Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 16(12)2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38927958

RESUMEN

Triple-negative breast cancer (TNBC) is the most aggressive subtype with high metastasis and mortality rates. Given the lack of actionable targets such as ER and HER2, TNBC still remains an unmet therapeutic challenge. Despite harboring high CDK4/6 expression levels, the efficacy of CDK4/6 inhibition in TNBC has been limited due to the emergence of resistance. The resistance to CDK4/6 inhibition is mainly mediated by RB1 inactivation. Since our aim is to overcome resistance to CDK4/6 inhibition, in this study, we primarily used the cell lines that do not express RB1. Following a screening for activated receptor tyrosine kinases (RTKs) upon CDK4/6 inhibition, we identified the TAM (Tyro3, Axl, and MerTK) RTKs as a crucial therapeutic vulnerability in TNBC. We show that targeting the TAM receptors with a novel inhibitor, sitravatinib, significantly sensitizes TNBC to CDK4/6 inhibitors. Upon prolonged HER2 inhibitor treatment, HER2+ breast cancers suppress HER2 expression, physiologically transforming into TNBC-like cells. We further show that the combined treatment is highly effective against drug-resistant HER2+ breast cancer as well. Following quantitative proteomics and RNA-seq data analysis, we extended our study into the immunophenotyping of TNBC. Given the roles of the TAM receptors in promoting the creation of an immunosuppressive tumor microenvironment (TME), we further demonstrate that the combination of CDK4/6 inhibitor abemaciclib and sitravatinib modifies the immune landscape of TNBC to favor immune checkpoint blockade. Overall, our study offers a novel and highly effective combination therapy against TNBC and potentially treatment-resistant HER2+ breast cancer that can be rapidly moved to the clinic.

2.
Cancer Res ; 82(12): 2254-2268, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-35413105

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is a highly metastatic disease with few effective treatments. Here we show that the mitochondrial calcium uniporter (MCU) promotes PDAC cell migration, invasion, metastasis, and metabolic stress resistance by activating the Keap1-Nrf2 antioxidant program. The cystine transporter SLC7A11 was identified as a druggable target downstream of the MCU-Nrf2 axis. Paradoxically, despite the increased ability to uptake cystine, MCU-overexpressing PDAC demonstrated characteristics typical of cystine-deprived cells and were hypersensitive to cystine deprivation-induced ferroptosis. Pharmacologic inhibitors of SLC7A11 effectively induced tumor regression and abrogated MCU-driven metastasis in PDAC. In patient-derived organoid models in vitro and patient-derived xenograft models in vivo, MCU-high PDAC demonstrated increased sensitivity to SLC7A11 inhibition compared with MCU-low tumors. These data suggest that MCU is able to promote resistance to metabolic stress and to drive PDAC metastasis in a cystine-dependent manner. MCU-mediated cystine addiction could be exploited as a therapeutic vulnerability to inhibit PDAC tumor growth and to prevent metastasis. SIGNIFICANCE: Elevated mitochondrial calcium uptake in PDAC promotes metastasis but exposes cystine addiction and ferroptosis sensitivity that could be targeted to improve pancreatic cancer treatment.


Asunto(s)
Canales de Calcio/metabolismo , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Calcio/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Cistina/metabolismo , Humanos , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas
3.
EMBO Mol Med ; 14(1): e14296, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34845836

RESUMEN

More than 60% of nonsmall cell lung cancer (NSCLC) patients show a positive response to the first ALK inhibitor, crizotinib, which has been used as the standard treatment for newly diagnosed patients with ALK rearrangement. However, most patients inevitably develop crizotinib resistance due to acquired secondary mutations in the ALK kinase domain, such as the gatekeeper mutation L1196M and the most refractory mutation, G1202R. Here, we develop XMU-MP-5 as a new-generation ALK inhibitor to overcome crizotinib resistance mutations, including L1196M and G1202R. XMU-MP-5 blocks ALK signaling pathways and inhibits the proliferation of cells harboring either wild-type or mutant EML4-ALK in vitro and suppresses tumor growth in xenograft mouse models in vivo. Structural analysis provides insights into the mode of action of XMU-MP-5. In addition, XMU-MP-5 induces significant regression of lung tumors in two genetically engineered mouse (GEM) models, further demonstrating its pharmacological efficacy and potential for clinical application. These preclinical data support XMU-MP-5 as a novel selective ALK inhibitor with high potency and selectivity. XMU-MP-5 holds great promise as a new therapeutic against clinically relevant secondary ALK mutations.


Asunto(s)
Quinasa de Linfoma Anaplásico/antagonistas & inhibidores , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Inhibidores de Proteínas Quinasas , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Resistencia a Antineoplásicos , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirazoles/uso terapéutico , Piridinas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Biochem Soc Trans ; 49(6): 2581-2589, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34854917

RESUMEN

The store-operated calcium (Ca2+) entry (SOCE) is the Ca2+ entry mechanism used by cells to replenish depleted Ca2+ store. The dysregulation of SOCE has been reported in metastatic cancer. It is believed that SOCE promotes migration and invasion by remodeling the actin cytoskeleton and cell adhesion dynamics. There is recent evidence supporting that SOCE is critical for the spatial and the temporal coding of Ca2+ signals in the cell. In this review, we critically examined the spatiotemporal control of SOCE signaling and its implication in the specificity and robustness of signaling events downstream of SOCE, with a focus on the spatiotemporal SOCE signaling during cancer cell migration, invasion and metastasis. We further discuss the limitation of our current understanding of SOCE in cancer metastasis and potential approaches to overcome such limitation.


Asunto(s)
Calcio/metabolismo , Metástasis de la Neoplasia , Neoplasias/metabolismo , Señalización del Calcio , Humanos , Transporte Iónico , Neoplasias/patología
5.
Cancer Lett ; 518: 230-242, 2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-34303764

RESUMEN

Fascin is a pro-metastatic actin-bundling protein that is upregulated in all metastatic carcinomas. Fascin promotes cancer cell migration and invasion by facilitating membrane protrusions, such as filopodia and invadopodia. Aerobic glycolysis is a key feature of cancer metabolism and provides critical intermediate metabolites for tumor growth. Here, we report that fascin increases glycolysis in lung cancer to promote tumor growth and metastasis. Fascin promotes glycolytic flux by increasing the expression and activities of phosphofructose-kinases 1 and 2 (PFK1 and 2). Fascin mediates glycolytic functions via activation of yes-associated protein 1 (YAP1) through its canonical actin-bundling activity by promoting the binding of YAP1 to a TEAD1/4 binding motif located 30 bp upstream of the PFKFB3 transcription start site to activate its transcription. Examination of the TCGA database suggests that the fascin-YAP1-PFKFB3 axis is likely conserved across different types of cancers. Importantly, pharmacological inhibitors of fascin suppressed YAP1-PFKFB3 signaling and glycolysis in cancer cell lines, organoid cultures, and xenograft metastasis models. Taken together, our data reveal that the glycolytic function of fascin is essential for the promotion of lung cancer growth and metabolism, and suggest that pharmacological inhibitors of fascin may be used to reprogram cancer metabolism in lung and potentially other cancers with fascin upregulation.


Asunto(s)
Proteínas Portadoras/genética , Proliferación Celular/genética , Glucólisis/genética , Neoplasias Pulmonares/genética , Proteínas de Microfilamentos/genética , Metástasis de la Neoplasia/genética , Fosfofructoquinasa-2/genética , Células A549 , Animales , Línea Celular Tumoral , Femenino , Células Hep G2 , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/patología , Transducción de Señal/genética , Transcripción Genética/genética , Proteínas Señalizadoras YAP/genética
6.
Eur J Med Chem ; 206: 112697, 2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-32814244

RESUMEN

Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death worldwide and targeted therapeutics exhibit limited success. Polo-like kinase 1 (PLK1), a Ser/Thr kinase, plays a pivotal role in cell-cycle regulation and is considered a promising target in HCC. Here, via structural optimization using both biochemical kinase assays and cellular antiproliferation assays, we discovered a potent and selective PLK1 kinase inhibitor, compound 31. Compound 31 exhibited biochemical activity with IC50 of < 0.508 nM against PLK1 and a KINOMEscan selectivity score (S(1)) of 0.02 at a concentration of 1 µM. Furthermore, 31 showed broad antiproliferative activity against a variety of cancer cell lines, with the lowest antiproliferative IC50 (11.1 nM) in the HCC cell line HepG2. A detailed mechanistic study of 31 revealed that inhibition of PLK1 by 31 induces mitotic arrest at the G2/M phase checkpoint, thus leading to cancer cell apoptosis. Moreover, 31 exhibited profound antitumor efficacy in a xenograft mouse model. Collectively, these results establish compound 31 as a good starting point for the development of PLK1 targeted therapeutics for HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Diseño de Fármacos , Neoplasias Hepáticas/patología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Tiofenos/química , Tiofenos/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Células Hep G2 , Humanos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Ratones , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasa Tipo Polo 1
7.
Adv Sci (Weinh) ; 7(15): 2001129, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32775170

RESUMEN

The analysis of nanoparticles' biocompatibility and immunogenicity is mostly performed under a healthy condition. However, more clinically relevant evaluation conducted under pathological condition is less known. Here, the immunogenicity and bio-nano interactions of porous silicon nanoparticles (PSi NPs) are evaluated in an acute liver inflammation mice model. Interestingly, a new mechanism in which PSi NPs can remit the hepatocellular damage and inflammation activation in a surface dependent manner through protein corona formation, which perturbs the inflammation by capturing the pro-inflammatory signaling proteins that are inordinately excreted or exposed under pathological condition, is found. This signal sequestration further attenuates the nuclear factor κB pathway activation and cytokines production from macrophages. Hence, the study proposes a potential mechanism for elucidating the altered immunogenicity of nanomaterials under pathological conditions, which might further offer insights to establish harmonized standards for assessing the biosafety of biomaterials in a disease-specific or personalized manner.

8.
FEBS Lett ; 594(14): 2294-2302, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32412649

RESUMEN

Receptor-interacting serine/threonine-protein kinase 3 (RIPK3) is a central protein in necroptosis with great potential as a target for treating necroptosis-associated diseases, such as Crohn's disease. However, blockade of RIPK3 kinase activity leads to unexpected RIPK3-initiated apoptosis. Herein, we found that PP2, a known SRC inhibitor, inhibits TNF-α-induced necroptosis without initiating apoptosis. Further investigation showed that PP2 acts as an inhibitor of not only SRC but also RIPK3. PP2 does not disturb the integrity of the RIPK1-RIPK3-mixed lineage kinase domain-like pseudokinase (MLKL) necroptosome or the autophosphorylation of RIPK3 at T231/S232 but disrupts RIPK3 oligomerization, thereby impairing the phosphorylation and oligomerization of MLKL. These results demonstrate the essential role of RIPK3 oligomerization in necroptosis and suggest a potential RIPK3 oligomerization-targeting strategy for therapeutic development.


Asunto(s)
Apoptosis , Necroptosis/efectos de los fármacos , Multimerización de Proteína/efectos de los fármacos , Pirimidinas/farmacología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Ratones , Fosforilación/efectos de los fármacos , Factor de Necrosis Tumoral alfa/farmacología , Familia-src Quinasas/antagonistas & inhibidores
9.
Br J Pharmacol ; 176(23): 4491-4509, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31364164

RESUMEN

BACKGROUND AND PURPOSE: Bruton's tyrosine kinase (BTK) plays a key role in B-cell receptor signalling by regulating cell proliferation and survival in various B-cell malignancies. Covalent low-MW BTK kinase inhibitors have shown impressive clinical efficacy in B-cell malignancies. However, the mutant BtkC481S poses a major challenge in the management of B-cell malignancies by disrupting the formation of the covalent bond between BTK and irreversible inhibitors, such as ibrutinib. The present studies were designed to develop novel BTK inhibitors targeting ibrutinib-resistant BtkC481S mutation. EXPERIMENTAL APPROACH: BTK-Ba/F3, BTK(C481S)-Ba/F3 cells, and human malignant B-cells JeKo-1, Ramos, and NALM-6 were used to evaluate cellular potency of BTK inhibitors. The in vitro pharmacological efficacy and compound selectivity were assayed via cell viability, colony formation, and BTK-mediated signalling. A tumour xenograft model with BTK-Ba/F3, Ramos and BTK(C481S)-Ba/F3 cells in Nu/nu BALB/c mice was used to assess in vivo efficacy of XMU-MP-3. KEY RESULTS: XMU-MP-3 is one of a group of low MW compounds that are potent non-covalent BTK inhibitors. XMU-MP-3 inhibited both BTK and the acquired mutant BTKC481S, in vitro and in vivo. Further computational modelling, site-directed mutagenesis analysis, and structure-activity relationships studies indicated that XMU-MP-3 displayed a typical Type-II inhibitor binding mode. CONCLUSION AND IMPLICATIONS: XMU-MP-3 directly targets the BTK signalling pathway in B-cell lymphoma. These findings establish XMU-MP-3 as a novel inhibitor of BTK, which could serve as both a tool compound and a lead for further drug development in BTK relevant B-cell malignancies, especially those with the acquired ibrutinib-resistant C481S mutation.


Asunto(s)
Agammaglobulinemia Tirosina Quinasa/antagonistas & inhibidores , Antineoplásicos/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirazoles/farmacología , Pirimidinas/farmacología , Adenina/análogos & derivados , Agammaglobulinemia Tirosina Quinasa/genética , Agammaglobulinemia Tirosina Quinasa/metabolismo , Animales , Antineoplásicos/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Células HEK293 , Humanos , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Simulación del Acoplamiento Molecular , Estructura Molecular , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Piperidinas , Inhibidores de Proteínas Quinasas/química , Pirazoles/química , Pirimidinas/química , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad
10.
Biomaterials ; 185: 322-332, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30267958

RESUMEN

Orally administrable drug delivery vehicles are developed to manage incurable inflammatory bowel disease (IBD), however, their therapeutic outcomes are compromised by the side effects of systemic drug exposure. Herein, we use hyaluronic acid functionalized porous silicon nanoparticle to bridge enzyme-responsive hydrogel and pH-responsive polymer, generating a hierarchical structured (nano-in-nano-in-micro) vehicle with programmed properties to fully and sequentially overcome the multiple obstacles for efficiently delivering drugs locally to inflamed sites of intestine. After oral administration, the pH-responsive matrix protects the embedded hybrid nanoparticles containing drug loaded hydrogels against the spatially variable physiological environments of the gastrointestinal tract until they reach the inflamed sites of intestine, preventing premature drug release. The negatively charged hybrid nanoparticles selectively target the inflamed sites of intestine, and gradually release drug in response to the microenvironment of inflamed intestine. Overall, the developed hierarchical structured and programmed vehicles load, protect, transport and release drugs locally to inflamed sites of intestine, contributing to superior therapeutic outcomes. Such strategy could also inspire the development of numerous hierarchical structured vehicles by other porous nanoparticles and stimuli-responsive materials for the local delivery of various drugs to treat plenty of inflammatory gastrointestinal diseases, including IBD, gastrointestinal cancers and viral infections.


Asunto(s)
Antiinflamatorios/administración & dosificación , Budesonida/administración & dosificación , Preparaciones de Acción Retardada/química , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Intestinos/efectos de los fármacos , Silicio/química , Administración Oral , Animales , Antiinflamatorios/farmacocinética , Antiinflamatorios/uso terapéutico , Budesonida/farmacocinética , Budesonida/uso terapéutico , Línea Celular , Sistemas de Liberación de Medicamentos , Humanos , Ácido Hialurónico/análogos & derivados , Concentración de Iones de Hidrógeno , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Intestinos/inmunología , Intestinos/patología , Masculino , Ratones Endogámicos C57BL , Nanopartículas/química , Polímeros/química , Porosidad
11.
J Med Chem ; 61(12): 5424-5434, 2018 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-29878764

RESUMEN

Methuosis is a novel nonapoptotic mode of cell death characterized by vacuole accumulation in the cytoplasm. In this article, we describe a series of azaindole-based compounds that cause vacuolization in MDA-MB-231 cells. The most potent vacuole inducer, compound 13 (compound 13), displayed differential cytotoxicities against a broad panel of cancer cell lines, such as MDA-MB-231, A375, HCT116, and MCF-7, but it did not inhibit the growth of the nontumorigenic epithelial cell line MCF-10A. A mechanism study confirmed that the cell death was caused by inducing methuosis. Furthermore, compound 13 exhibited substantial pharmacological efficacy in the suppression of tumor growth in a xenograft mouse model of MDA-MB-231 cells without apparent side effects, which makes this compound the first example of a methuosis inducer with potent in vivo efficacy. These results demonstrate that methuosis inducers might serve as novel therapeutics for the treatment of cancer.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Antineoplásicos/farmacocinética , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Semivida , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Bibliotecas de Moléculas Pequeñas/química , Vacuolas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Eur J Pharmacol ; 822: 51-58, 2018 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-29355554

RESUMEN

Thrombin has long been suggested as a desirable antithrombotic target, but anti-thrombin therapy without anti-platelet thereby has never achieved the ideal effect. HY023016 is a novel compound, in our previous study, it exerted better anti-thrombotic than dabigatran etexilate. The present study aims to illustrate the excess anti-thrombotic molecular mechanisms of HY023016 through thrombin anion exosites and the platelet membrane receptor subunit glycoprotein Ibα (GPIbα). HY023016 strongly inhibited the conversion of fibrinogen to fibrous may via blocking thrombin exosite I. We also discovered that HY023016 remarkably inhibited exosite II by a loss of affinity for the γ'-peptide of fibrinogen and for heparin. Furthermore, a solid phase binding assay revealed that HY023016 inhibited ristocetin-induced washed platelets bind to von Willebrand factor (vWF). In GST pull-down assay, HY023016 decreased the binding of recombinant vWF-A1 to GPIbα N-terminal. Thus, HY023016 provides an innovative idea for designing multi-targeted anti-thrombotic drugs and laying a scientific foundation for reducing "total thrombosis risk" in a clinical drug treatment.


Asunto(s)
Dabigatrán/farmacología , Fibrinolíticos/farmacología , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Trombina/química , Trombina/metabolismo , Sitios de Unión , Humanos , Complejo GPIb-IX de Glicoproteína Plaquetaria/química , Glicoproteínas de Membrana Plaquetaria/metabolismo , Unión Proteica/efectos de los fármacos , Dominios Proteicos
13.
Adv Mater ; 30(24): e1703393, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29024054

RESUMEN

Herein, a novel nanohybrid based on porous silicon, gold nanoparticles (Au NPs), and acetalated dextran (DPSi/DAu@AcDEX) is reported to encapsulate and deliver one drug and increase the computer tomography (CT) signal for acute-liver-failure (ALF) theranostics. A microfluidic-assisted method is used to co-encapsulate different NPs in a single step. By alternating the surface properties of different NPs and by modulating the composition of the organic phase, both PSi and Au NPs are effectively encapsulated into the polymer matrix simultaneously, thus further achieving a multifunctional application. This system can be used to identify pathologically changes in the tissues and selectively deliver drugs to these sites. The loading of a therapeutic compound (XMU-MP-1) improves the drug solubility, precise, in situ drug delivery, and the drug-functioning time. In vivo results confirm a superior treatment effect and better compliance of this newly developed nanoformulation than free compound. This nanosystem plays a crucial role in targeting the lesion area, thus increasing the local drug concentration important for ALF reverse-effect. Moreover, the residence of Au NPs within the matrix further endows our system for CT-imaging. Altogether, these results support that this nanohybrid is a potential theranostic platform for ALF.


Asunto(s)
Nanopartículas , Dextranos , Oro , Humanos , Fallo Hepático Agudo , Regeneración Hepática , Silicio , Nanomedicina Teranóstica
14.
Cancer Res ; 77(1): 175-186, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27758886

RESUMEN

Approximately 80% of breast cancers overexpress the kinase breast tumor kinase (BRK)/protein tyrosine kinase 6, which has various oncogenic roles in breast cancer cell proliferation, survival, and migration. However, BRK inhibitors have yet to be explored as possible therapeutic tools. In this study, we used a parallel compound-centric approach to discover a new class of pharmaceutical agents, exemplified by XMU-MP-2, as potent and selective BRK inhibitors. XMU-MP-2 exhibited target-specific inhibition of BRK kinase activity and disrupted signaling pathways mediated by this activity, thereby reducing proliferation in BRK-positive breast cancer cells. In mouse xenograft models, XMU-MP-2 repressed the growth of tumors driven by oncogenic BRK, including BRK-transformed Ba/F3 cells and BRK-positive breast cancer cells. Notably, XMU-MP-2 cooperated strongly with HER2 inhibitor or ER blockade to block breast cancer cell proliferation in vitro and in vivo Overall, our findings offer a preclinical proof of concept for therapeutic targeting of the BRK kinase in breast cancer. Cancer Res; 77(1); 175-86. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Animales , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , Transcriptoma , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Sci Transl Med ; 8(352): 352ra108, 2016 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-27535619

RESUMEN

Tissue repair and regenerative medicine address the important medical needs to replace damaged tissue with functional tissue. Most regenerative medicine strategies have focused on delivering biomaterials and cells, yet there is the untapped potential for drug-induced regeneration with good specificity and safety profiles. The Hippo pathway is a key regulator of organ size and regeneration by inhibiting cell proliferation and promoting apoptosis. Kinases MST1 and MST2 (MST1/2), the mammalian Hippo orthologs, are central components of this pathway and are, therefore, strong target candidates for pharmacologically induced tissue regeneration. We report the discovery of a reversible and selective MST1/2 inhibitor, 4-((5,10-dimethyl-6-oxo-6,10-dihydro-5H-pyrimido[5,4-b]thieno[3,2-e][1,4]diazepin-2-yl)amino)benzenesulfonamide (XMU-MP-1), using an enzyme-linked immunosorbent assay-based high-throughput biochemical assay. The cocrystal structure and the structure-activity relationship confirmed that XMU-MP-1 is on-target to MST1/2. XMU-MP-1 blocked MST1/2 kinase activities, thereby activating the downstream effector Yes-associated protein and promoting cell growth. XMU-MP-1 displayed excellent in vivo pharmacokinetics and was able to augment mouse intestinal repair, as well as liver repair and regeneration, in both acute and chronic liver injury mouse models at a dose of 1 to 3 mg/kg via intraperitoneal injection. XMU-MP-1 treatment exhibited substantially greater repopulation rate of human hepatocytes in the Fah-deficient mouse model than in the vehicle-treated control, indicating that XMU-MP-1 treatment might facilitate human liver regeneration. Thus, the pharmacological modulation of MST1/2 kinase activities provides a novel approach to potentiate tissue repair and regeneration, with XMU-MP-1 as the first lead for the development of targeted regenerative therapeutics.


Asunto(s)
Factor de Crecimiento de Hepatocito/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Regeneración/efectos de los fármacos , Sulfonamidas/farmacología , Acetaminofén/toxicidad , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Colitis/inducido químicamente , Colitis/prevención & control , Cristalización , Factor de Crecimiento de Hepatocito/química , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/trasplante , Ensayos Analíticos de Alto Rendimiento , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lesión Pulmonar/tratamiento farmacológico , Ratones , Modelos Moleculares , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/química , Medicina Regenerativa , Serina-Treonina Quinasa 3 , Transducción de Señal/efectos de los fármacos , Sulfonamidas/química , Investigación Biomédica Traslacional
16.
Eur J Pharmacol ; 781: 190-7, 2016 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-27085896

RESUMEN

The generation of thrombus is not considered as an isolated progression without other pathologic processes, which may also enhance procoagulant state. The purpose of this study was to assess whether HY023016, a novel dabigatran prodrug and an oral direct thrombin inhibitor, or dabigatran etexilate, another thrombin inhibitor can improve the state of whole blood hypercoagulability in vitro/vivo. By using whole blood flow cytometry we explored the effects of HY023016 and dabigatran etexilate on thrombin and ADP-induced human platelet-leukocyte aggregation generated in vitro. With the method of continuous infusion of thrombin intravenous, we successfully established a rat hypercoagulable model and evaluated the effect of HY023016 or dabigatran etexilate in vivo. HY023016 was able to inhibit thrombin- or ADP-induced platelet P-selectin or CD40L expression, leukocyte CD11b expression and formation of platelet-leukocyte aggregates in dose-dependent manner. Dabigatran etexilate was unable to affect ADP-induced platelet P-selectin or CD40L expression, leukocyte CD11b expression and formation of platelet-leukocyte aggregates. Based on rat hypercoagulable model, dabigatran etexilate could reverse thrombin-induced circulatory system hypercoagulable state in a concentration-dependent manner. Dabigatran etexilate also inhibited electrical stimulation induced formation of arterial thrombus in rat under hypercoagulable state, and extracorporal circulation-induced formation of thrombus in dose-dependent manner. Compared with dabigatran etexilate, HY023016 showed nearly equal or even better antithrombotic activity, regardless of reversing the cycle of rat hypercoagulable state or inhibiting platelet-leukocyte aggregation. In surrmary, HY023016 could effectively improve hypercoagulable state of circulatory system.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Fibrinolíticos/farmacología , Animales , Antígeno CD11b/metabolismo , Modelos Animales de Enfermedad , Fibrinolíticos/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Nocicepción/efectos de los fármacos , Activación Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Ratas , Trombina/administración & dosificación , Trombina/farmacología , Trombosis/tratamiento farmacológico , Trombosis/fisiopatología
17.
Neurosci Lett ; 588: 154-8, 2015 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-25545553

RESUMEN

Evodiamine is a major alkaloid compound extracted from the dry unripened fruit Evodia fructus (Evodia rutaecarpa Benth., Rutaceae), which has a variety of pharmacological activities. The present study aims to determine the antidepressant-like effect of evodiamine in a rat model of chronic unpredictable mild stress (CUMS). We identified that evodiamine could reverse the following CUMS-induced behavioural deficits and biochemical changes in rats: the decreases of sucrose preference, number of crossings, 5-HT and NA levels, as well as the increase of immobility time. Evodiamine treatments also ameliorated the corticosterone hypersecretion induced by CUMS. Furthermore, we found that evodiamine was able to up-regulate the expression of brain-derived neurotrophic factor (BDNF) and phosphorylated tropomyosin-related kinase B (pTrkB) without altering TrkB. This study suggests potential antidepressant-like effect of evodiamine on CUMS rats, and its underlying mechanisms can be potentially linked to their modulating effects on the monoamine transmitters and BDNF-TrkB signaling in the hippocampus.


Asunto(s)
Antidepresivos/uso terapéutico , Quinazolinas/uso terapéutico , Estrés Psicológico/tratamiento farmacológico , Animales , Antidepresivos/farmacología , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Enfermedad Crónica , Corticosterona/sangre , Hipocampo/metabolismo , Masculino , Norepinefrina/metabolismo , Fosforilación , Quinazolinas/farmacología , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Receptor trkB/metabolismo , Serotonina/metabolismo , Estrés Psicológico/metabolismo
18.
Environ Toxicol Pharmacol ; 36(2): 288-295, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23686009

RESUMEN

This study aimed to investigate the effects of galangin on liver fibrosis in rats induced by subcutaneous injection of carbon tetrachloride (CCl4). The administration of CCl4 to rats for 12 weeks caused significant increase of hyaluronic acid, laminin, alanine transaminase, aspartate transaminase and decrease of total protein, albumin in serum, while the influences could be reversed by galangin. Galangin markedly reduced hepatic malondialdehyde, hydroxyproline concentration, increased activities of liver superoxide dismutase, glutathione peroxidase compared with CCl4-treated rats. Histological results indicated that galangin alleviated liver damage. In addition, treatment with galangin significantly down-regulated expressions of α-smooth muscle actin and transforming growth factor ß1. These results suggest galangin can inhibit liver fibrosis induced by CCl4 in rats, which was probably associated with its effect on removing oxygen free radicals, decreasing lipid peroxidation, as well as inhibiting hepatic stellate cells activation and proliferation.


Asunto(s)
Flavonoides/farmacología , Cirrosis Hepática Experimental/prevención & control , Hígado/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Tetracloruro de Carbono , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Células Estrelladas Hepáticas/efectos de los fármacos , Células Estrelladas Hepáticas/metabolismo , Células Estrelladas Hepáticas/patología , Peroxidación de Lípido/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática Experimental/inducido químicamente , Cirrosis Hepática Experimental/metabolismo , Cirrosis Hepática Experimental/patología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
19.
Thromb Res ; 131(5): 425-35, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23535565

RESUMEN

INTRODUCTION: Thrombin is a multifunctional trypsin-like serine protease that plays key roles in coagulation and thrombogenesis. HY023016, a novel Dabigatran prodrug, is an oral direct thrombin inhibitor. The purpose of this study was to compare the anti-thrombotic activities and haemorrhagic effects of HY023016 with Dabigatran etexilate and tetramethylpyrazine in several animal thrombosis models. METHODS: To investigate drug exposure, liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was used to determine the pharmacokinetic profile of HY023016. After single intragastric administrations of HY023016, Dabigatran etexilate or tetramethylpyrazine, the anti-thrombotic activities were evaluated through rabbit jugular vein thrombosis model, rat inferior vena cava thrombosis model, ex vivo rabbit platelet aggregation assay, in vivo rabbit coagulation assay, and direct thrombin binding assay. Meanwhile, we evaluated the effect of HY023016 on expression of tissue factor (TF) by RT-PCR. Rabbit cuticle bleeding assay and mouse tail bleeding assay were applied to evaluate the effects of HY023016 on haemorrhage. RESULTS: Pharmacokinetic parameters indicated that HY023016 can convert to Dabigatran and tetramethylpyrazine. Our studies showed that HY023016 was able to significantly inhibit thrombus formation in a dose-dependent manner in rabbit and rat models (P<0.05). Similarly, it was able to dose-dependently inhibit thrombin- or ADP-induced platelet aggregation, prolonging the activated partial thromboplastin time (APTT) and prothrombin time (PT), inhibiting the activity of thrombin and inhibiting thrombin- or ADP-induced expression of TF (P<0.05 or 0.01). Dabigatran etexilate was also able to dose-dependently and significantly inhibit thrombus formation (P<0.01) but was unable to affect ADP-induced platelet aggregation and expression of TF. In contrast, tetramethylpyrazine could only exhibit mild antithrombotic activity compared with HY023016 and Dabigatran etexilate (P<0.05). HY023016 could prolong bleeding time (P<0.001), but the prolongations were significantly less than Dabigatran etexilate (P<0.05). CONCLUSION: HY023016 showed thrombosis-inhibition activities comparable to those of Dabigatran etexilate, but better than those of tetramethylpyrazine. The attendant bleeding risk of HY023016 was lower than Dabigatran etexilate in rabbits and mice.


Asunto(s)
Antitrombinas/farmacología , Bencimidazoles/farmacología , Profármacos/farmacología , Trombosis/tratamiento farmacológico , Trombosis/metabolismo , beta-Alanina/análogos & derivados , Animales , Antitrombinas/sangre , Antitrombinas/farmacocinética , Bencimidazoles/sangre , Bencimidazoles/farmacocinética , Dabigatrán , Modelos Animales de Enfermedad , Ratones , Profármacos/farmacocinética , ARN Mensajero/sangre , ARN Mensajero/genética , Conejos , Ratas , Trombina/metabolismo , Tromboplastina/biosíntesis , Tromboplastina/genética , Trombosis/sangre , beta-Alanina/sangre , beta-Alanina/farmacocinética , beta-Alanina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA