Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Chem Inf Model ; 64(5): 1615-1627, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38356220

RESUMEN

Cancer immunotherapy harnesses the immune system to combat tumors and has emerged as a major cancer treatment modality. The PD-1/PD-L1 immune checkpoint modulates interactions between tumor cells and T cells and has been extensively targeted in cancer immunotherapy. However, the monoclonal antibodies known to target this immune checkpoint have considerable side effects, and novel PD-1/PD-L1 inhibitors are therefore required. Herein, a peptide inhibitor to disrupt PD-1/PD-L1 interactions was designed through structure-driven phage display engineering coupled to computational modification and optimization. BetaPb, a novel peptide library constructed by using the known structure of PD-1/PD-L, was used to develop inhibitors against the immune checkpoint, and specific peptides with high affinity toward PD-1 were screened through enzyme-linked immunosorbent assays, homogeneous time-resolved fluorescence, and biolayer interferometry. A potential inhibitor, B8, was preliminarily screened through biopanning. The binding affinity of B8 toward PD-1 was confirmed through computation-aided optimization. Assessment of B8 variants (B8.1, B8.2, B8.3, B8.4, and B8.5) demonstrated their attenuation of PD-1/PD-L1 interactions. B8.4 exhibited the strongest attenuation efficiency at a half-maximal effective concentration of 0.1 µM and the strongest binding affinity to PD-1 (equilibrium dissociation constant = 0.1 µM). B8.4 outperformed the known PD-1/PD-L1 interaction inhibitor PL120131 in disrupting PD-1/PD-L1 interactions, revealing that B8.4 has remarkable potential for modification to yield an antitumor agent. This study provides valuable information for the future development of peptide-based drugs, therapeutics, and immunotherapies for cancer.


Asunto(s)
Bacteriófagos , Neoplasias , Humanos , Inhibidores de Puntos de Control Inmunológico , Receptor de Muerte Celular Programada 1/química , Antígeno B7-H1/química , Péptidos/farmacología , Péptidos/química , Bacteriófagos/metabolismo
2.
Cell Mol Life Sci ; 80(4): 101, 2023 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-36935456

RESUMEN

Pdia4 has been characterized as a key protein that positively regulates ß-cell failure and diabetes via ROS regulation. Here, we investigated the function and mechanism of PS1, a Pdia4 inhibitor, in ß-cells and diabetes. We found that PS1 had an IC50 of 4 µM for Pdia4. Furthermore, PS1 alone and in combination with metformin significantly reversed diabetes in db/db mice, 6 to 7 mice per group, as evidenced by blood glucose, glycosylated hemoglobin A1c (HbA1c), glucose tolerance test, diabetic incidence, survival and longevity (P < 0.05 or less). Accordingly, PS1 reduced cell death and dysfunction in the pancreatic ß-islets of db/db mice as exemplified by serum insulin, serum c-peptide, reactive oxygen species (ROS), islet atrophy, and homeostatic model assessment (HOMA) indices (P < 0.05 or less). Moreover, PS1 decreased cell death in the ß-islets of db/db mice. Mechanistic studies showed that PS1 significantly increased cell survival and insulin secretion in Min6 cells in response to high glucose (P < 0.05 or less). This increase could be attributed to a reduction in ROS production and the activity of electron transport chain complex 1 (ETC C1) and Nox in Min6 cells by PS1. Further, we found that PS1 inhibited the enzymatic activity of Pdia4 and mitigated the interaction between Pdia4 and Ndufs3 or p22 in Min6 cells (P < 0.01 or less). Taken together, this work demonstrates that PS1 negatively regulated ß-cell pathogenesis and diabetes via reduction of ROS production involving the Pdia4/Ndufs3 and Pdia4/p22 cascades.


Asunto(s)
Diabetes Mellitus Tipo 2 , Células Secretoras de Insulina , Ratones , Animales , Diabetes Mellitus Tipo 2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Glucemia/metabolismo , Ratones Endogámicos , Ratones Endogámicos C57BL , Proteína Disulfuro Isomerasas/metabolismo
3.
Biochem Biophys Res Commun ; 680: 161-170, 2023 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-37741263

RESUMEN

Studies have shown that the high expression of EphA4 in gastric cancer tissues may correlate with unfavorable clinical pathological characteristics. Therefore, EphA4 may be an effective target for treating gastric cancer in addition to HER-2/neu. In this study, generated scFv S3 can bind endogenous EphA4 of gastric cancer cells and has significant membrane staining. Additionally, scFv S3 binding to EphA4 inhibits the growth and migration of cancer cells and the growth induction that ephrinA1 generates in gastric cancer cells. We found that EphA4 molecules may degrade through antibody treatment of cells, and the increase in LAMP1 and LAMP2 indicates that lysosome is involved in the degradation. The scFv S3 administration leads to the signals pAKT, pERK, and pSTAT3 decrease in cancer cells. The xenograft model of HER-2/neu low expressing gastric cancer cell SNU-16 exhibits better therapeutic effects by scFv S3 than trastuzumab scFv. The scFv S3 administration in vivo can degrade EphA4 molecules in tumor tissues, decreasing Ki67 and increasing cleaved C3 molecule expression. Furthermore, we identified and validated that scFv S3 generates essential ionic bonding with R162 on EphA4. The antibody may provide effective treatment for patients with gastric cancer and abnormal activation or overexpression of EphA4 signaling.


Asunto(s)
Anticuerpos de Cadena Única , Neoplasias Gástricas , Humanos , Transducción de Señal , Neoplasias Gástricas/tratamiento farmacológico , Anticuerpos de Cadena Única/farmacología , Animales
4.
Biochem Biophys Res Commun ; 688: 149214, 2023 12 25.
Artículo en Inglés | MEDLINE | ID: mdl-37951154

RESUMEN

Pancreatic adenocarcinoma, a highly aggressive form of cancer with a poor prognosis, necessitates the development of innovative treatment strategies. Our prior research showcased the growth-inhibiting effects of the anti-EphA2 antibody drug hSD5 on pancreatic cancer tumors. This antibody targets and induces the degradation of the EphA2 receptor while also prompting the antibody's internalization. A deeper dive into the hSD5 Fab crystallographic structure and docking studies revealed that hSD5's CDRH3 drives the primary interaction between hSD5 and the EphA2 active site. In this study, we developed a novel antibody-drug conjugate (ADC)-the auristatin-based hSD5-vedotin specifically targeting EphA2 in pancreatic cancer cells. This ADC aims at the tumor-specific antigen EphA2, triggering endocytosis and releasing the conjugated payload molecule Monomethyl auristatin E (MMAE), amplifying the tumor-killing effect. Upon cellular entry, hSD5-vedotin demonstrated an impressive tumor-killing response, inhibiting tumor cell growth and promoting apoptosis even at lower antibody concentrations. In a pancreatic cancer xenograft animal model, hSD5-vedotin showcased the potential to suppress tumor growth entirely. Notably, potential immune resistance responses were also observed in recurrent pancreatic cancer tumors. Our empirical results underscore the possibility of developing hSD5-vedotin further, which we anticipate will have a broader and more potent therapeutic impact on pancreatic cancer and other EphA2-related cancers.


Asunto(s)
Adenocarcinoma , Inmunoconjugados , Neoplasias Pancreáticas , Animales , Humanos , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Inmunoconjugados/química , Neoplasias Pancreáticas/patología , Adenocarcinoma/tratamiento farmacológico , Línea Celular Tumoral , Recurrencia Local de Neoplasia , Ensayos Antitumor por Modelo de Xenoinjerto , Neoplasias Pancreáticas
5.
J Nat Prod ; 86(6): 1428-1436, 2023 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-37267066

RESUMEN

Traditional herbal medicine offers opportunities to discover novel therapeutics against SARS-CoV-2 mutation. The dried aerial part of mint (Mentha canadensis L.) was chosen for bioactivity-guided extraction. Seven constituents were isolated and characterized by nuclear magnetic resonance (NMR) and mass spectrometry (MS). Syringic acid and methyl rosmarinate were evaluated in drug combination treatment. Ten amide derivatives of methyl rosmarinate were synthesized, and the dodecyl (13) and 3-ethylphenyl (19) derivatives demonstrated significant improvement in the anti-SARS-CoV-2 plaque reduction assay, achieving IC50 of 0.77 and 2.70 µM, respectively, against Omicron BA.1 as compared to methyl rosmarinate's IC50 of 57.0 µM. Spike protein binding and 3CLpro inhibition assays were performed to explore the viral inhibition mechanism. Molecular docking of compounds 13 and 19 to 3CLpro was performed to reveal potential interaction. In summary, natural products with anti-Omicron BA.1 activity were isolated from Mentha canadensis and derivatives of methyl rosmarinate were synthesized, showing 21- to 74-fold improvement in antiviral activity against Omicron BA.1.


Asunto(s)
Productos Biológicos , COVID-19 , Mentha , Antivirales/farmacología , Simulación del Acoplamiento Molecular , SARS-CoV-2 , Antiinflamatorios no Esteroideos , Antioxidantes , Productos Biológicos/farmacología , Cinamatos , Depsidos
6.
Bioorg Chem ; 138: 106581, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37172437

RESUMEN

Inhibition of steroid sulfatase (STS) decreases estrogen production and thus, suppresses tumor proliferation. Inspired by irosustat, the first STS inhibitor in clinical trials, we explored twenty-one tricyclic and tetra-heterocyclic coumarin-based derivatives. Their STS enzyme kinetic parameters, docking models, and cytotoxicity toward breast cancer and normal cells were evaluated. Tricyclic derivative 9e and tetracyclic derivative 10c were the most promising irreversible inhibitors developed in this study, with KI of 0.05 and 0.4 nM, and kinact/KI ratios of 28.6 and 19.1 nM-1min-1 on human placenta STS, respectively.


Asunto(s)
Neoplasias de la Mama , Esteril-Sulfatasa , Embarazo , Femenino , Humanos , Cinética , Relación Estructura-Actividad , Ácidos Sulfónicos , Neoplasias de la Mama/tratamiento farmacológico , Cumarinas/farmacología , Cumarinas/uso terapéutico , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico
7.
Mar Drugs ; 21(4)2023 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-37103364

RESUMEN

Immunogenic cell death (ICD) refers to a type of cell death that stimulates immune responses. It is characterized by the surface exposure of damage-associated molecular patterns (DAMPs), which can facilitate the uptake of antigens by dendritic cells (DCs) and stimulate DC activation, resulting in T cell immunity. The activation of immune responses through ICD has been proposed as a promising approach for cancer immunotherapy. The marine natural product crassolide, a cembranolide isolated from the Formosan soft coral Lobophytum michaelae, has been shown to have cytotoxic effects on cancer cells. In this study, we investigated the effects of crassolide on the induction of ICD, the expression of immune checkpoint molecules and cell adhesion molecules, as well as tumor growth in a murine 4T1 mammary carcinoma model. Immunofluorescence staining for DAMP ectolocalization, Western blotting for protein expression and Z'-LYTE kinase assay for kinase activity were performed. The results showed that crassolide significantly increased ICD and slightly decreased the expression level of CD24 on the surface of murine mammary carcinoma cells. An orthotopic tumor engraftment of 4T1 carcinoma cells indicated that crassolide-treated tumor cell lysates stimulate anti-tumor immunity against tumor growth. Crassolide was also found to be a blocker of mitogen-activated protein kinase 14 activation. This study highlights the immunotherapeutic effects of crassolide on the activation of anticancer immune responses and suggests the potential clinical use of crassolide as a novel treatment for breast cancer.


Asunto(s)
Antozoos , Antineoplásicos , Carcinoma , Proteína Quinasa 14 Activada por Mitógenos , Animales , Ratones , Muerte Celular Inmunogénica , Antineoplásicos/farmacología , Línea Celular Tumoral
8.
Pharmacol Res ; 184: 106424, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36064077

RESUMEN

The global COVID-19 pandemic remains a critical public health threat, as existing vaccines and drugs appear insufficient to halt the rapid transmission. During an outbreak from May to August 2021 in Taiwan, patients with severe COVID-19 were administered NRICM102, which was a traditional Chinese medicine (TCM) formula developed based on its predecessor NRICM101 approved for treating mild cases. This study aimed to explore the mechanism of NRICM102 in ameliorating severe COVID-19-related embolic and fibrotic pulmonary injury. NRICM102 was found to disrupt spike protein/ACE2 interaction, 3CL protease activity, reduce activation of neutrophils, monocytes and expression of cytokines (TNF-α, IL-1ß, IL-6, IL-8), chemokines (MCP-1, MIP-1, RANTES) and proinflammatory receptor (TLR4). NRICM102 also inhibited the spread of virus and progression to embolic and fibrotic pulmonary injury through reducing prothrombotic (vWF, PAI-1, NET) and fibrotic (c-Kit, SCF) factors, and reducing alveolar type I (AT1) and type II (AT2) cell apoptosis. NRICM102 may exhibit its protective capability via regulation of TLRs, JAK/STAT, PI3K/AKT, and NET signaling pathways. The study demonstrates the ability of NRICM102 to ameliorate severe COVID-19-related embolic and fibrotic pulmonary injury in vitro and in vivo and elucidates the underlying mechanisms.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Lesión Pulmonar , Embolia Pulmonar , Enzima Convertidora de Angiotensina 2 , Quimiocina CCL5 , Citocinas , Fibrosis , Humanos , Interleucina-6/metabolismo , Interleucina-8 , Lesión Pulmonar/tratamiento farmacológico , Pandemias , Fosfatidilinositol 3-Quinasas , Inhibidor 1 de Activador Plasminogénico , Proteínas Proto-Oncogénicas c-akt , Embolia Pulmonar/tratamiento farmacológico , Glicoproteína de la Espiga del Coronavirus , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Factor de von Willebrand
9.
Pharmacol Res ; 184: 106412, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36007774

RESUMEN

BACKGROUND: Viral- and host-targeted traditional Chinese medicine (TCM) formulae NRICM101 and NRICM102 were administered to hospitalized patients with COVID-19 during the mid-2021 outbreak in Taiwan. We report the outcomes by measuring the risks of intubation or admission to intensive care unit (ICU) for patients requiring no oxygen support, and death for those requiring oxygen therapy. METHODS: This multicenter retrospective study retrieved data of 840 patients admitted to 9 hospitals between May 1 and July 26, 2021. After propensity score matching, 302 patients (151 received NRICM101 and 151 did not) and 246 patients (123 received NRICM102 and 123 did not) were included in the analysis to assess relative risks. RESULTS: During the 30-day observation period, no endpoint occurred in the patients receiving NRICM101 plus usual care while 14 (9.27%) in the group receiving only usual care were intubated or admitted to ICU. The numbers of deceased patients were 7 (5.69%) in the group receiving NRICM102 plus usual care and 27 (21.95%) in the usual care group. No patients receiving NRICM101 transitioned to a more severe status; NRICM102 users were 74.07% less likely to die than non-users (relative risk= 25.93%, 95% confidence interval 11.73%-57.29%). CONCLUSION: NRICM101 and NRICM102 were significantly associated with a lower risk of intubation/ICU admission or death among patients with mild-to-severe COVID-19. This study provides real-world evidence of adopting broad-spectrum oral therapeutics and shortening the gap between outbreak and effective response. It offers a new vision in our preparation for future pandemics.


Asunto(s)
COVID-19 , COVID-19/terapia , Humanos , Medicina Tradicional China , Puntaje de Propensión , Estudios Retrospectivos , SARS-CoV-2
10.
Bioorg Chem ; 129: 106148, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36244324

RESUMEN

Steroid sulfatase inhibitors block the local production of estrogenic steroids and are attractive agents for the treatment of estrogen-dependent cancers. Inspiration of coumarin-based inhibitors, we synthesized thirty-two 5-oxa-1,2,3,4-tetrahydro-2H-chromeno-(3,4-c)pyridin-8-yl sulfamates, focusing on the substitution derivatives on the adjacent phenyl ring and evaluated their abilities to block STS from human placenta and MCF-7 cells. SAR analysis revealed that the incorporation of chlorine at either meta and/or para position of the adjacent phenyl ring of the tricyclic skeleton enhanced STS inhibition. Di-substitutions at the adjacent phenyl ring were superior to mono and tri-substitutions. Further kinetic analysis of these compounds revealed that chloride-bearing compounds, such as 19m, 19v, and 19w, had KI of 0.02 to 0.11 nM and kinact/KI ratios of 8.8-17.5 nM-1min-1, a parameter indicated for the efficiency of irreversible inhibition. We also used the docking model to illustrate the difference in STS inhibitory potency of compounds. Finally, the safety and anti-cancer activity of selected compounds 19m, 19v, and 19w were also studied, showing the results of low cytotoxicity on NHDF cell line and being more potent than irosustat on ZR-75-1 cell, which was a hormone-dependent cancer cell line with high STS expression.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos , Placenta , Esteril-Sulfatasa , Ácidos Sulfónicos , Femenino , Humanos , Embarazo , Inhibidores Enzimáticos/farmacología , Cinética , Esteril-Sulfatasa/antagonistas & inhibidores , Relación Estructura-Actividad , Ácidos Sulfónicos/química , Ácidos Sulfónicos/farmacología , Placenta/enzimología , Células MCF-7
11.
Bioorg Chem ; 96: 103618, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32059152

RESUMEN

Steroid sulfatase (STS) is a sulfatase enzyme that catalyzes the conversion of sulfated steroid precursors to free steroid. The inhibition of STS could abate estrogenic steroids that stimulate the proliferation and development of breast cancer, and therefore STS is a potential target for adjuvant endocrine therapy. In this study, a series of 3-benzylaminocoumarin-7-O-sulfamate derivatives targeting STS were designed and synthesized. Structure-relationship activities (SAR) analysis revealed that attachment of a benzylamino group at the 3-position of coumarin improved inhibitory activity. Compound 3j was found to have the highest inhibition activity against human placenta isolated STS (IC50  0.13 µM) and MCF-7 cell lines (IC50 1.35 µM). Kinetic studies found compound 3j to be an irreversible inhibitor of STS, with KI and kinact value of 86.9 nM and 158.7 min-1, respectively.


Asunto(s)
Cumarinas/química , Cumarinas/farmacología , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Esteril-Sulfatasa/antagonistas & inhibidores , Aminación , Compuestos de Bencilo/síntesis química , Compuestos de Bencilo/química , Compuestos de Bencilo/farmacología , Cumarinas/síntesis química , Inhibidores Enzimáticos/síntesis química , Femenino , Humanos , Células MCF-7 , Placenta/enzimología , Embarazo , Esteril-Sulfatasa/metabolismo , Relación Estructura-Actividad , Ácidos Sulfónicos/síntesis química , Ácidos Sulfónicos/química , Ácidos Sulfónicos/farmacología
12.
Anim Biotechnol ; 30(4): 293-301, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30261812

RESUMEN

The human cluster of differentiation 19 (CD19) is highly expressed in most leukemia, rendering is a promising therapeutic target. In this study, we generated anti-CD19 single-chain variable fragments (scFv) from immunized chickens by phage display technology. After constructing a scFv antibody library with 2.5 × 108 compositional diversity for panning, one representative scFv clone S2 which can specifically recognize to the CD19 protein was isolated and characterized. The binding reactivity of the scFv S2 to the endogenous CD19 protein of the ARH-77 leukemia cancer cell was verified through flow cytometry and the binding affinity of scFv S2 is 6.9 × 10-8 M determined by the surface plasmon resonance system. Compared with the chicken germline, hyper mutation in the complementarity-determining regions (CDRs) suggested that scFv S2 could be generated through an antigen-driven humoral response. By molecular modeling, the possible CDR configurations of scFv S2 were constructed rationally. Furthermore, the characteristics of chicken antibodies of a protein database were investigated. The findings in this study contribute to antibody development and engineering because they reveal the geometric structures and properties of the CDRs in chicken antibodies.


Asunto(s)
Antígenos CD19/inmunología , Anticuerpos de Cadena Única/química , Animales , Línea Celular Tumoral , Técnicas de Visualización de Superficie Celular , Pollos/inmunología , Regiones Determinantes de Complementariedad/inmunología , Humanos , Modelos Moleculares , Anticuerpos de Cadena Única/sangre , Anticuerpos de Cadena Única/genética , Resonancia por Plasmón de Superficie
14.
Toxicol Appl Pharmacol ; 357: 88-105, 2018 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-30189238

RESUMEN

An aminopropyl carbazole compound, P7C3, has been shown to be a potent neurogenesis promoting agent; however, its fundamental signaling action has yet to be elucidated. A cerebral ischemic/reperfusional (CI/R) injury model in mice was implemented to elucidate the neuronal protective mechanism(s) of P7C3. Treating CI/R mice using P7C3 (50-100 µg/kg, i.v.) significantly improved tracking distance and walking behavior, and reduced brain damage. Specifically, P7C3 promoted the expression of neurogenesis-associated proteins, including doublecortin, beta tubulin III (ß-tub3), adam11 and adamts20, near the peri-infarct cortex, accompanied by glycogen synthase kinase 3 (GSK-3) inhibition and ß-catenin upregulation. The application of a specific inhibitor against glucagon-like peptide 1 receptor (GLP-1R), exendin(9-39), revealed that the beneficial effects of P7C3 involved triggering the activation of GLP-1R-associated PKA/Akt signaling. P7C3 elicited the GLP-1R-dependent intracellular cAMP increment and the insulin secretion in cellular models. Surface plasmon resonance assay of P7C3 showed a Kd value of 0.53 µM for GLP-1R binding, and the docking of P7C3 to the putative active site on GLP-1R was successfully predicted by molecular modeling. Our findings indicate that P7C3 promotes the expression of neurogenesis proteins by activation of the cAMP/PKA-dependent and Akt/GSK3-associated ß-catenin through positive allosteric stimulation of GLP-1R. Within the P7C3 class of neuroprotective molecules, this mechanism appears to be unique to the prototypical P7C3 molecule, as other active derivatives such as P7C2-A20 and P7C3-S243 they do not engage this same pathway and have been shown to work by nicotinamide phosphoribosyltransferase (NAMPT) stimulation.


Asunto(s)
Carbazoles/farmacología , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Neurogénesis/efectos de los fármacos , Daño por Reperfusión/tratamiento farmacológico , Animales , Barrera Hematoencefálica , Isquemia Encefálica , Carbazoles/química , Regulación de la Expresión Génica/efectos de los fármacos , Receptor del Péptido 1 Similar al Glucagón/genética , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Masculino , Ratones , Estructura Molecular , Resonancia por Plasmón de Superficie
15.
Planta Med ; 84(18): 1348-1354, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29986352

RESUMEN

Phytochemical investigation of ethanol extracts from two Taiwanese collections of Vernonia cinerea resulted in the isolation of eighteen hirsutinolide-type sesquiterpenoids, including seven new ones designated as vernolides E - K (1: -7: ). All structures were determined by a combination of detailed spectroscopic analyses (NMR and MS) and comparison with reported data. In an in vitro anti-inflammatory assay, compounds 3, 7, 9, 11: , and 14: exhibited strong inhibitory activities toward NO production by LPS-induced RAW264.7 macrophages, with IC50 values of 1.18, 0.85, 0.66, 0.71 and 0.45 µM, respectively, without affecting cellular viability at 40 µM. Preliminary structure-activity relationships indicate that the ester groups at C-8 and C-13 may enhance inhibition of NO production.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Óxido Nítrico/biosíntesis , Sesquiterpenos/química , Sesquiterpenos/farmacología , Vernonia/química , Animales , Antiinflamatorios no Esteroideos/química , Evaluación Preclínica de Medicamentos/métodos , Concentración 50 Inhibidora , Lipopolisacáridos/farmacología , Espectroscopía de Resonancia Magnética , Ratones , Estructura Molecular , Células RAW 264.7 , Espectrometría de Masa por Ionización de Electrospray , Relación Estructura-Actividad
16.
Org Biomol Chem ; 15(46): 9910-9922, 2017 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-29159346

RESUMEN

Peramivir is an effective anti-influenza drug in the clinical treatment of influenza, but its efficacy toward the H275Y mutant is reduced. The previously reported cocrystal structures of inhibitors in the mutant neuraminidase (NA) suggest that the hydrophobic side chain should be at the origin of reduced binding affinity. In contrast, zanamivir having a hydrophilic glycerol side chain still possesses high affinity toward the H275Y NA. We thus designed five peramivir analogues (5-9) carrying hydrophilic glycol or glycerol side chains, and evaluated their roles in anti-influenza activity, especially for the H275Y mutant. The synthetic sequence involves a key step of (3 + 2) cycloaddition reactions between alkenes and nitrile oxides to construct the scaffold of peramivir carrying the desired hydrophilic side chains and other appropriate functional groups. The molecular docking experiments reveal that the hydrophilic side chain can provide extra hydrogen bonding with the translocated Glu-276 residue in the H275Y NA active site. Thus, the H275Y mutant may be even more sensitive than wild-type virus toward the peramivir analogues bearing hydrophilic side chains. Notably, the peramivir analogue bearing a glycerol side chain inhibits the H275Y mutant with an IC50 value of 35 nM, which is better than the WSN virus by 9 fold.


Asunto(s)
Antivirales/farmacología , Ciclopentanos/farmacología , Inhibidores Enzimáticos/farmacología , Guanidinas/farmacología , Neuraminidasa/antagonistas & inhibidores , Orthomyxoviridae/efectos de los fármacos , Ácidos Carbocíclicos , Animales , Antivirales/síntesis química , Antivirales/química , Ciclopentanos/síntesis química , Ciclopentanos/química , Perros , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Guanidinas/síntesis química , Guanidinas/química , Interacciones Hidrofóbicas e Hidrofílicas , Células de Riñón Canino Madin Darby , Pruebas de Sensibilidad Microbiana , Modelos Moleculares , Estructura Molecular , Mutación , Neuraminidasa/metabolismo , Orthomyxoviridae/enzimología , Relación Estructura-Actividad
17.
J Biomed Sci ; 23: 27, 2016 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-26892079

RESUMEN

BACKGROUND: The accumulation of soluble oligomeric amyloid-ß peptide (oAß) proceeding the formation of senile plaques contributes to synaptic and memory deficits in Alzheimer's disease. Our previous studies have indentified scavenger receptor A (SR-A), especially SR-A type I (SR-AI), as prominent scavenger receptors on mediating oAß clearance by microglia while glycan moiety and scavenger receptor cysteine-rich (SRCR) domain may play the critical role. Macrophage receptor with collagenous structure (MARCO), another member of class A superfamily with a highly conserved SRCR domain, may also play the similar role on oAß internalization. However, the role of N-glycosylation and SRCR domain of SR-AI and MARCO on oAß internalization remains unclear. RESULT: We found that oAß internalization was diminished in the cells expressing SR-AI harboring mutations of dual N-glycosylation sites (i.e. N120Q-N143Q and N143Q-N184Q) while they were normally surface targeted. Normal oAß internalization was observed in 10 SR-AI-SRCR and 4 MARCO-SRCR surface targeted mutants. Alternatively, the SRCR mutants at ß-sheet and α-helix and on disulfide bone formation obstructed receptor's N-glycosylation and surface targeting. CONCLUSION: Our study reveals that N-glycan moiety is more critical than SRCR domain for SR-A-mediated oAß internalization.


Asunto(s)
Proteínas Portadoras/metabolismo , Receptores Inmunológicos/metabolismo , Sustitución de Aminoácidos , Péptidos beta-Amiloides , Animales , Células COS , Proteínas Portadoras/genética , Chlorocebus aethiops , Glicosilación , Células HEK293 , Humanos , Mutación Missense , Estructura Terciaria de Proteína , Transporte de Proteínas/genética , Receptores Inmunológicos/genética , Factores de Empalme Serina-Arginina
18.
Bioorg Med Chem Lett ; 26(2): 265-271, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26707392

RESUMEN

Malaria, caused by infections of the human malaria parasites Plasmodium falciparum, is a global infectious parasitic disease. Each year, about three million people died from malaria and the majority of whom are pregnant women and young children. Recently, a number of research attempt to reduce malaria parasite resistance and the toxicity of anti-malarial drugs. Nowadays, Plasmodium falciparum dihydroorotate dehydrogenase (PfDHODH) was validated as a potent drug target to inhibit malarial activity by blocking pyrimidine biosynthesis. In this study, we employed 3D-QSAR Pharmacophore Generation and Docking-Based Pharmacophore Development to build the pharmacophore by using the collected 67 effective inhibitors against PfDHODH. 3D-QSAR Pharmacophore model, Hypo1, shows the high correlation coefficient (0.935), the lowest RMS deviation (2.15), the predicting accuracy of successful rates to training set (89.4%) and test set compounds (72.4%), respectively, revealing favorable predictive ability and is a reliable for further study. Additionally, Docking-Based Pharmacophore model, DBP-All255, exhibits comparable predictive capability to that of Hypo1, while DBP-Top1 shows poor statistical significance. This study reveals pharmacophore features of Hypo1, built by 3D-QSAR Pharmacophore Generation, are well-complementary to the functional residues in the active site of PfDHODH and is of great reliable for database screening.


Asunto(s)
Antimaláricos/química , Derivados del Benceno/química , Compuestos Heterocíclicos/química , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Plasmodium falciparum/enzimología , Dihidroorotato Deshidrogenasa , Enlace de Hidrógeno , Ligandos , Simulación del Acoplamiento Molecular , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/química , Relación Estructura-Actividad Cuantitativa
19.
Mol Pharmacol ; 87(2): 218-30, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25403678

RESUMEN

Tyrosinase, a key copper-containing enzyme involved in melanin biosynthesis, is closely associated with hyperpigmentation disorders, cancer, and neurodegenerative diseases, and as such, it is an essential target in medicine and cosmetics. Known tyrosinase inhibitors possess adverse side effects, and there are no safety regulations; therefore, it is necessary to develop new inhibitors with fewer side effects and less toxicity. Peptides are exquisitely specific to their in vivo targets, with high potencies and relatively few off-target side effects. Thus, we systematically and comprehensively investigated the tyrosinase-inhibitory abilities of N- and C-terminal cysteine/tyrosine-containing tetrapeptides by constructing a phage-display random tetrapeptide library and conducting computational molecular docking studies on novel tyrosinase tetrapeptide inhibitors. We found that N-terminal cysteine-containing tetrapeptides exhibited the most potent tyrosinase-inhibitory abilities. The positional preference of cysteine residues at the N terminus in the tetrapeptides significantly contributed to their tyrosinase-inhibitory function. The sulfur atom in cysteine moieties of N- and C-terminal cysteine-containing tetrapeptides coordinated with copper ions, which then tightly blocked substrate-binding sites. N- and C-terminal tyrosine-containing tetrapeptides functioned as competitive inhibitors against mushroom tyrosinase by using the phenol ring of tyrosine to stack with the imidazole ring of His263, thus competing for the substrate-binding site. The N-terminal cysteine-containing tetrapeptide CRVI exhibited the strongest tyrosinase-inhibitory potency (with an IC50 of 2.7 ± 0.5 µM), which was superior to those of the known tyrosinase inhibitors (arbutin and kojic acid) and outperformed kojic acid-tripeptides, mimosine-FFY, and short-sequence oligopeptides at inhibiting mushroom tyrosinase.


Asunto(s)
Cisteína/metabolismo , Monofenol Monooxigenasa/metabolismo , Oligopéptidos/metabolismo , Biblioteca de Péptidos , Azufre/metabolismo , Agaricales/enzimología , Agaricales/genética , Secuencia de Bases , Cisteína/genética , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/metabolismo , Datos de Secuencia Molecular , Monofenol Monooxigenasa/antagonistas & inhibidores , Oligopéptidos/administración & dosificación , Oligopéptidos/genética , Unión Proteica/fisiología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
20.
Biochem Biophys Res Commun ; 460(2): 170-6, 2015 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-25769957

RESUMEN

Trimeresurus mucrosquamatus (TM) is one of majorities of snake envenomation with necrotic and hemorrhagic toxin in Taiwan. In this study, chickens were used as an alternative animal model for immunization with TM venom. Using phage display technology to process four rounds of panning, selected single chain variable fragments (scFv) could specifically recognize TM venom proteins, which were later identified as a group of homogeneous venom serine protease. The specific scFv antibodies showed various inhibitory effects on sheep RBC lysis induced by TM venom using an indirect hemolytic assay in vitro. In addition, the survival times of mice were extended to certain degrees when treated with these scFv antibodies individually or in a combination. To elucidate the inhibitory mechanism, we used molecular modeling to build up the serine protease structure to simulate the possible interactions with scFv antibodies. The results suggested that the CDR-loop of the scFv antibodies (3S10 or 4S1) might bind at the 99-loop of venom serine protease so as to affect substrate access due to the partial collapse of the subsite S2 and the partial movement of the subsite S4. It is hoped these chicken-derived antibodies could be applied to develop diagnostic and therapeutic agents against snakebites.


Asunto(s)
Venenos de Crotálidos/toxicidad , Inhibidores de Serina Proteinasa/farmacología , Anticuerpos de Cadena Única/farmacología , Animales , Anticuerpos Neutralizantes/inmunología , Formación de Anticuerpos , Western Blotting , Pollos , Venenos de Crotálidos/antagonistas & inhibidores , Venenos de Crotálidos/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Simulación del Acoplamiento Molecular , Trimeresurus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA