in dichloromethane, affording diverse 1,3,4-thiadiazole analogs in high yields and purities. The 1,3,4-thiadiazole analogs show a different distribution of physicochemical and biological properties compared with our previously constructed 1,3,4-oxadiazole and 1,3,4-thiadiazole libraries in a range of orally available drug properties.


Subject(s)
Resins, Synthetic/chemistry , Semicarbazides/chemistry , Small Molecule Libraries/chemistry , Thiadiazoles/chemistry , Acylation , Alkylation , Amides/chemistry , Amines/chemistry , Combinatorial Chemistry Techniques , Cyclization , Molecular Structure , Oxadiazoles/chemistry , Small Molecule Libraries/chemical synthesis , Solid-Phase Synthesis Techniques , Thiadiazoles/chemical synthesis
18.
Free Radic Res ; 50(6): 678-90, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27002191

ABSTRACT

Recently, we have reported that N-adamantyl-4-methylthiazol-2-amine (KHG26693) successfully reduced the production of oxidative stress in streptozotocin-induced diabetic rats and lipopolysaccharide-induced BV-2 microglial cells by increasing their antioxidant capacity. However, antioxidative effects of KHG26693 against Aß (Aß)-induced oxidative stress have not yet been reported. In the present study, we further investigated the antioxidative function of KHG26693 in Aß-mediated primary cultured cortical neurons. We showed here that KHG26693 attenuated Aß-induced cytotoxicity, increase of Bax/Bcl-2 ratio, elevation of caspase-3 expression, and impairment of mitochondrial membrane potential in cultured primary cortical neurons. KHG26693 also decreases the Aß-mediated formation of malondialdehyde, reactive oxygen species, and NO production by decreasing nitric oxide synthase (iNOS) and NADPH oxidase level. Moreover, KHG26693 suppress the Aß-induced oxidative stress through a possible mechanism involving attenuation of GSH and antioxidant enzyme activities such as glutathione reductase and glutathione peroxidase (GPx). Finally, pretreatment of cortical neurons with KHG26693 significantly reduced the Aß-induced protein oxidation and nitration. To our knowledge, this is the first report, showing that KHG26693 significantly attenuates Aß-induced oxidative stress in primary cortical neurons, and may prove attractive strategies to reduce Aß-induced neural cell death.


Subject(s)
Adamantane/analogs & derivatives , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Neurons/metabolism , Thiazoles/metabolism , Adamantane/metabolism , Animals , Oxidation-Reduction , Oxidative Stress , Rats , Rats, Sprague-Dawley
19.
Neuropharmacology ; 102: 32-41, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26522435

ABSTRACT

It is well documented that a maternal immune response to infection during pregnancy can cause neurodevelopmental damage. We demonstrate in our current study that maternally administered 2-cyclopropylimino-3-methyl-1,3-thiazoline hydrochloride (KHG26377), a novel thiazole derivative, prevents fetal malformations and neurodevelopmental deficits in offspring by blocking lipopolysaccharide (LPS)-induced inflammation. Administration of KHG26377 effectively regulated LPS-induced inflammatory markers and mediators such as soluble intercellular adhesion molecule-1, se-Selectin, macrophage chemoattractant protein-1, and cytokine-induced neutrophil chemoattractant-1 in the maternal serum. Furthermore, maternally administered KHG26377 showed an inhibitory effect on the LPS-induced developmental toxicity by selectively suppressing the TNF-α level in maternal serum, amniotic fluid, placenta, fetal liver, and fetal brain as well as by suppression of LPS-induced nitric oxide synthase (iNOS), cyclooxygenase 2 (COX-2), and myelin basic protein (MBP) levels in the fetal brain. In addition, pretreatment of neuronal cells with KHG26377 effectively reestablished the cell body morphology and microtubule-associated protein 2 (MAP2) staining compared to the LPS-treated group in cortex primary neuronal cultures. Although the clinical relevance of our findings remains to be determined, our results provide novel insights into KHG26377 as a possible therapeutic agent to protect fetuses against various inflammatory responses.


Subject(s)
Brain/drug effects , Cytokines/metabolism , Fetus/drug effects , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Neurons/drug effects , Thiazoles/pharmacology , Animals , Brain/cytology , Brain/metabolism , Cell Shape/drug effects , Cells, Cultured , Female , Fetus/cytology , Fetus/metabolism , Male , Mice , Microtubule-Associated Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Pregnancy
20.
ACS Comb Sci ; 17(12): 732-41, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26524617

ABSTRACT

A 2-amino/amido-1,3,4-oxadiazole and 1,3,4-thiadiazole library has been constructed on solid-phase organic synthesis. The key step on this solid-phase synthesis involves the preparation of polymer-bound 2-amino-1,3,4-oxadiazole and 1,3,4-thiadiazole core skeleton resin by cyclization of thiosemicarbazide with EDC·HCl and p-TsCl, respectively. The resulting core skeleton undergoes functionalization reaction with various electrophiles such as alkyl halides, and acid chlorides to generate N-alkylamino and N-acylamino-1,3,4-oxadiazole, and 1,3,4-thiadiazole resin, respectively. Finally, the 2-amino and 2-amido-1,3,4-oxadiazole and 1,3,4-thiadiazole library was then generated in good yields and high purities by cleavage of the respective resin under trifluoroacetic acid(TFA) in dichloromethane(DCM). The constructed library shows reasonable, oral bioavailability drug properties as determine by using the Lipinski's Rule and similar parameters.


Subject(s)
Oxadiazoles/chemistry , Semicarbazides/chemical synthesis , Thiadiazoles/chemistry , Cyclization , Indicators and Reagents/chemistry , Molecular Structure , Semicarbazides/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...