Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Cytokine ; 148: 155710, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34564023

RESUMEN

The pathogenicity of HCC could be enhanced by TNF-α and NFκB, which are crucial parts of the inflammatory pathway inside the HCC microenvironment. Therefore, we aimed to discover the therapeutic effects of QNZ, an inhibitor of both TNF-α and NFκB, in an experimental model of HCC in rats. HCC was experimentally induced in rats by thioacetamide, and some of the rats were treated with QNZ. The expression levels of nuclear factor (NF)κB, tumor necrosis factor (TNF)-α, apoptosis signal regulating kinase (ASK)-1, ß-catenin, glycogen synthase kinase (GSK)-3 and TNF receptor-associated factor (TRAF) were examined in hepatic samples. In addition, hepatic tissues were stained with hematoxylin/eosin and anti-TNF-α antibodies. QNZ blocked HCC-induced expression of both NFκB and TNF-α. It significantly reduced both α-fetoprotein and the average number of nodules and increased the survival rate of the HCC rats. Moreover, hematoxylin and eosin liver sections from the HCC rats showed vacuolated cytoplasm and necrotic nodules. All of these effects were alleviated by QNZ treatment. Finally, treating HCC rats with QNZ resulted in a reduction in the expression of TRAF, ASK-1 and ß-catenin, as well as increased expression of GSK-3. In conclusion, inhibition of the inflammatory pathway in HCC with QNZ produced therapeutic effects, as indicated by an increased survival rate, reduced serum α-fetoprotein levels, decreased liver nodules and improved the hepatocyte structure. In addition, QNZ significantly reduced the expression of TRAF, ASK-1 and ß-catenin that were associated with increased expression of GSK-3.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Éteres Fenílicos/uso terapéutico , Quinazolinas/uso terapéutico , Animales , Carcinoma Hepatocelular/complicaciones , Carcinoma Hepatocelular/patología , Modelos Animales de Enfermedad , Glucógeno Sintasa Quinasa 3/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/patología , Inflamación/complicaciones , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/complicaciones , Neoplasias Hepáticas/patología , MAP Quinasa Quinasa Quinasa 5/metabolismo , FN-kappa B/metabolismo , Especificidad de Órganos/efectos de los fármacos , Éteres Fenílicos/farmacología , Quinazolinas/farmacología , Ratas , Ratas Sprague-Dawley , Análisis de Supervivencia , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , alfa-Fetoproteínas/metabolismo , beta Catenina/metabolismo
2.
Biochem Pharmacol ; 188: 114522, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33741334

RESUMEN

Programmed death protein 1 (PD-1)/programmed death protein ligand 1 (PD-L1) pathway is one of the most actively pursued targets in cancer immunotherapy. In a continuation of our research interest in this pathway, we synthesized and evaluated the pharmacological activities of a series of resorcinol biphenyl ether analogs as small molecule PD-1/PD-L1 inhibitors for cancer treatment. Among the 27 newly synthesized compounds, CH1 was found to have the highest inhibitory effect against PD-1/PDL-1 with an IC50 value of 56.58 nM in the HTRF (homogenous time-resolved fluorescence) assay. In addition, CH1 dose-dependently promoted HepG2 cell death in a co-culture model of HepG2/hPD-L1 and Jurkat T cells. Furthermore, molecular modeling study indicated that CH1 binds with high affinity to the binding interface of PD-L1. Moreover, CH1 effectively inhibited tumor growth (TGI of 76.4% at 90 mg/kg) in an immune checkpoint humanized mouse model with no obvious toxicity. Finally, CH1 did not cause in vivo cardiotoxicity and bone marrow suppression (myelosuppression) to BALB/c mice. Taken together, these results suggest that CH1 deserves further investigation as a potent and safe PD-1/PDL-1 inhibitor for cancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Éteres Fenílicos/farmacología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Resorcinoles/farmacología , Células A549 , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Antígeno B7-H1/metabolismo , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Células HeLa , Células Hep G2 , Humanos , Células Jurkat , Células MCF-7 , Masculino , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/metabolismo , Ratones , Simulación del Acoplamiento Molecular/métodos , Éteres Fenílicos/química , Éteres Fenílicos/uso terapéutico , Receptor de Muerte Celular Programada 1/metabolismo , Resorcinoles/química , Resorcinoles/uso terapéutico
3.
J Med Chem ; 63(15): 8338-8358, 2020 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-32667799

RESUMEN

Novel small molecule compounds based on various scaffolds including chalcone, flavonoid, and resorcinol dibenzyl ether were designed and tested for their inhibitory activity against the Programmed Cell Death-1/Programmed Cell Death-Ligand 1 (PD-1/PD-L1) pathway. Among them, compound NP19 inhibited the human PD-1/PD-L1 interaction with IC50 values of 12.5 nM in homogeneous time-resolved fluorescence (HTRF) binding assays. In addition, NP19 dose-dependently elevated IFN-γ production in a coculture model of Hep3B/OS-8/hPD-L1 and CD3 T cells. Furthermore, NP19 displayed significant in vivo antitumor efficacy in two different mouse models of cancer (a melanoma B16-F10 tumor model and an H22 hepatoma tumor model). Moreover, H&E staining and flow cytometry data suggested that NP19 activated the immune microenvironment in the tumor, which may contribute to its antitumor effects. This work shows NP19 is a promising lead compound for further development as a new generation of small molecule inhibitors targeting the PD-1/PD-L1 pathway.


Asunto(s)
Antineoplásicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Éteres Fenílicos/farmacología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Resorcinoles/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Antígeno B7-H1/metabolismo , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Descubrimiento de Drogas , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/metabolismo , Masculino , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/metabolismo , Ratones , Ratones Endogámicos BALB C , Simulación del Acoplamiento Molecular , Éteres Fenílicos/química , Éteres Fenílicos/uso terapéutico , Receptor de Muerte Celular Programada 1/metabolismo , Mapas de Interacción de Proteínas/efectos de los fármacos , Ratas Sprague-Dawley , Resorcinoles/química , Resorcinoles/uso terapéutico , Transducción de Señal/efectos de los fármacos
4.
F1000Res ; 82019.
Artículo en Inglés | MEDLINE | ID: mdl-31069052

RESUMEN

Despite recent advances in understanding and treating trigeminal neuralgia, its management remains a considerable challenge. Better classification of different types of facial pain and the identification of prognostic factors for different treatment options lead the way toward better quality of life for the individual patient. Although the principles of treating trigeminal neuralgia remain basically the same, antiepileptic drugs, muscle relaxants, and neuroleptic agents are widely used medical treatment options. They were not originally developed for treating trigeminal neuralgia. Carbamazepine was studied in adequate placebo-controlled clinical trials in the 1960s and is still considered the most effective drug. Among emerging treatment options currently under clinical investigation are local botulinum neurotoxin type A injections and a novel sodium channel blocker (CNV1014802) that selectively blocks the Na v1.7 sodium channel. Non-pharmacological treatment options are non-invasive electrical stimulation with either transcranial direct-current stimulation or repetitive transcranial magnetic stimulation which both require further evaluation in regard to applicability. Surgical options remain a valid choice for patients not responding to medical treatment and include Gasserian ganglion percutaneous techniques, gamma knife surgery, and microvascular decompression. There is continual effort to improve these techniques and predict the outcome for better patient selection.


Asunto(s)
Radiocirugia , Estimulación Transcraneal de Corriente Directa , Neuralgia del Trigémino/terapia , Carbamazepina/uso terapéutico , Humanos , Éteres Fenílicos/uso terapéutico , Prolina/análogos & derivados , Prolina/uso terapéutico , Calidad de Vida , Bloqueadores de los Canales de Sodio/uso terapéutico , Neuralgia del Trigémino/tratamiento farmacológico
5.
J Cancer Res Ther ; 15(1): 120-125, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30880766

RESUMEN

AIM OF STUDY: This study is to investigate the effects of a novel peroxisome proliferator-activated receptor (PPAR) α/γ dual agonist TZD18 on cell growth, apoptosis, caspase activity, mitochondrial membrane potential, cytochrome c release, and apoptotic-related protein expression in MKN-45 cells. MATERIALS AND METHODS: 3-(4, 5-dimethylthiazolyl)-2,5-diphenyltetrazolium bromide assay against various human cancer cell lines was performed to investigate the whether TZD18 could in reduce the proliferation rates of cancer cells. The percentages of apoptotic cells and mitochondrial membrane potential level were determined by flow cytometry. The subcellular localization of cytochrome c was examined by immunofluorescence microscopy. Western blotting assay was performed to reveal the expression of apoptosis-related proteins. RESULTS: The results showed that the administration of TZD18 could inhibit the growth of MKN-45 cells in a dose- and time-dependent manner. In addition, the apoptotic ratio increased sharply along with a significant increase of caspase activities, mitochondrial membrane potential, and cytochrome c release following TZD18 exposure. The expression of Bax and p27kip1 increased significantly, whereas the expression level of Bcl-2 protein was downregulated. CONCLUSION: These results indicated that the administration of PPAR α/γ agonist TZD18 may inhibit cell growth by inducing the apoptotic process in MKN-45 cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Éteres Fenílicos/farmacología , Neoplasias Gástricas/tratamiento farmacológico , Tiazolidinedionas/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Humanos , PPAR alfa/agonistas , PPAR gamma/agonistas , Éteres Fenílicos/uso terapéutico , Neoplasias Gástricas/patología , Tiazolidinedionas/uso terapéutico
6.
Am J Trop Med Hyg ; 99(5): 1201-1205, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30226153

RESUMEN

Dipylidium caninum is a cosmopolitan cestode infecting dogs, cats, and humans. Praziquantel is a highly effective cestocidal drug and resistance in adult cestodes has not been reported. From 2016 to 2018, a population of dogs with cestode infections that could not be eliminated despite multiple treatments with praziquantel or epsiprantel was identified. Cases of D. caninum were clinically resistant to praziquantel and could not be resolved despite increasing the dose, frequency, and duration of treatment. Resistant isolates were identified and characterized by sequencing the 28S, 12S, and voltage-gated calcium channel beta subunit genes. Cases were only resolved following treatment with nitroscanate or a compounded pyrantel/praziquantel/oxantel product. Clinicians should be aware of this alarming development as treatment options for cestodes are limited in both human and veterinary medicine.


Asunto(s)
Antihelmínticos/farmacología , Cestodos/efectos de los fármacos , Infecciones por Cestodos/veterinaria , Enfermedades de los Perros/tratamiento farmacológico , Resistencia a Múltiples Medicamentos , Praziquantel/farmacología , Animales , Antihelmínticos/uso terapéutico , Cestodos/genética , Infecciones por Cestodos/tratamiento farmacológico , Enfermedades de los Perros/parasitología , Perros , Heces/parasitología , Éteres Fenílicos/uso terapéutico , Praziquantel/análogos & derivados , Praziquantel/uso terapéutico , Pirantel/análogos & derivados , Pirantel/uso terapéutico , ARN Ribosómico/genética , ARN Ribosómico 28S/genética , Tiocianatos/uso terapéutico , Resultado del Tratamiento
7.
Expert Opin Biol Ther ; 18(10): 1049-1059, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30129371

RESUMEN

INTRODUCTION: Stargardt macular dystrophy (STGD1) is a hereditary retinal degeneration that lacks effective treatment options. Gene therapy, stem cell therapy, and pharmacotherapy with visual cycle modulators (VCMs) and complement inhibitors are discussed as potential treatments. AREAS COVERED: Investigational therapies for STGD1 aim to reduce toxic bisretinoids and lipofuscin in the retina and retinal pigment epithelium (RPE). These agents include C20-D3-vitamin A (ALK-001), isotretinoin, VM200, emixustat, and A1120. Avacincaptad pegol is a C5 complement inhibitor that may reduce inflammation-related RPE damage. Animal models of STGD1 show promising data for these treatments, though proof of efficacy in humans is lacking. Fenretinide and emixustat are VCMs for dry AMD and STGD1 that failed to halt geographic atrophy progression or improve vision in trials for AMD. A1120 prevents retinol transport into RPE and may spare side effects typically seen with VCMs (nyctalopia and chromatopsia). Stem cell transplantation suggests potential biologic plausibility in a phase I/II trial. Gene therapy aims to augment the mutated ABCA4 gene, though results of a phase I/II trial are pending. EXPERT OPINION: Stem cell transplantation, ABCA4 gene therapy, VCMs, and complement inhibitors offer biologically plausible treatment mechanisms for treatment of STGD1. Further trials are warranted to assess efficacy and safety in humans.


Asunto(s)
Degeneración Macular/congénito , Terapias en Investigación/métodos , Terapias en Investigación/tendencias , Transportadoras de Casetes de Unión a ATP/genética , Animales , Inactivadores del Complemento/uso terapéutico , Terapia Genética/métodos , Terapia Genética/tendencias , Humanos , Lipofuscina/uso terapéutico , Degeneración Macular/terapia , Éteres Fenílicos/uso terapéutico , Propanolaminas/uso terapéutico , Enfermedad de Stargardt , Trasplante de Células Madre
8.
Biochem Pharmacol ; 150: 267-279, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29476714

RESUMEN

RDD648, a novel derivative of a natural molecule riccardin D, exhibited potent anticancer activity by targeting lysosomes in vitro and in vivo. Mechanistic studies revealed that RDD648 facilitated STAT3 to translocate into the nucleus, and this activity was involved in lysosome-mediated cell death as evidenced by our finding that inhibition of STAT3 alleviated lysosomal membrane permeabilization. Further investigation indicated that nuclear STAT3 directly interacted with transcription factor TFEB, leading to the partial loss of function of TFEB, which is essential for lysosome turnover. The present study first uncovers that STAT3 contributes to lysosomal-mediated cell death in RDD648-treated breast cancer cells though interacting with TFEB, and the findings may be significant in the design of treatments for breast cancers where STAT3 is constitutively expressed.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Neoplasias de la Mama/metabolismo , Lisosomas/metabolismo , Éteres Fenílicos/farmacología , Factor de Transcripción STAT3/metabolismo , Estilbenos/farmacología , Animales , Neoplasias de la Mama/tratamiento farmacológico , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Relación Dosis-Respuesta a Droga , Estrés del Retículo Endoplásmico/efectos de los fármacos , Estrés del Retículo Endoplásmico/fisiología , Femenino , Humanos , Lisosomas/efectos de los fármacos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Éteres Fenílicos/uso terapéutico , Factor de Transcripción STAT3/antagonistas & inhibidores , Estilbenos/uso terapéutico
9.
Mol Pharm ; 15(1): 116-126, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29185769

RESUMEN

By screening a drug library comprising FDA approved compounds, we discovered a potent interaction between the antifungal agent haloprogin and the experimental organometallic drug RAPTA-T, to synergistically induce cancer cell killing. The combination of these two small molecules, even at low doses, elicited an improved therapeutic response on tumor growth over either agent alone or the current treatment used in the clinic in the highly aggressive syngeneic B16F10 melanoma tumor model, where classical cytotoxic chemotherapeutic agents show little efficacy. The combination with the repurposed chemodrug haloprogin provides the basis for a new powerful treatment option for cutaneous melanoma. Importantly, because synergistic induction of tumor cell death is achieved with low individual drug doses, and cellular targets for RAPTA-T are different from those of classical chemotherapeutic drugs, a therapeutic strategy based on this approach could avoid toxicities and potentially resistance mechanisms, and could even inhibit metastatic progression.


Asunto(s)
Antifúngicos/uso terapéutico , Reposicionamiento de Medicamentos/métodos , Melanoma/tratamiento farmacológico , Compuestos Organometálicos/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Antifúngicos/química , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Compuestos Organometálicos/química , Éteres Fenílicos/uso terapéutico , Melanoma Cutáneo Maligno
10.
Eur Rev Med Pharmacol Sci ; 21(24): 5765-5773, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-29272013

RESUMEN

OBJECTIVE: To investigate the changes in myocardial energy metabolism and the effect of peroxisome proliferator-activated receptor alpha/gamma (PPARα/γ) dual agonist TZD18 on myocardial energy metabolism in rats with heart failure after myocardial infarction. MATERIALS AND METHODS: The myocardial infarction model was established by ligating the left anterior descending coronary artery. The rats were randomly divided into the myocardial infarction group (MI group), the TZD18 intervention group (TZD18 group), and the shame surgery group (sham group). 8 weeks later, the blood flow parameters were measured by carotid arterial cannulas, and ventricular remodeling indexes were calculated. Hearts were extracted from rats after the execution. The expressions of PPARα/γ mRNA and α/ß-MHC mRNA were detected by reverse transcription-polymerase chain reaction (RT-PCR). The mitochondrial oxidative respiration activity was measured by a bio-tissue oxygen consumption meter, the content of adenosine in mitochondria was measured by high-performance liquid chromatography, and tritium-labeled adenosine diphosphate incorporation assay was used to detect the transport activity of adenosine nucleotide translocases (ANT). RESULTS: The expression of PPARα/γ mRNA and the ratio of α/ß-MHC mRNA in the MI group were significantly decreased, the content of high energy phosphates, respiration activity, ANT transport activity in mitochondria were significantly decreased, the hemodynamic indexes were disturbed and left ventricular weight/body weight ratio (LVW/BW) significantly became higher. TZD18 intervention could increase the expression level of PPARα/γ mRNA and up-regulate the ratio of α/ß-MHC mRNA, thus improving mitochondrial respiratory activity and ANT transport activity in rats with heart failure after myocardial infarction, increasing the content of high energy phosphates in mitochondria and improving the remodeling indexes in the ventricle. CONCLUSIONS: TZD18 increases both the expression of enzymes related to myocardial energy metabolism and the content of high-energy phosphates in mitochondria. Also, it improves the respiratory activity and ANT transport activity by activating PPARα/γ genes, thus improving the generation and delivery of myocardial energy and protecting the myocardial cells.


Asunto(s)
Infarto del Miocardio/tratamiento farmacológico , Miocardio/metabolismo , PPAR alfa/agonistas , PPAR gamma/agonistas , Éteres Fenílicos/uso terapéutico , Tiazolidinedionas/uso terapéutico , Remodelación Ventricular , Animales , Masculino , Mitocondrias/metabolismo , Infarto del Miocardio/metabolismo , PPAR alfa/genética , PPAR gamma/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar
11.
PLoS One ; 12(7): e0180319, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28746336

RESUMEN

The beta-1 adrenergic receptor (ADRB1) is a promising therapeutic target intrinsically involved in the cognitive deficits and pathological features associated with Alzheimer's disease (AD). Evidence indicates that ADRB1 plays an important role in regulating neuroinflammatory processes, and activation of ADRB1 may produce neuroprotective effects in neuroinflammatory diseases. Novel small molecule modulators of ADRB1, engineered to be highly brain permeable and functionally selective for the G protein with partial agonistic activity, could have tremendous value both as pharmacological tools and potential lead molecules for further preclinical development. The present study describes our ongoing efforts toward the discovery of functionally selective partial agonists of ADRB1 that have potential therapeutic value for AD and neuroinflammatory disorders, which has led to the identification of the molecule STD-101-D1. As a functionally selective agonist of ADRB1, STD-101-D1 produces partial agonistic activity on G protein signaling with an EC50 value in the low nanomolar range, but engages very little beta-arrestin recruitment compared to the unbiased agonist isoproterenol. STD-101-D1 also inhibits the tumor necrosis factor α (TNFα) response induced by lipopolysaccharide (LPS) both in vitro and in vivo, and shows high brain penetration. Other than the therapeutic role, this newly identified, functionally selective, partial agonist of ADRB1 is an invaluable research tool to study mechanisms of G protein-coupled receptor signal transduction.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 1/uso terapéutico , Encéfalo/metabolismo , Proteínas de Unión al GTP/metabolismo , Trastornos Neurocognitivos/tratamiento farmacológico , Receptores Adrenérgicos beta 1/metabolismo , Agonistas de Receptores Adrenérgicos beta 1/química , Agonistas de Receptores Adrenérgicos beta 1/farmacocinética , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Animales , Células CHO , Línea Celular Tumoral , Células Cultivadas , Cricetinae , Cricetulus , Cristalografía por Rayos X , Descubrimiento de Drogas , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Ratones Endogámicos C57BL , Modelos Químicos , Modelos Moleculares , Estructura Molecular , Trastornos Neurocognitivos/metabolismo , Permeabilidad , Éteres Fenílicos/química , Éteres Fenílicos/farmacocinética , Éteres Fenílicos/uso terapéutico , Propanolaminas/química , Propanolaminas/farmacocinética , Propanolaminas/uso terapéutico , Unión Proteica , Ratas Sprague-Dawley , Receptores Adrenérgicos beta 1/química , Relación Estructura-Actividad
12.
Sci Rep ; 7(1): 1728, 2017 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-28496142

RESUMEN

This study aimed to investigate the role of src-homology protein tyrosine phosphatase-1 (SHP-1)-signal transducer and activator of transcription 3 (STAT3) pathway in liver fibrogenesis and the anti-fibrotic effect of SHP-1 agonist. The antifibrotic activity of SC-43, a sorafenib derivative with an enhanced SHP-1 activity, was evaluated in two fibrosis mouse models by carbon tetrachloride induction and bile duct ligation. Rat, human, and primary mouse hepatic stellate cells (HSCs) were used for mechanistic investigations. The results showed that SHP-1 protein primarily localized in fibrotic areas of human and mouse livers. SC-43 treatment reduced the activated HSCs and thus effectively prevented and regressed liver fibrosis in both fibrosis mouse models and improved mouse survival. In vitro studies revealed that SC-43 promoted HSC apoptosis, increased the SHP-1 activity and inhibited phospho-STAT3. The enhanced SHP-1 activity in HSCs significantly inhibited HSC proliferation, whereas SHP-1 inhibition rescued SC-43-induced HSC apoptosis. Furthermore, SC-43 interacted with the N-SH2 domain of SHP-1 to enhance the activity of SHP-1 as its antifibrotic mechanism. In conclusion, the SHP-1-STAT3 pathway is crucial in fibrogenesis. SC-43 significantly ameliorates liver fibrosis through SHP-1 upregulation. A SHP-1-targeted antifibrotic therapy may represent a druggable strategy for antifibrotic drug discovery.


Asunto(s)
Cirrosis Hepática/tratamiento farmacológico , Éteres Fenílicos/uso terapéutico , Compuestos de Fenilurea/uso terapéutico , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Animales , Apoptosis/efectos de los fármacos , Conductos Biliares/patología , Tetracloruro de Carbono , Línea Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Humanos , Ligadura , Cirrosis Hepática/patología , Cirrosis Hepática/prevención & control , Masculino , Ratones Endogámicos C57BL , Mutación/genética , Éteres Fenílicos/química , Éteres Fenílicos/farmacología , Compuestos de Fenilurea/química , Compuestos de Fenilurea/farmacología , Dominios Proteicos , Proteína Tirosina Fosfatasa no Receptora Tipo 6/química , Ratas , Factor de Transcripción STAT3/metabolismo , Sorafenib/química , Sorafenib/farmacología , Sorafenib/uso terapéutico
13.
Toxins (Basel) ; 8(3)2016 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-26999206

RESUMEN

Loss-of-function mutations of Na(V)1.7 lead to congenital insensitivity to pain, a rare condition resulting in individuals who are otherwise normal except for the inability to sense pain, making pharmacological inhibition of Na(V)1.7 a promising therapeutic strategy for the treatment of pain. We characterized a novel mouse model of Na(V)1.7-mediated pain based on intraplantar injection of the scorpion toxin OD1, which is suitable for rapid in vivo profiling of Na(V)1.7 inhibitors. Intraplantar injection of OD1 caused spontaneous pain behaviors, which were reversed by co-injection with Na(V)1.7 inhibitors and significantly reduced in Na(V)1.7(-/-) mice. To validate the use of the model for profiling Na(V)1.7 inhibitors, we determined the Na(V) selectivity and tested the efficacy of the reported Na(V)1.7 inhibitors GpTx-1, PF-04856264 and CNV1014802 (raxatrigine). GpTx-1 selectively inhibited Na(V)1.7 and was effective when co-administered with OD1, but lacked efficacy when delivered systemically. PF-04856264 state-dependently and selectively inhibited Na(V)1.7 and significantly reduced OD1-induced spontaneous pain when delivered locally and systemically. CNV1014802 state-dependently, but non-selectively, inhibited Na(V) channels and was only effective in the OD1 model when delivered systemically. Our novel model of Na(V)1.7-mediated pain based on intraplantar injection of OD1 is thus suitable for the rapid in vivo characterization of the analgesic efficacy of Na(V)1.7 inhibitors.


Asunto(s)
Canal de Sodio Activado por Voltaje NAV1.7/fisiología , Dolor/tratamiento farmacológico , Péptidos/uso terapéutico , Éteres Fenílicos/uso terapéutico , Prolina/análogos & derivados , Venenos de Escorpión/uso terapéutico , Bloqueadores de los Canales de Sodio/uso terapéutico , Venenos de Araña/uso terapéutico , Analgésicos , Animales , Conducta Animal/efectos de los fármacos , Células CHO , Cricetulus , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Masculino , Ratones Endogámicos C57BL , Canal de Sodio Activado por Voltaje NAV1.7/genética , Fibras Nerviosas/efectos de los fármacos , Fibras Nerviosas/fisiología , Dolor/inducido químicamente , Prolina/uso terapéutico , Vena Safena/inervación , Sulfonamidas/uso terapéutico
14.
J Med Food ; 18(6): 677-84, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25919915

RESUMEN

We investigated the protective ability of 3-(4'-hydroxyl-3',5'-dimethoxyphenyl)propionic acid (HDMPPA), an active principle in Korean cabbage kimchi, against the production of proinflammatory mediators and cytokines, and the mechanisms involved in lipopolysaccharide (LPS)-stimulated BV2 microglial cells. HDMPPA significantly suppressed the production of nitric oxide (NO) and prostaglandin E2, along with the expression of inducible NO synthase and cyclooxygenase-2 in LPS-stimulated BV2 cells, at concentrations with no cytotoxicity. HDMPPA also attenuated the LPS-induced expression and secretion of proinflammatory cytokines, such as tumor necrosis factor-α and interleukin-1ß. Furthermore, HDMPPA inhibited LPS-induced nuclear factor-κB (NF-κB) activation, which was associated with the abrogation of IκB-α degradation and phosphorylation, and subsequent decreases in NF-κB p65 levels. Moreover, the phosphorylation of mitogen-activated protein kinases (MAPKs) and Akt, a downstream molecule of phosphatidylinositol-3-kinase (PI3K), in LPS-stimulated BV2 cells was suppressed markedly by HDMPPA. This effect was associated with a significant reduction in the formation of intracellular reactive oxygen species. The findings in this study suggest that HDMPPA may exert anti-inflammatory responses by suppressing LPS-induced expression of proinflammatory mediators and cytokines through blockage of NF-κB, MAPKs, and PI3K/Akt signaling pathways and oxidative stress in microglia.


Asunto(s)
Antiinflamatorios/uso terapéutico , Brassica/química , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Inflamación/tratamiento farmacológico , Microglía/efectos de los fármacos , Éteres Fenílicos/uso terapéutico , Propionatos/uso terapéutico , Animales , Antiinflamatorios/farmacología , Línea Celular , Fermentación , Inflamación/inducido químicamente , Inflamación/metabolismo , Lipopolisacáridos , Ratones , Microglía/metabolismo , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Estrés Oxidativo/efectos de los fármacos , Éteres Fenílicos/farmacología , Fitoterapia , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Propionatos/farmacología , Verduras
15.
Int J Toxicol ; 33(4): 319-324, 2014 07.
Artículo en Inglés | MEDLINE | ID: mdl-24801487

RESUMEN

The aim of this study was to compare the cytotoxic effects of a newly synthesized thialo benzene derivative 2,4-dithiophenoxy-1-iodo-4-bromobenzene (C18H12S2IBr) and a well-known antifungal agent, fluconazole, in L929 cells. L929 cells were treated with 250, 500, or 1000 µg/mL of C18H12S2IBr and with the same doses of fluconazole. Cytotoxicity tests including 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), lactate dehydrogenase (LDH) leakage, and protein content were compared. Glucose and lactate concentrations were measured to determine alterations in metabolic activity. Apoptosis was investigated by TUNEL test and results were supported with survivin enzyme-linked immunosorbent assay. Treatment with C18H12S2IBr resulted in a concentration-dependent cytotoxicity as indicated by MTT, LDH leakage assay, and decreased protein concentration. The loss of cell viability and the increased LDH leakage in 500 µg/mL and 1000 µg/mL C18H12S2IBr and fluconazole groups indicated cell membrane damage and necrotic cell death. In all groups, metabolic activities were altered but apoptosis was not induced. We have previously investigated lower doses of C18H12S2IBr; there was no cytotoxicity in L929 cells. In this study, higher doses caused cytotoxicity and alterations in metabolic activity . When we consider the similar results obtained from fluconazole and especially the lowest dose of C18H12S2IBr, this newly synthesized compound may be a good alternative antifungal agent.


Asunto(s)
Antifúngicos/efectos adversos , Bromobencenos/efectos adversos , Drogas en Investigación/efectos adversos , Metabolismo Energético/efectos de los fármacos , Yodobencenos/efectos adversos , Éteres Fenílicos/efectos adversos , Compuestos de Sulfhidrilo/efectos adversos , Animales , Antifúngicos/uso terapéutico , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Bromobencenos/uso terapéutico , Candida/efectos de los fármacos , Candida/crecimiento & desarrollo , Candidiasis/tratamiento farmacológico , Línea Celular , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Drogas en Investigación/uso terapéutico , Fluconazol/efectos adversos , Fluconazol/uso terapéutico , Etiquetado Corte-Fin in Situ , Proteínas Inhibidoras de la Apoptosis/metabolismo , Yodobencenos/uso terapéutico , Ratones , Concentración Osmolar , Éteres Fenílicos/uso terapéutico , Proteínas Represoras/metabolismo , Compuestos de Sulfhidrilo/uso terapéutico , Survivin
16.
Basic Clin Pharmacol Toxicol ; 115(6): 488-98, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24836118

RESUMEN

Riccardin D, a liverwort-derived naturally occurring macrocyclic bisbibenzyl, has been found to exert anticancer effects in multiple cancer cell types. In this study, we investigated the effect and mechanism of Riccardin D on human breast cancer. Experiments were performed on human breast cancer MCF-7 and MDA-MB-231 cells. The antitumour effects of Riccardin D were assessed by the 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay and human breast cancer xenografts mice model. TRAPeze(®) XL Telomerase Detection assay was used for the detection of telomerase activity. γ-H2 AX foci formation was tested for the induction of DNA damage response. Cell cycle distribution was analysed by flow cytometry, and cell apoptosis was determined by annexin V-FITC/PI staining, terminal deoxynucleotidyl transferase-mediated dUTP nick end labelling (TUNEL) assay and Western blotting. Riccardin D effectively inhibited the growth of MCF-7 and MDA-MB-231 cells in vitro. And Riccardin D also effectively delayed the growth of MCF-7 and MDA-MB-231-luc-D3H2LN xenografts without significant loss of body-weight. Further analysis suggested that Riccardin D's effects may arise from its suppression of telomerase activity, which led to telomere dysfunction. Telomerase inhibition and telomere dysfunction could activate the canonical ataxia telangiectasia-mutated (ATM) kinase-mediated DNA damage response, as shown by elevated expression of γ-H2 AX, p-ATM and p-Chk2. This is finally followed by the induction of cell cycle arrest and apoptosis, as shown by the increase of TUNEL-stained cells, caspase activation, PARP cleavage and the increase of bax/bcl-2 ratio. Moreover, Riccardin D induced p53-proficient MCF-7 cells to arrest in G1 phase and p53-deficient MDA-MB-231 cells to arrest in G2/M phase. Overall, these results demonstrate that Riccardin D may inhibit human breast cancer growth through suppression of telomerase activity.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Éteres Fenílicos/uso terapéutico , Estilbenos/uso terapéutico , Telomerasa/antagonistas & inhibidores , Animales , Western Blotting , Línea Celular Tumoral , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Etiquetado Corte-Fin in Situ , Células MCF-7/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Reacción en Cadena en Tiempo Real de la Polimerasa
17.
Eur J Pharmacol ; 723: 473-80, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24334279

RESUMEN

Osteoclasts are polykaryons that have the unique capacity to degrade bone. Modulation of osteoclast formation and function is a promising strategy for the treatment of bone-destructive diseases. Here, we report that obovatol, a natural compound isolated from Magnolia obovata, inhibits receptor activator of nuclear factor kappa B (NF-κB) ligand (RANKL)-induced osteoclast differentiation in vitro and inflammatory bone loss in vivo. We found that obovatol strongly inhibited osteoclast formation from bone marrow-derived macrophages in a dose-dependent manner without cytotoxicity. Obovatol significantly suppressed RANKL-induced activation of NF-κB, c-Jun-N-terminal kinase, and extracellular signal-regulated kinase signaling pathways. Obovatol also inhibited RANKL-induced expression of the genes c-Fos and nuclear factor of activated T cells c1, which are transcription factors important for osteoclastogenesis. In addition to osteoclast differentiation, obovatol blocked cytoskeletal organization and abrogated the bone resorbing activity of mature osteoclast. Obovatol also accelerated osteoclast apoptosis through the induction of caspase-3 activation. Consistent with its in vitro anti-resorptive effect, obovatol prevented bone loss induced by lipopolysaccharide in vivo. Together, our data suggest that obovatol may be a useful therapeutic agent for the treatment of pathological bone disorders characterized by excessive osteoclastic bone resorption.


Asunto(s)
Compuestos de Bifenilo/farmacología , Compuestos de Bifenilo/uso terapéutico , Conservadores de la Densidad Ósea/farmacología , Conservadores de la Densidad Ósea/uso terapéutico , Resorción Ósea/tratamiento farmacológico , Osteoclastos/efectos de los fármacos , Éteres Fenílicos/farmacología , Éteres Fenílicos/uso terapéutico , Animales , Densidad Ósea/efectos de los fármacos , Resorción Ósea/inducido químicamente , Resorción Ósea/diagnóstico por imagen , Caspasa 3/metabolismo , Diferenciación Celular/efectos de los fármacos , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Lipopolisacáridos , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ligando RANK/farmacología , Radiografía
18.
J Pharmacol Sci ; 122(3): 223-31, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23877018

RESUMEN

Glioblastoma is the most lethal brain cancer. In spite of intensive therapy, the prognosis of patients with glioblastoma is very poor. To discover novel therapeutic agents, we screened a combinatorial compound library containing 372 thiazolidinone compounds using U87MG human glioblastoma cells. (2E,5Z)-5-(2-hydroxybenzylidene)-2-((4-phenoxyphenyl)imino) thiazolidin-4-one (HBPT) was identified as the most potent anti-glioblastoma compound. HBPT inhibits U87MG human glioblastoma cell proliferation with an IC50 of 20 µM, which is almost 5-fold more potent than temozolomide (a widely used drug for treating malignant glioma in the clinic). Mechanistic investigation demonstrated that HBPT is a novel microtubule-depolymerizing agent, which arrests cancer cells at the G2/M phase of the cell cycle and induces cell apoptosis. In the mouse U87MG xenograft model, HBPT elicits a robust tumor inhibitory effect. More importantly, no obvious toxicity was observed for HBPT therapy in animal experiments. These findings indicate that HBPT has the potential to be developed as a novel agent for the treatment of glioblastoma. [Supplementary Tables: available only at http://dx.doi.org/10.1254/jphs.13064FP].


Asunto(s)
Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Microtúbulos/metabolismo , Microtúbulos/patología , Éteres Fenílicos/farmacología , Éteres Fenílicos/uso terapéutico , Tiazolidinas/farmacología , Tiazolidinas/uso terapéutico , Moduladores de Tubulina , Animales , Antineoplásicos/química , Apoptosis/efectos de los fármacos , División Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Modelos Animales de Enfermedad , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Éteres Fenílicos/química , Temozolomida , Tiazolidinas/química , Células Tumorales Cultivadas
19.
Antibiot Khimioter ; 57(5-6): 28-31, 2012.
Artículo en Ruso | MEDLINE | ID: mdl-23156041

RESUMEN

The aim of the study was to estimate the use of hypoxen (antihypoxant) during the early postoperative course in gynecologic patients. The patients were divided into two groups according to the treatment scheme. 339 patients of the control group were under the routine therapy after the laparatomic gynecologic operations. 52 patients were additionally treated with hypoxen (antioxidant) (the main group). The impact of hypoxen on the antioxidant system and lipid peroxidation and its preventive effect on prolongation of the postoperative pain syndrom were estimated. The results of the study showed that hypoxen inhibited lipid peroxidation and activated the antioxidant system in the postoperative patients vs. the control group. It was also observed that among the patients additionally treated with hypoxen the percentage of those with prolonged postoperative pains was statistically lower (p<0.05, chi2 test). It was concluded that hypoxen normalized lipid peroxidation and antioxidant system after abdominal gynecologic operations. The additional use of hypoxen in the routine therapy of the gynecologic patients prevented prolongation of the postoperative pain syndrom.


Asunto(s)
Procedimientos Quirúrgicos Ginecológicos/efectos adversos , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Dolor Postoperatorio/tratamiento farmacológico , Dolor Postoperatorio/prevención & control , Éteres Fenílicos/uso terapéutico , Adulto , Anciano , Femenino , Humanos , Persona de Mediana Edad , Factores de Tiempo , Adulto Joven
20.
PLoS One ; 7(3): e33243, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22432006

RESUMEN

BACKGROUND: Mutation of tumor suppressor gene, adenomatous polyposis coli (APC), is the primary molecular event in the development of most intestinal carcinomas. Animal model with APC gene mutation is an effective tool for study of preventive approaches against intestinal carcinomas. We aimed to evaluate the effect of Riccardin D, a macrocyclic bisbibenzyl compound, as a chemopreventive agent against intestinal adenoma formation in APC(Min/+) mice. METHODS: APC(Min/+) mice were given Riccardin D by p.o. gavage for 7 weeks. Mice were sacrificed, and the number, size and histopathology of intestinal polyps were examined under a microscope. We performed immunohistochemical staining, western blotting, reverse transcriptase-polymerase chain reaction (RT-PCR) and enzyme-linked immunosorbent assay (ELISA) in intestinal polyps to investigate the mechanism of chemopreventive effect of Riccardin D. RESULTS: Riccardin D treatment resulted in a significant inhibition of intestinal adenoma formation, showing a reduction of polyp number by 41.7%, 31.1% and 44.4%, respectively, in proximal, middle and distal portions of small intestine. The activity of Riccardin D against polyp formation was more profound in colon, wherein Riccardin D decreased polyp number by 79.3%. Size distribution analysis revealed a significant reduction in large-size polyps (2-3 mm) by 40.0%, 42.5% and 33.3%, respectively, in proximal, middle and distal portions of small intestine, and 77.8% in colon. Histopathological analysis of the intestinal polyps revealed mostly hyperplastic morphology without obvious dysplasia in Riccardin D-treated mice. Molecular analyses of the polyps suggested that the inhibitory effect of Riccardin D on intestinal adenoma formation was associated with its abilities of reduction in cell proliferation, induction of apoptosis, antiangiogenesis, inhibition of the Wnt signaling pathway and suppression of inflammatory mediators in polyps. CONCLUSIONS: Our results suggested that Riccardin D exerts its chemopreventive effect against intestinal adenoma formation through multiple mechanisms including anti-proliferative, apoptotic, anti-angiogenic and anti-inflammatory activity.


Asunto(s)
Adenoma/prevención & control , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Productos Biológicos/farmacología , Hepatophyta/química , Neoplasias Intestinales/prevención & control , Éteres Fenílicos/farmacología , Lesiones Precancerosas/prevención & control , Estilbenos/farmacología , Adenoma/irrigación sanguínea , Adenoma/patología , Animales , Apoptosis/efectos de los fármacos , Productos Biológicos/química , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Colon/efectos de los fármacos , Colon/patología , Ciclina D1/metabolismo , Ciclooxigenasa 2/metabolismo , Inflamación/patología , Neoplasias Intestinales/irrigación sanguínea , Neoplasias Intestinales/patología , Pólipos Intestinales/enzimología , Pólipos Intestinales/patología , Pólipos Intestinales/prevención & control , Intestino Delgado/efectos de los fármacos , Intestino Delgado/patología , Ratones , FN-kappa B/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Éteres Fenílicos/química , Éteres Fenílicos/uso terapéutico , Lesiones Precancerosas/patología , Transducción de Señal/efectos de los fármacos , Estilbenos/química , Estilbenos/uso terapéutico , beta Catenina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA