Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
1.
Int J Mol Sci ; 22(3)2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530496

RESUMEN

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.


Asunto(s)
Apoptosis/genética , Calgranulina A/genética , Regulación de la Expresión Génica , Hipoxia/genética , Hipoxia/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Animales , Biomarcadores , Calgranulina A/metabolismo , Línea Celular , Citocinas/metabolismo , Susceptibilidad a Enfermedades , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Técnicas de Silenciamiento del Gen , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Mediadores de Inflamación/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Fosforilación
2.
Biochem Biophys Res Commun ; 495(1): 151-156, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29111328

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Azetidinas/farmacología , Inflamasomas/inmunología , Microglía/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Naftalenos/farmacología , Animales , Línea Celular , Interleucina-1beta/inmunología , Interleucina-6/inmunología , Lipopolisacáridos/inmunología , Ratones , Microglía/inmunología , Especies Reactivas de Oxígeno/inmunología , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/inmunología
3.
Adv Sci (Weinh) ; 11(21): e2308763, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38552157

RESUMEN

The CRISPR/Cas system has been introduced as an innovative tool for therapy, however achieving specific delivery to the target has been a major challenge. Here, an antibody-CRISPR/Cas conjugate platform that enables specific delivery and target gene editing in HER2-positive cancer is introduced. The CRISPR/Cas system by replacing specific residues of Cas9 with an unnatural amino acid is engineered, that can be complexed with a nanocarrier and bioorthogonally functionalized with a monoclonal antibody targeting HER2. The resultant antibody-conjugated CRISPR/Cas nanocomplexes can be specifically delivered and induce gene editing in HER2-positive cancer cells in vitro. It is demonstrated that the in vivo delivery of the antibody-CRISPR/Cas nanocomplexes can effectively disrupt the plk1 gene in HER2-positive ovarian cancer, resulting in substantial suppression of tumor growth. The current study presents a useful therapeutic platform for antibody-mediated delivery of CRISPR/Cas for the treatment of various cancers and genetic diseases.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Humanos , Ratones , Animales , Femenino , Línea Celular Tumoral , Neoplasias Ováricas/genética , Neoplasias Ováricas/terapia , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Anticuerpos Monoclonales/genética , Neoplasias/terapia , Neoplasias/genética , Receptor ErbB-2/genética
4.
J Org Chem ; 78(2): 438-44, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23215154

RESUMEN

A regioselective, reagent-based method for the cyclization reaction of 2-amino-1,3,4-oxadiazole and 2-amino-1,3,4-thiadiazole core skeletons is described. The thiosemicarbazide intermediate 3 was reacted with EDC·HCl in DMSO or p-TsCl, triethylamine in N-methyl-2-pyrrolidone to give the corresponding 2-amino-1,3,4-oxadiazoles 4 and 2-amino-1,3,4-thiadiazoles 5 through regioselcective cyclization processes. The regioselectivity was affected by both R(1) and R(2) in p-TsCl mediated cyclization. It is shown in select sets of thiosemicarbazide 3 with R(1)(benzyl) and R(2)(phenyl). 2-Amino-1,3,4-oxadiazole 4 was also shown in the reaction of p-TsCl mediated cyclization. The resulting 2-amino-1,3,4-oxadiazole and 2-amino-1,3,4-thiadiazole core skeleton are functionalized with various electrophiles such as alkyl halide, acid halides, and sulfornyl chloride in high yields.


Asunto(s)
Indicadores y Reactivos/química , Oxadiazoles/síntesis química , Oxazoles/síntesis química , Semicarbacidas/síntesis química , Tiadiazoles/síntesis química , Ciclización , Espectroscopía de Resonancia Magnética , Estructura Molecular , Oxadiazoles/química , Oxazoles/química , Semicarbacidas/química , Estereoisomerismo , Tiadiazoles/química
5.
Adv Sci (Weinh) ; 10(27): e2302253, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37485817

RESUMEN

Bioconjugation of proteins can substantially expand the opportunities in biopharmaceutical development, however, applications are limited for the gene editing machinery despite its tremendous therapeutic potential. Here, a self-delivered nanomedicine platform based on bioorthogonal CRISPR/Cas9 conjugates, which can be armed with a chemotherapeutic drug for combinatorial therapy is introduced. It is demonstrated that multi-functionalized Cas9 with a drug and polymer can form self-condensed nanocomplexes, and induce significant gene editing upon delivery while avoiding the use of a conventional carrier formulation. It is shown that the nanomedicine platform can be applied for combinatorial therapy by incorporating the anti-cancer drug olaparib and targeting the RAD52 gene, leading to significant anti-tumor effects in BRCA-mutant cancer. The current development provides a versatile nanomedicine platform for combination treatment of human diseases such as cancer.


Asunto(s)
Sistemas CRISPR-Cas , Técnicas de Transferencia de Gen , Humanos , Sistemas CRISPR-Cas/genética , Preparaciones Farmacéuticas , Nanomedicina , Edición Génica
6.
BMB Rep ; 55(9): 447-452, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35651331

RESUMEN

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].


Asunto(s)
Lipopolisacáridos , MicroARNs , Antiinflamatorios , Apoptosis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Caspasas/metabolismo , Citocinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Inflamasomas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos/farmacología , MicroARNs/genética , MicroARNs/metabolismo , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
Biochip J ; 16(4): 441-450, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36091642

RESUMEN

We introduce a lateral flow assay (LFA) integrated with a modified isothermal nucleic acid amplification procedure for rapid and simple genetic testing. Padlock probes specific for the target DNA were designed for ligation, followed by rolling circle amplification (RCA) using capture ligand-modified oligonucleotides as primers. After hybridization with detection linker probes, the amplified target DNA is flowed through an LFA membrane strip for binding of gold nanoparticles as the substrate for colorimetric detection. We established and validated the "RCA-LFA" method for detection of mecA, the antibiotic resistance gene for methicillin-resistant Staphylococcus aureus (MRSA). The assay was optimized using various concentrations of primers and probes for RCA and LFA, respectively. The sensitivity was determined by performing RCA-LFA using various amounts of mecA target DNA, showing a detection limit of ~ 1.3 fmol. The specificity of the assay was examined using target DNAs for other resistance genes as the controls, which demonstrated positive detection signals only for mecA DNA, when added either individually or in combinations with the control targets. Furthermore, applying the RCA-LFA method using specifically designed probes for RNA-dependent RNA polymerase (RdRp) and receptor binding domain (RBD) gene for SARS-CoV-2, which demonstrated feasibility of the method for viral gene targets. The current method suggests a useful platform which can be universally applied for various nucleic acid targets, allowing rapid and sensitive diagnosis at point-of-care. Supplementary Information: The online version contains supplementary material available at 10.1007/s13206-022-00080-1.

8.
BMB Rep ; 54(11): 557-562, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34353430

RESUMEN

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].


Asunto(s)
Antiinflamatorios/farmacología , Bromuros/química , Inflamasomas/efectos de los fármacos , Inflamación/tratamiento farmacológico , Lipopolisacáridos/toxicidad , Microglía/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Animales , Antiinflamatorios/química , Citocinas/metabolismo , Inflamasomas/inmunología , Inflamasomas/metabolismo , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Peroxidación de Lípido , Activación de Macrófagos , Ratones , Microglía/metabolismo , Microglía/patología , FN-kappa B/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
9.
BMB Rep ; 53(10): 527-532, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32635984

RESUMEN

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].


Asunto(s)
Adamantano/análogos & derivados , Autofagia/efectos de los fármacos , Neuronas/metabolismo , Tiazoles/farmacología , Adamantano/metabolismo , Adamantano/farmacología , Animales , Antioxidantes/farmacología , Muerte Celular Autofágica , Autofagia/fisiología , Corteza Cerebral/metabolismo , Ácido Glutámico/efectos adversos , Ácido Glutámico/metabolismo , Glutatión/metabolismo , Glutatión Peroxidasa/metabolismo , Neuronas/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Tiazoles/metabolismo
10.
Front Immunol ; 11: 1258, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32903598

RESUMEN

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.


Asunto(s)
Apoptosis , Calgranulina A/metabolismo , Calgranulina B/metabolismo , Síndrome Hipereosinofílico/etiología , Síndrome Hipereosinofílico/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Células Cultivadas , Enfermedad Crónica , Resistencia a Antineoplásicos , Femenino , Expresión Génica , Humanos , Síndrome Hipereosinofílico/tratamiento farmacológico , Síndrome Hipereosinofílico/patología , Mesilato de Imatinib/farmacología , Mesilato de Imatinib/uso terapéutico , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes
11.
BMB Rep ; 52(10): 613-618, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30940325

RESUMEN

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].


Asunto(s)
Antiinflamatorios/farmacología , Inflamasomas/metabolismo , Microglía/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Ácido Tióctico/farmacología , Animales , Caspasa 1/metabolismo , Línea Celular , Citocinas/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Lipopolisacáridos/farmacología , Activación de Macrófagos , Glicoproteínas de Membrana/metabolismo , Ratones , Microglía/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Eur J Pharmacol ; 856: 172412, 2019 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-31129157

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzotiazoles/farmacología , Ácido Glutámico/efectos adversos , Inflamasomas/metabolismo , Mitocondrias/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuroblastoma/patología , Adenosina Trifosfato/metabolismo , Calcio/metabolismo , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/patología , Estrés Oxidativo/efectos de los fármacos
13.
Biotechnol Appl Biochem ; 51(Pt 2): 107-10, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18248328

RESUMEN

In the nervous system, GDH (glutamate dehydrogenase) is enriched in astrocytes and is important for recycling glutamate, a major excitatory neurotransmitter. The function of hGDH (human GDH) may be important in neurodegenerative diseases such as Parkinson's disease. To test the effect of decreased hGDH expression, several vector-based plasmidlinked hGDH siRNAs (small interfering RNAs) were expressed intracellularly in BE(2)C human neuroblastoma cells. Immunoblotting and reverse-transcription-PCR confirmed that expression of hGDH protein and mRNA was knocked down by co-transfection with phGDH-siRNA vectors in BE(2)C human neuroblastoma cells. TUNEL (terminal uridine deoxynucleotidyl transferase dUTP nick-end labelling) and DNA fragmentation assays 48 h after transfection of phGDH-siRNAs revealed that inhibition of hGDH expression induced cellular apoptosis and activated phospho-ERK1/2 (phospho-extracellular-signal-regulated kinase 1/2). These findings show that inhibition of hGDH expression by siRNA is related to apoptosis in neuronal cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Glutamato Deshidrogenasa/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , ARN Interferente Pequeño/farmacología , Fragmentación del ADN/efectos de los fármacos , Perfilación de la Expresión Génica , Vectores Genéticos/genética , Glutamato Deshidrogenasa/biosíntesis , Glutamato Deshidrogenasa/deficiencia , Humanos , Immunoblotting , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección
14.
ACS Comb Sci ; 20(2): 82-97, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29309123

RESUMEN

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.


Asunto(s)
Oxadiazoles/síntesis química , Bibliotecas de Moléculas Pequeñas/síntesis química , Técnicas de Síntesis en Fase Sólida/métodos , Tiadiazoles/síntesis química , Amidas/química , Aminas/química , Aminoácidos/química , Técnicas Químicas Combinatorias/métodos , Ciclización , Estructura Molecular , Semicarbacidas/química , Relación Estructura-Actividad , Tiourea/química
15.
ACS Comb Sci ; 20(5): 282-291, 2018 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-29557646

RESUMEN

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.


Asunto(s)
Bencimidazoles/síntesis química , Resinas Sintéticas/química , Técnicas de Síntesis en Fase Sólida/métodos , Tiourea/química , Acilación , Aminas/química , Ciclización , Bibliotecas de Moléculas Pequeñas/química
16.
BMB Rep ; 51(11): 590-595, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29966582

RESUMEN

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].


Asunto(s)
1-Metil-4-fenilpiridinio/toxicidad , Azetidinas/farmacología , Citoprotección/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Naftalenos/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Estrés Oxidativo/efectos de los fármacos , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Microglía/efectos de los fármacos , Microglía/fisiología , Mitocondrias/metabolismo , Neuronas/fisiología , Síndromes de Neurotoxicidad/patología , Síndromes de Neurotoxicidad/prevención & control , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
17.
Toxicology ; 239(3): 156-66, 2007 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-17714846

RESUMEN

We have investigated the effect of KHG21834, a benzothiazole derivative, on the amyloid beta protein (Abeta)-induced cell death in rat pheochromocytoma (PC12) cells and rat cortical and mesencephalic neuron-glia cultures. KHG21834 attenuated the Abeta(25-35)-induced apoptotic death in PC12 cells determined by characteristic morphological alterations and positive in situ terminal end-labeling (TUNEL). In the cortical neuron-glia cultures, KHG21834 reduced the Abeta(25-35)-induced apoptosis determined by TUNEL staining. Immunocytochemical analysis and Western blot analysis of Abeta(25-35)-induced neurotoxicity in mesencephalic neuron-glia cultures with anti-tyrosine hydroxylase (TH) antibody showed that Abeta(25-35) decreased the expression of TH protein by 60% and KHG21834 significantly attenuated the Abeta(25-35)-induced reduction in the expression of TH. Moreover, KHG21834 attenuates Abeta(25-35)-induced toxicity concomitant with the reduction of activation of extracellular signal-regulated kinase (ERK)1/2 to a lesser extent. ERK1 was more sensitively affected than ERK2 in attenuation of Abeta(25-35)-induced phosphorylation by KHG21834. These results demonstrated that KHG21834 was capable of protecting neuronal cells from Abeta(25-35)-induced degeneration.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Benzotiazoles/farmacología , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/toxicidad , Animales , Anticuerpos/inmunología , Apoptosis/efectos de los fármacos , Benzotiazoles/química , Western Blotting , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Fragmentación del ADN/efectos de los fármacos , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Mesencéfalo/efectos de los fármacos , Mesencéfalo/metabolismo , Mesencéfalo/patología , Microscopía Confocal , Microscopía Fluorescente , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Estructura Molecular , Neuronas/metabolismo , Neuronas/patología , Fármacos Neuroprotectores/química , Células PC12 , Ratas , Ratas Sprague-Dawley , Tirosina 3-Monooxigenasa/inmunología , Tirosina 3-Monooxigenasa/metabolismo
18.
J Biochem Mol Biol ; 40(6): 1077-82, 2007 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-18047806

RESUMEN

Human glutamate dehydrogenase exists in hGDH1 (housekeeping isozyme) and in hGDH2 (nerve-specific isozyme), which differ markedly in their allosteric regulation. In the nervous system, GDH is enriched in astrocytes and is important for recycling glutamate, a major excitatory neurotransmitter during neurotransmission. Chloroquine has been known to be a potent inhibitor of house-keeping GDH1 in permeabilized liver and kidney-cortex of rabbit. However, the effects of chloroquine on nerve-specific GDH2 have not been reported yet. In the present study, we have investigated the effects of chloroquine on hGDH2 at various conditions and showed that chloroquine could inhibit the activity of hGDH2 at dose-dependent manner. Studies of the chloroquine inhibition on enzyme activity revealed that hGDH2 was relatively less sensitive to chloroquine inhibition than house-keeping hGDH1. Incubation of hGDH2 was uncompetitive with respect of NADH and non-competitive with respect of 2-oxoglutarate. The inhibitory effect of chloroquine on hGDH2 was abolished, although in part, by the presence of ADP and L-leucine, whereas GTP did not change the sensitivity to chloroquine inhibition. Our results show a possibility that chloroquine may be used in regulating GDH activity and subsequently glutamate concentration in the central nervous system.


Asunto(s)
Cloroquina/farmacología , Glutamato Deshidrogenasa/antagonistas & inhibidores , Regulación Alostérica , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/enzimología , Inhibidores Enzimáticos/farmacología , Escherichia coli/genética , Glutamato Deshidrogenasa/genética , Humanos , Técnicas In Vitro , Isoenzimas/antagonistas & inhibidores , Isoenzimas/genética , Cinética , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/enzimología
19.
J Biochem Mol Biol ; 40(5): 690-6, 2007 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-17927902

RESUMEN

There are conflicting views for the polymerization process of human UDP-glucose dehydrogenase (UGDH) and no clear evidence has been reported yet. Based on crystal coordinates for Streptococcus pyogenes UGDH, we made double mutant A222Q/S233G. The double mutagenesis had no effects on expression, stability, and secondary structure. Interestingly, A222Q/S233G was a dimeric form and showed an UGDH activity, although it showed increased Km values for substrates. These results suggest that Ala222 and Ser233 play an important role in maintaining the hexameric structure and the reduced binding affinities for substrates are attributable to its altered subunit communication although quaternary structure may not be critical for catalysis.


Asunto(s)
Mutación , Uridina Difosfato Glucosa Deshidrogenasa/química , Uridina Difosfato Glucosa Deshidrogenasa/genética , Alanina/química , Alanina/genética , Alanina/metabolismo , Sustitución de Aminoácidos , Sitios de Unión/genética , Cromatografía en Gel , Cromatografía Líquida de Alta Presión , Humanos , Cinética , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Serina/química , Serina/genética , Serina/metabolismo , Relación Estructura-Actividad , Uridina Difosfato Glucosa Deshidrogenasa/metabolismo
20.
ACS Comb Sci ; 19(12): 738-747, 2017 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-29095593

RESUMEN

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.


Asunto(s)
Imidazoles/química , Bibliotecas de Moléculas Pequeñas/química , Compuestos de Sulfhidrilo/química , Tionas/química , Ciclización , Imidazoles/síntesis química , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/síntesis química , Técnicas de Síntesis en Fase Sólida , Compuestos de Sulfhidrilo/síntesis química , Termodinámica , Tionas/síntesis química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA