Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Int J Mol Sci ; 23(1)2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-35008983

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease with a multifactorial etiology. A multitarget treatment that modulates multifaceted biological functions might be more effective than a single-target approach. Here, the therapeutic efficacy of combination treatment using anti-Aß antibody NP106 and curcumin analog TML-6 versus monotherapy was investigated in an APP/PS1 mouse model of AD. Our data demonstrate that both combination treatment and monotherapy attenuated brain Aß and improved the nesting behavioral deficit to varying degrees. Importantly, the combination treatment group had the lowest Aß levels, and insoluble forms of Aß were reduced most effectively. The nesting performance of APP/PS1 mice receiving combination treatment was better than that of other APP/PS1 groups. Further findings indicate that enhanced microglial Aß phagocytosis and lower levels of proinflammatory cytokines were concurrent with the aforementioned effects of NP106 in combination with TML-6. Intriguingly, combination treatment also normalized the gut microbiota of APP/PS1 mice to levels resembling the wild-type control. Taken together, combination treatment outperformed NP106 or TML-6 monotherapy in ameliorating Aß pathology and the nesting behavioral deficit in APP/PS1 mice. The superior effect might result from a more potent modulation of microglial function, cerebral inflammation, and the gut microbiota. This innovative treatment paradigm confers a new avenue to develop more efficacious AD treatments.


Asunto(s)
Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/antagonistas & inhibidores , Precursor de Proteína beta-Amiloide/deficiencia , Anticuerpos Monoclonales/farmacología , Curcumina/farmacología , Presenilina-1/deficiencia , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/patología , Animales , Conducta Animal/efectos de los fármacos , Biomarcadores , Curcumina/análogos & derivados , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Inmunohistoquímica , Ratones , Ratones Noqueados , Microbiota/efectos de los fármacos , Microglía/efectos de los fármacos , Microglía/metabolismo , Terapia Molecular Dirigida , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/patología
2.
Int J Mol Sci ; 22(15)2021 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-34360989

RESUMEN

Increasing evidence suggests that elderly people with dementia are vulnerable to the development of severe coronavirus disease 2019 (COVID-19). In Alzheimer's disease (AD), the major form of dementia, ß-amyloid (Aß) levels in the blood are increased; however, the impact of elevated Aß levels on the progression of COVID-19 remains largely unknown. Here, our findings demonstrate that Aß1-42, but not Aß1-40, bound to various viral proteins with a preferentially high affinity for the spike protein S1 subunit (S1) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the viral receptor, angiotensin-converting enzyme 2 (ACE2). These bindings were mainly through the C-terminal residues of Aß1-42. Furthermore, Aß1-42 strengthened the binding of the S1 of SARS-CoV-2 to ACE2 and increased the viral entry and production of IL-6 in a SARS-CoV-2 pseudovirus infection model. Intriguingly, data from a surrogate mouse model with intravenous inoculation of Aß1-42 show that the clearance of Aß1-42 in the blood was dampened in the presence of the extracellular domain of the spike protein trimers of SARS-CoV-2, whose effects can be prevented by a novel anti-Aß antibody. In conclusion, these findings suggest that the binding of Aß1-42 to the S1 of SARS-CoV-2 and ACE2 may have a negative impact on the course and severity of SARS-CoV-2 infection. Further investigations are warranted to elucidate the underlying mechanisms and examine whether reducing the level of Aß1-42 in the blood is beneficial to the fight against COVID-19 and AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Fragmentos de Péptidos/metabolismo , SARS-CoV-2/enzimología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células A549 , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Animales , COVID-19/complicaciones , COVID-19/metabolismo , Chlorocebus aethiops , Humanos , Interleucina-6/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Fragmentos de Péptidos/química , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Células Vero , Internalización del Virus
3.
Artículo en Inglés | MEDLINE | ID: mdl-33139286

RESUMEN

We discovered that neuropilin 1 (NRP1) is a new receptor candidate to mediate enterovirus A71 (EVA71) into cells. In the engineered form as a decoy receptor, NRP1 was able to recognize and neutralize EVA71 but not enterovirus D68 or coxsackievirus B3 (CVB3). NRP1 recognizes EVA71 through a novel domain on the VP3 capsid protein. The principle in the design, engineering, and refinement of the NRP1-based decoy receptor described in this study represents a general and well-suited antiviral strategy.


Asunto(s)
Enterovirus Humano A , Infecciones por Enterovirus , Enterovirus , Enterovirus Humano A/genética , Humanos , Neuropilina-1/genética , Receptores Virales/genética
4.
Artículo en Inglés | MEDLINE | ID: mdl-32152074

RESUMEN

In the past few decades, enterovirus A71 (EVA71) has caused devastating outbreaks in the Asia-Pacific region, resulting in serious sequelae in infected young children. No preventive or therapeutic interventions are currently available for curing EVA71 infection, highlighting a great unmet medical need for this disease. Here, we showed that one novel single-domain antibody (sdAb), F1, isolated from an immunized llama, could alleviate EVA71 infection both in vitro and in vivo We also confirmed that the sdAb clone F1 recognizes EVA71 through a novel conformational epitope comprising the highly conserved region of VP3 capsid protein by using competitive-binding and overlapping-peptide enzyme-linked immunosorbent assays (ELISAs). Because of the virion's icosahedral structure, we reasoned that adjacent epitopes must be clustered within molecular ranges that may be simultaneously bound by an engineered antibody with multiple valency. Therefore, two single-domain binding modules (F1) were fused to generate an sdAb-in-tandem design so that the capture of viral antigens could be further increased by valency effects. We showed that the tetravalent construct F1×F1-hFc, containing two sdAb-in-tandem on a fragment crystallizable (Fc) scaffold, exhibits more potent neutralization activity against EVA71 than does the bivalent sdAb F1-hFc by at least 5.8-fold. We also demonstrated that, using a human scavenger receptor class B member 2 (hSCARB2) transgenic mouse model, a half dose of the F1×F1-hFc provided better protection against EVA71 infection than did the F1-hFc. Thus, our study furnishes important insights into multivalent sdAb engineering against viral infection and provides a novel strategic deployment approach for preparedness of emerging infectious diseases such as EVA71.


Asunto(s)
Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/inmunología , Enterovirus Humano A/inmunología , Infecciones por Enterovirus/terapia , Anticuerpos de Dominio Único/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/farmacología , Antígenos Virales/inmunología , Camélidos del Nuevo Mundo , Línea Celular Tumoral , Infecciones por Enterovirus/inmunología , Infecciones por Enterovirus/prevención & control , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Ratones , Ratones Transgénicos , Anticuerpos de Dominio Único/farmacología
5.
Artículo en Inglés | MEDLINE | ID: mdl-32669265

RESUMEN

The coronavirus (CoV) disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome CoV-2 (SARS-CoV-2) is a health threat worldwide. Viral main protease (Mpro, also called 3C-like protease [3CLpro]) is a therapeutic target for drug discovery. Herein, we report that GC376, a broad-spectrum inhibitor targeting Mpro in the picornavirus-like supercluster, is a potent inhibitor for the Mpro encoded by SARS-CoV-2, with a half-maximum inhibitory concentration (IC50) of 26.4 ± 1.1 nM. In this study, we also show that GC376 inhibits SARS-CoV-2 replication with a half-maximum effective concentration (EC50) of 0.91 ± 0.03 µM. Only a small portion of SARS-CoV-2 Mpro was covalently modified in the excess of GC376 as evaluated by mass spectrometry analysis, indicating that improved inhibitors are needed. Subsequently, molecular docking analysis revealed that the recognition and binding groups of GC376 within the active site of SARS-CoV-2 Mpro provide important new information for the optimization of GC376. Given that sufficient safety and efficacy data are available for GC376 as an investigational veterinary drug, expedited development of GC376, or its optimized analogues, for treatment of SARS-CoV-2 infection in human is recommended.


Asunto(s)
Antivirales/química , Betacoronavirus/efectos de los fármacos , Cisteína Endopeptidasas/química , Inhibidores de Proteasas/química , Pirrolidinas/química , Proteínas no Estructurales Virales/química , Secuencias de Aminoácidos , Animales , Antivirales/farmacología , Betacoronavirus/patogenicidad , Dominio Catalítico , Chlorocebus aethiops , Proteasas 3C de Coronavirus , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Expresión Génica , Simulación del Acoplamiento Molecular , Inhibidores de Proteasas/farmacología , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Pirrolidinas/farmacología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , SARS-CoV-2 , Ácidos Sulfónicos , Termodinámica , Células Vero , Proteínas no Estructurales Virales/antagonistas & inhibidores , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/efectos de los fármacos
6.
Biomed Pharmacother ; 173: 116404, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38471275

RESUMEN

High-fat diet (HFD)-induced fatty liver disease is a deteriorating risk factor for Alzheimer's disease (AD). Mitigating fatty liver disease has been shown to attenuate AD-like pathology in animal models. However, it remains unclear whether enhancing Aß clearance through immunotherapy would in turn attenuate HFD-induced fatty liver or whether its efficacy would be compromised by long-term exposure to HFD. Here, the therapeutic potentials of an anti-Aß antibody, NP106, was investigated in APP/PS1 mice by HFD feeding for 44 weeks. The data demonstrate that NP106 treatment effectively reduced Aß burden and pro-inflammatory cytokines in HFD-fed APP/PS1 mice and ameliorated HFD-aggravated cognitive impairments during the final 18 weeks of the study. The rejuvenating characteristics of microglia were evident in APP/PS1 mice with NP106 treatment, namely enhanced microglial Aß phagocytosis and attenuated microglial lipid accumulation, which may explain the benefits of NP106. Surprisingly, NP106 also reduced HFD-induced hyperglycemia, fatty liver, liver fibrosis, and hepatic lipids, concomitant with modifications in the expressions of genes involved in hepatic lipogenesis and fatty acid oxidation. The data further reveal that brain Aß burden and behavioral deficits were positively correlated with the severity of fatty liver disease and fasting serum glucose levels. In conclusion, our study shows for the first time that anti-Aß immunotherapy using NP106, which alleviates AD-like disorders in APP/PS1 mice, ameliorates fatty liver disease. Minimizing AD-related pathology and symptoms may reduce the vicious interplay between central AD and peripheral fatty liver disease, thereby highlighting the importance of developing AD therapies from a systemic disease perspective.


Asunto(s)
Enfermedad de Alzheimer , Hígado Graso , Hepatopatías , Ratones , Animales , Precursor de Proteína beta-Amiloide/metabolismo , Ratones Transgénicos , Dieta Alta en Grasa/efectos adversos , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Hepatopatías/metabolismo , Hígado Graso/metabolismo , Modelos Animales de Enfermedad , Péptidos beta-Amiloides/metabolismo
7.
Biotechnol Bioeng ; 108(12): 2932-40, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21755496

RESUMEN

Cytochrome P450 1A2 (CYP1A2) is an important member of cytochrome P450 involved in drug metabolism. In this study, a cell line, Huh7-1A2-I-E, with high expression level of CYP1A2 is established based on Huh7 cells. To achieve this, we constructed a recombinant lentiviral vector, pLenti-1A2-I-E, containing a single promoter encoding CYP1A2 followed by an internal ribosome entry site (IRES) to permit the translation of enhanced green fluorescence protein (EGFP). Such a design has greatly facilitated the selection of stable cell lines because the translations of CYP1A2 and EGFP proteins would be based on a single bi-cistronic mRNA. The Huh7-1A2-I-E cells were evaluated as a cell-based model for identification of CYP1A2 inhibitors and for studies of cytotoxicity resulted from CYP-mediated drug metabolism. Treatment of Huh7-1A2-I-E cells and the Huh7-E control cells with aflatoxin B1 showed that cells with CYP1A2 expression are much more sensitive to aflatoxin B1 and the cellular toxicity of aflatoxin B1 in Huh7-1A2-I-E cells could be prevented by furafylline, a CYP1A2 inhibitor. A collection of approximately 200 drugs were screened using this system and results indicate that for most drugs the metabolism by CYP1A2 is unlikely to have made a major contribution to the in vitro cytotoxicity except for thimerosal and evoxine. Several previously unidentified CYP1A2 inhibitors such as evoxine and berberine were also identified in this study.


Asunto(s)
Inhibidores del Citocromo P-450 CYP1A2 , Citocromo P-450 CYP1A2/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos/farmacología , Berberina/metabolismo , Berberina/farmacología , Línea Celular , Citocromo P-450 CYP1A2/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Inhibidores Enzimáticos/metabolismo , Humanos , Proteínas Recombinantes/antagonistas & inhibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Teofilina/análogos & derivados , Teofilina/metabolismo , Teofilina/farmacología
8.
Sci Rep ; 10(1): 8929, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32488021

RESUMEN

Zika virus (ZIKV) of the flaviviridae family, is the cause of emerging infections characterized by fever, Guillain-Barré syndrome (GBS) in adults and microcephaly in newborns. There exists an urgent unmet clinical need for anti-ZIKV drugs for the treatment of infected individuals. In the current work, we aimed at the promising virus drug target, ZIKV NS3 protease and constructed a Pharmacophore Anchor (PA) model for the active site. The PA model reveals a total of 12 anchors (E, H, V) mapped across the active site subpockets. We further identified five of these anchors to be critical core anchors (CEH1, CH3, CH7, CV1, CV3) conserved across flaviviral proteases. The ZIKV protease PA model was then applied in anchor-enhanced virtual screening yielding 14 potential antiviral candidates, which were tested by in vitro assays. We discovered FDA drugs Asunaprevir and Simeprevir to have potent anti-ZIKV activities with EC50 values 4.7 µM and 0.4 µM, inhibiting the viral protease with IC50 values 6.0 µM and 2.6 µM respectively. Additionally, the PA model anchors aided in the exploration of inhibitor binding mechanisms. In conclusion, our PA model serves as a promising guide map for ZIKV protease targeted drug discovery and the identified 'previr' FDA drugs are promising for anti-ZIKV treatments.


Asunto(s)
Antivirales/farmacología , Descubrimiento de Drogas/métodos , Serina Endopeptidasas/efectos de los fármacos , Proteínas Virales/efectos de los fármacos , Virus Zika/efectos de los fármacos , Dominio Catalítico/efectos de los fármacos , Modelos Químicos , Simulación del Acoplamiento Molecular , Alineación de Secuencia , Virus Zika/enzimología , Virus Zika/genética
9.
Antimicrob Agents Chemother ; 53(7): 2740-7, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19414569

RESUMEN

Enterovirus 71 (EV71) has emerged as an important virulent neurotropic enterovirus in young children. DTriP-22 (4{4-[(2-bromo-phenyl)-(3-methyl-thiophen-2-yl)-methyl]-piperazin-1-yl}-1-pheny-1H-pyrazolo[3,4-d]pyrimidine) was found to be a novel and potent inhibitor of EV71. The molecular target of this compound was identified by analyzing DTriP-22-resistant viruses. A substitution of lysine for Arg163 in EV71 3D polymerase rendered the virus drug resistant. DTriP-22 exhibited the ability to inhibit viral replication by reducing viral RNA accumulation. The compound suppressed the accumulated levels of both positive- and negative-stranded viral RNA during virus infection. An in vitro polymerase assay indicated that DTriP-22 inhibited the poly(U) elongation activity, but not the VPg uridylylation activity, of EV71 polymerase. These findings demonstrate that the nonnucleoside analogue DTriP-22 acts as a novel inhibitor of EV71 polymerase. DTriP-22 also exhibited a broad spectrum of antiviral activity against other picornaviruses, which highlights its potential in the development of antiviral agents.


Asunto(s)
Antivirales/farmacología , Enterovirus/efectos de los fármacos , Enterovirus/enzimología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Perros , Enterovirus/genética , Infecciones por Enterovirus/tratamiento farmacológico , Células HeLa , Humanos , ARN Polimerasa Dependiente del ARN/genética , Proteínas Recombinantes/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Vero
10.
Sci Rep ; 9(1): 2405, 2019 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-30787373

RESUMEN

Morphine is a unique opioid analgesic that activates the mu-opioid receptor (MOR) without efficiently promoting its endocytosis that may underlie side effects. Our objective was to discover a novel enhancer of ligand-induced MOR endocytosis and determine its effects on analgesia, tolerance and dependence. We used high-throughput screening to identify convallatoxin as an enhancer of ligand-induced MOR endocytosis with high potency and efficacy. Treatment of cells with convallatoxin enhanced morphine-induced MOR endocytosis through an adaptor protein 2 (AP2)/clathrin-dependent mechanism, attenuated morphine-induced phosphorylation of MOR, and diminished desensitization of membrane hyperpolarization. Furthermore, co-treatment with chronic convallatoxin reduced morphine tolerance in animal models of acute thermal pain and chronic inflammatory pain. Acute convallatoxin administration reversed morphine tolerance and dependence in morphine-tolerant mice. These findings suggest convallatoxin are potentially therapeutic for morphine side effects and open a new avenue to study MOR trafficking.


Asunto(s)
Analgésicos/farmacología , Morfina/farmacología , Receptores Opioides mu/genética , Estrofantinas/farmacología , Analgesia/métodos , Analgésicos/química , Animales , Modelos Animales de Enfermedad , Endocitosis/efectos de los fármacos , Humanos , Ligandos , Ratones , Receptores Opioides mu/efectos de los fármacos
11.
Phytother Res ; 22(5): 605-13, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18398904

RESUMEN

This study isolated agonists of peroxisome proliferator activated receptors (PPARs) from the green algae Chlorella sorokiniana, using a bioassay-guided purification strategy. PPARs are widely recognized as the molecular drug targets for many diseases including hyperglycemia, diabetes, obesity and cancer. Two independent bioassays were developed. The first is the scintillation proximity assay, a ligand binding assay. The other is the cell-based transcriptional activation assay which uses the Dual-Luciferase reporter system as the reporter gene under the control of the PPAR response element. Using these two assays, a PPARgamma-active fraction, CE 3-3, was obtained from C. sorokiniana extracts, which was also able to activate PPARalphamediated gene expression. To elucidate the active ingredients in the CE 3-3 fraction, GC-MS analysis was employed. The results showed that the CE 3-3 fraction consisted of at least ten fatty acids (FAs). The bioactivities of several of the individual FAs were evaluated for their PPARgamma activity and the results showed that linolenic acid and linoleic acid were the most potent FAs tested. Our studies indicate that Chlorella sorokiniana could have potential health benefits through the dual activation of PPARalpha/gamma via its unique FA constituents.


Asunto(s)
Bioensayo/métodos , Chlorella/química , PPAR alfa/agonistas , PPAR gamma/agonistas , Línea Celular Tumoral , Ácidos Grasos/aislamiento & purificación , Ácidos Grasos/farmacología , Cromatografía de Gases y Espectrometría de Masas , Humanos , Ácido Linoleico/aislamiento & purificación , Ácido Linoleico/farmacología , PPAR alfa/genética , PPAR gamma/genética , Activación Transcripcional/efectos de los fármacos , Ácido alfa-Linolénico/aislamiento & purificación , Ácido alfa-Linolénico/farmacología
12.
J Med Chem ; 49(21): 6421-4, 2006 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-17034149

RESUMEN

Type 2 diabetes has rapidly reached an epidemic proportion becoming a major threat to global public health. PPAR agonists have emerged as a leading class of oral antidiabetic drugs. We report a structure biology analysis of novel indole-based PPAR agonists to explain the structure-activity relationships and present a critical analysis of reasons for change in selectivity with change in the orientation of the same scaffolds. The results would be helpful in designing novel PPAR agonists.


Asunto(s)
Indoles/química , Receptores Activados del Proliferador del Peroxisoma/agonistas , Receptores Activados del Proliferador del Peroxisoma/química , Acetatos/química , Alquilación , Sitios de Unión , Cristalografía por Rayos X , Humanos , Técnicas In Vitro , Indoles/síntesis química , Indoles/farmacología , Ligandos , Modelos Moleculares , Estructura Molecular , Naftalenos/síntesis química , Naftalenos/química , Naftalenos/farmacología , Receptores Activados del Proliferador del Peroxisoma/genética , Propionatos/química , Relación Estructura-Actividad , Termodinámica , Activación Transcripcional/efectos de los fármacos
13.
Theranostics ; 6(1): 118-30, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26722378

RESUMEN

The overexpression of HER2/neu and EGFR receptors plays important roles in tumorigenesis and tumor progression. Targeting these two receptors simultaneously can have a more widespread application in early diagnosis of cancers. In this study, a new multifunctional nanoparticles (MnMEIO-CyTE777-(Bis)-mPEG NPs) comprising a manganese-doped iron oxide nanoparticle core (MnMEIO), a silane-amino functionalized poly(ethylene glycol) copolymer shell, a near infrared fluorescence dye (CyTE777), and a covalently conjugated anti-HER2/neu and anti-EGFR receptors bispecific antibody (Bis) were successfully developed. In vitro T2-weighted MR imaging studies in SKBR-3 and A431 tumor cells incubated with MnMEIO-CyTE777-(Bis)-mPEG NPs showed - 94.8 ± 3.8 and - 84.1 ± 2.8% negative contrast enhancement, respectively. Pharmacokinetics study showed that MnMEIO-CyTE777-(Bis)-mPEG NPs were eliminated from serum with the half-life of 21.3 mins. In vivo MR imaging showed that MnMEIO-CyTE777-(Bis)-mPEG NPs could specifically and effectively target to HER2/neu- and EGFR-expressing tumors in mice; the relative contrast enhancements were 11.8 (at 2 hrs post-injection) and 61.5 (at 24 hrs post-injection) fold higher in SKBR-3 tumors as compared to Colo-205 tumors. T2-weighted MR and optical imaging studies revealed that the new contrast agent (MnMEIO-CyTE777-(Bis)-mPEG NPs) could specifically and effectively target to HER2/neu- and/or EGFR-expressing tumors. Our results demonstrate that MnMEIO-CyTE777-(Bis)-mPEG NPs are able to recognize the tumors expressing both HER2/neu and/or EGFR, and may provide a novel molecular imaging tool for early diagnosis of cancers expressing HER2/neu and/or EGFR.


Asunto(s)
Anticuerpos Biespecíficos/farmacocinética , Receptores ErbB/análisis , Compuestos Férricos/farmacocinética , Imagen por Resonancia Magnética/métodos , Manganeso/farmacocinética , Neoplasias/diagnóstico , Receptor ErbB-2/análisis , Animales , Magnetismo , Ratones , Neoplasias/patología , Coloración y Etiquetado/métodos
14.
Mol Cancer Ther ; 15(10): 2323-2333, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27512117

RESUMEN

Acute myelogenous leukemia (AML) carrying t(8;21)(q22;q22) or inv(16)/t(16;16)(p13;q22) is classified as core binding factor (CBF)-AML and accounts for approximately 15% of AML. c-KIT mutation can be detected in 17%∼46% of CBF-AML and is associated with poor prognosis. c-KIT mutation is a crucial hit and cooperates with AML1-ETO resulting from t(8;21)(q22;q22) to cause overt AML. Tyrosine kinase inhibitors (TKI) targeting c-KIT, such as imatinib, has been used successfully to treat c-KIT driven gastrointestinal stromal tumors. However, the effect of TKI on c-KIT-driven leukemia, including CBF-AML and systemic mastocytosis (SM), has not been satisfactory. BPR1J373 is a 5-phenylthiazol-2-ylamine-pyriminide derivative targeting multiple tyrosine kinases. It was shown to inhibit cell proliferation and induce apoptosis in AML cells with constitutively activated c-KIT via inhibiting c-KIT phosphorylation and its downstream signals. The compound induced apoptosis by the mitochondrial intrinsic pathway through upregulation of proapoptotic proteins Bax and Bak and caspase 8 and 9 activation in c-KIT mutant Kasumi-1 cells. Furthermore, it induced cell-cycle arrest via targeting aurora kinase B in c-KIT wild-type KG-1 cells. The antitumor response of BPR1J373 was also shown in subcutaneously grafted SCID mice. BPR1J373 was shown to effectively suppress c-KIT phosphorylation of D816V mutation by treating c-KIT-null COS-1 cells transfected with c-KIT D816V mutant plasmid. In conclusion, BPR1J373 inhibits cell proliferation of c-KIT-driven AML cells via induction of apoptosis and cell-cycle arrest. It is also effective for multiple drug-resistant c-KIT D816V mutation. BPR1J373 deserves further development for clinical use in c-KIT-driven myeloid leukemia. Mol Cancer Ther; 15(10); 2323-33. ©2016 AACR.


Asunto(s)
Leucemia Mieloide Aguda/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación , Fosforilación , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal/efectos de los fármacos , Activación Transcripcional , Translocación Genética , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Antiviral Res ; 112: 18-25, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25448086

RESUMEN

Enterovirus 71 (EV71) infections can cause hand, foot, and mouth disease with severe neurological complications. Because no clinical drug is available for treating EV71 infections, developing an efficient antiviral medication against EV71 infection is crucial. This study indicated that 6-bromo-2-[1-(2,5-dimethylphenyl)-5-methyl-1H-pyrazol-4-yl] quinoline-4-carboxylic acid (BPR-3P0128) exhibits excellent antiviral activity against EV71 (EC50 = 0.0029 µM). BPR-3P0128 inhibits viral replication during the early post infection stage, targets EV71 RNA-dependent RNA polymerase and VPg uridylylation, and also reduces viral RNA accumulation levels and inhibits viral replication of EV71.


Asunto(s)
Antivirales/farmacología , Enterovirus Humano A/efectos de los fármacos , Enterovirus Humano A/enzimología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirazoles/farmacología , Quinolinas/farmacología , ARN Polimerasa Dependiente del ARN/antagonistas & inhibidores , Replicación Viral/efectos de los fármacos , Línea Celular , Efecto Citopatogénico Viral , Enterovirus Humano A/fisiología , Humanos , Concentración 50 Inhibidora , Ensayo de Placa Viral , Proteínas Virales/metabolismo
16.
Int J Biol Sci ; 9(4): 403-11, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23678290

RESUMEN

Small synthetic compounds have been implicated in treatment of human cancers. We have synthesized a small compound, BPR1K0609S1 (hereafter, BP), which inhibits Aurora-A kinase. In the present study, we studied the mechanism of BP suppression of tumorigenesis induced by Aurora-A. Given our previous results that inactivation of p53 accelerates MMTV-Aurora-A-mediated tumorigenesis in vivo, we studied the roles of p53 pathway using the isogenic human colon carcinoma cell lines of HCT116, in which p53, Puma, Bax, p21 or Chk2 is deleted. When these isogenic cell lines are treated with BP for 48 h, accumulation of G2M phase and aneuploidy are commonly observed, and HCT116 p21(-) cells show increase in apoptosis. In xenograft assay, s.c. injection of BP efficiently inhibits tumorigenesis of HCT116 deficient for Chk2 or p21. Re-transplantation of BP-resistant tumors indicates that these resistant cells do not acquire advanced tumor growth. Significantly, 5-FU (5-fluorouracil) treatment further induces apoptosis of BP-resistant HCT116 deficient for Chk2 or Puma. These results demonstrate that p21 deficiency enhances BP-mediated suppression of tumor growth, and that BP and 5-FU can collaborate for tumor regression.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Fluorouracilo/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Aurora Quinasa A , Aurora Quinasas , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores Enzimáticos/uso terapéutico , Femenino , Fluorouracilo/uso terapéutico , Células HCT116 , Humanos , Ratones , Ratones Desnudos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores
17.
J Med Chem ; 56(10): 3889-903, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23611691

RESUMEN

The Asp-Phe-Gly (DFG) motif plays an important role in the regulation of kinase activity. Structure-based drug design was performed to design compounds able to interact with the DFG motif; epidermal growth factor receptor (EGFR) was selected as an example. Structural insights obtained from the EGFR/2a complex suggested that an extension from the meta-position on the phenyl group (ring-5) would improve interactions with the DFG motif. Indeed, introduction of an N,N-dimethylamino tail resulted in 4b, which showed almost 50-fold improvement in inhibition compared to 2a. Structural studies confirmed this N,N-dimethylamino tail moved toward the DFG motif to form a salt bridge with the side chain of Asp831. That the interactions with the DFG motif greatly contribute to the potency of 4b is strongly evidenced by synthesizing and testing compounds 2a, 3g, and 4f: when the charge interactions are absent, the inhibitory activity decreased significantly.


Asunto(s)
Receptores ErbB/antagonistas & inhibidores , Oligopéptidos/síntesis química , Oligopéptidos/farmacología , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Cristalografía por Rayos X , Diseño de Fármacos , Receptores ErbB/genética , Humanos , Modelos Moleculares , Mutación , Unión Proteica , Conformación Proteica , Inhibidores de Proteínas Quinasas/química , Reacción en Cadena en Tiempo Real de la Polimerasa , Relación Estructura-Actividad
18.
PLoS One ; 6(8): e23485, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21887256

RESUMEN

BACKGROUND: Over-expression of Aurora kinases promotes the tumorigenesis of cells. The aim of this study was to determine the preclinical profile of a novel pan-Aurora kinase inhibitor, BPR1K653, as a candidate for anti-cancer therapy. Since expression of the drug efflux pump, MDR1, reduces the effectiveness of various chemotherapeutic compounds in human cancers, this study also aimed to determine whether the potency of BPR1K653 could be affected by the expression of MDR1 in cancer cells. PRINCIPAL FINDINGS: BPR1K653 specifically inhibited the activity of Aurora-A and Aurora-B kinase at low nano-molar concentrations in vitro. Anti-proliferative activity of BPR1K653 was evaluated in various human cancer cell lines. Results of the clonogenic assay showed that BPR1K653 was potent in targeting a variety of cancer cell lines regardless of the tissue origin, p53 status, or expression of MDR1. At the cellular level, BPR1K653 induced endo-replication and subsequent apoptosis in both MDR1-negative and MDR1-positive cancer cells. Importantly, it showed potent activity against the growth of xenograft tumors of the human cervical carcinoma KB and KB-derived MDR1-positive KB-VIN10 cells in nude mice. Finally, BPR1K653 also exhibited favorable pharmacokinetic properties in rats. CONCLUSIONS AND SIGNIFICANCE: BPR1K653 is a novel potent anti-cancer compound, and its potency is not affected by the expression of the multiple drug resistant protein, MDR1, in cancer cells. Therefore, BPR1K653 is a promising anti-cancer compound that has potential for the management of various malignancies, particularly for patients with MDR1-related drug resistance after prolonged chemotherapeutic treatments.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacología , Compuestos Bicíclicos Heterocíclicos con Puentes/farmacología , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Aurora Quinasa A , Aurora Quinasa B , Aurora Quinasas , Compuestos Bicíclicos Heterocíclicos con Puentes/química , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Ciclina B1/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Histonas/metabolismo , Humanos , Ratones , Compuestos de Fenilurea/química , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Pirimidinas/química , Ratas , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Biochemistry ; 44(30): 10349-59, 2005 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-16042412

RESUMEN

Viral proteases are essential for pathogenesis and virulence of severe acute respiratory syndrome coronavirus (SARS-CoV). Little information is available on SARS-CoV papain-like protease 2 (PLP2), and development of inhibitors against PLP2 is attractive for antiviral therapy. Here, we report the characterization of SARS-CoV PLP2 (from residues 1414 to 1858) purified from baculovirus-infected insect cells. We demonstrate that SARS-CoV PLP2 by itself differentially cleaves between the amino acids Gly180 and Ala181, Gly818 and Ala819, and Gly2740 and Lys2741 of the viral polypeptide pp1a, as determined by reversed-phase high-performance liquid chromatography analysis coupled with mass spectrometry. This protease is especially selective for the P1, P4, and P6 sites of the substrate. The study demonstrates, for the first time among coronaviral PLPs, that the reaction mechanism of SARS-CoV PLP2 is characteristic of papain and compatible with the involvement of the catalytic dyad (Cys)-S(-)/(His)-Im(+)H ion pair. With a fluorogenic inhibitor-screening platform, we show that zinc ion and its conjugates potently inhibit the enzymatic activity of SARS-CoV PLP2. In addition, we provided evidence for evolutionary reclassification of SARS-CoV. The results provide important insights into the biochemical properties of the coronaviral PLP family and a promising therapeutic way to fight SARS-CoV.


Asunto(s)
Inhibidores de Cisteína Proteinasa/farmacología , Papaína/genética , Papaína/aislamiento & purificación , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/enzimología , Proteínas Virales/genética , Proteínas Virales/aislamiento & purificación , Secuencia de Aminoácidos , Catálisis , Dominio Catalítico , Proteasas Similares a la Papaína de Coronavirus , Coronavirus Bovino/enzimología , Inhibidores de Cisteína Proteinasa/síntesis química , Hidrólisis , Concentración 50 Inhibidora , Cinética , Datos de Secuencia Molecular , Virus de la Hepatitis Murina/enzimología , Papaína/antagonistas & inhibidores , Papaína/biosíntesis , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/patogenicidad , Especificidad por Sustrato , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/biosíntesis , Zinc/farmacología
20.
Protein Expr Purif ; 35(1): 142-6, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15039077

RESUMEN

DPP8 is a new member of the prolyl dipeptidases, many of which have important biological functions in vivo. DPP8 catalyzes the cleavage at the carboxyl side of the proline residue at the penultimate position. To study its structure and biochemical properties, we have overexpressed the human DPP8 protein in baculovirus infected Sf9 cells. The protein is soluble and can be purified to homogeneity. Using the chromogenic H-Gly-Pro-pNA as the substrate, a kinetic study shows that purified DPP8 is active and has a similar kcat value as that of DPP-IV, a prolyl dipeptidase that is a drug target for type II diabetes. The kinetic constants of DPP8 are also determined for other chromogenic substrates, and the results indicate that DPP8 has substrate preference at both the P1 and P2 sites. The expression system provides means of better understanding the structure, catalytic mechanism, and biological function of DPP8 protein.


Asunto(s)
Dipeptidasas/aislamiento & purificación , Dipeptidasas/metabolismo , Animales , Baculoviridae/genética , Baculoviridae/metabolismo , Línea Celular , Dipeptidasas/genética , Humanos , Insectos , Péptidos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA