Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Phytother Res ; 38(1): 131-146, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37821355

RESUMEN

Neuroblastoma and glioblastoma are primary malignant tumors of the nervous system, with frequent relapse and limited clinical therapeutic drugs. The failure of their treatment is due to the tumor cells exhibiting cancer stem-like cells (CSLCs) properties. Octamer binding transcription factor 4 (Oct4) is involved in mediating CSLCs, our previous work found that Oct4-driven reprogramming of astrocytes into induced neural stem cells was potentiated with continuous sonic hedgehog (Shh) stimulation. In this study, we aimed to study the importance of Oct4 and Shh combination in the stemness properties induction of neuroblastoma and glioblastoma cells, and evaluate the anti-stemness effect of dauricine (DAU), a natural product of bis-benzylisoquinoline alkaloid. The effect of Oct4 and Shh co-activation on cancer stemness was evaluated by tumor spheres formation model and flow cytometry analysis. Then the effects of DAU on SH-SY5Y and T98-G cells were assessed by the MTT, colony formation, and tumor spheres formation model. DAU acts on Oct4 were verified using the Western blotting, MTT, and so on. Mechanistic studies were explored by siRNA transfection assay, Western blotting, and flow cytometry analysis. We identified that Shh effectively improved Oct4-mediated generation of stemness in SH-SY5Y and T98-G cells, and Oct4 and Shh co-activation promoted cell growth, the resistance of apoptosis. In addition, DAU, a natural product, was found to be able to attenuate Oct4/Shh co-activated stemness and induce cell cycle arrest and apoptosis via blocking AKT/ß-catenin signaling in neuroblastoma and glioblastoma, which contributed to the neuroblastoma and glioblastoma cells growth inhibition by DAU. In summary, our results indicated that the treatment of DAU may be served as a potential therapeutic method in neuroblastoma and glioblastoma.


Asunto(s)
Bencilisoquinolinas , Productos Biológicos , Glioblastoma , Neuroblastoma , Tetrahidroisoquinolinas , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Proteínas Hedgehog/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , beta Catenina/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Línea Celular Tumoral , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/metabolismo , Neuroblastoma/patología , Recurrencia Local de Neoplasia/metabolismo , Recurrencia Local de Neoplasia/patología , Bencilisoquinolinas/farmacología , Células Madre Neoplásicas , Proliferación Celular , Apoptosis , Productos Biológicos/farmacología
2.
J Transl Med ; 21(1): 597, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37670360

RESUMEN

BACKGROUND: Triple negative breast cancer (TNBC) is a major subtype of breast cancer, with limited therapeutic drugs in clinical. Epidermal growth factor receptor (EGFR) is reported to be overexpressed in various TNBC cells. Cantharidin is an effective ingredient in many clinical traditional Chinese medicine preparations, such as Delisheng injection, Aidi injection, Disodium cantharidinate and vitamin B6 injection. Previous studies showed that cantharidin had satisfactory pharmacological activity on a variety of tumors. In this study, we aimed to study the therapeutic potential of cantharidin for TNBC treatment by targeting EGFR, and expound its novel regulator miR-607. METHODS: The effect of cantharidin on breast cancer in vivo was evaluated by 4T1 mice model. Then the effects of cantharidin on TNBC cells was assessed by the MTT, colony formation, and AnnexinV-PE/7AAD staining. Cantharidin acts on EGFR were verified using the cell membrane chromatography, RT-PCR, Western blotting, MTT, and so on. Mechanistic studies were explored by dual-luciferase report assay, RT-PCR, western blotting, and immunofluorescence staining assay. RESULTS: Cantharidin inhibited TNBC cell growth and induce apoptosis by targeting EGFR. miR-607 was a novel EGFR regulator and exhibited suppressive functions on TNBC cell behaviors. Mechanistic study showed that cantharidin blocked the downstream PI3K/AKT/mTOR and ERK/MAPK signaling pathway. CONCLUSION: Our results revealed that cantharidin may be served as a potential candidate for TNBC treatment by miR-607-mediated downregulation of EGFR.


Asunto(s)
MicroARNs , Neoplasias de la Mama Triple Negativas , Animales , Ratones , Humanos , Cantaridina , Regulación hacia Abajo , Fosfatidilinositol 3-Quinasas , Receptores ErbB , Apoptosis
3.
Phytother Res ; 35(8): 4567-4578, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34058790

RESUMEN

Regorafenib (RGF), a second-line multi-kinase inhibitor in the treatment of HCC (hepatocellular carcinoma) after sorafenib failure, exposes to the risk of drug resistance and subsequent progression of HCC patients. Toosendanin (TSN), a triterpenoid has presented excellent inhibition on several tumors. The purpose of this study is to investigate the inhibitory effect of the combination of TSN and RGF on HCC cells. We identified that TSN and RGF combination (TRC) synergistically inhibited the proliferation and migration of MHCC-97L cells. The upregulation of WWOX (WW-domain containing oxidoreductase) played a vital role in the HCC cell growth treated with TRC. TRC suppressed the phosphorylation of Stat3 and expression of DVL2, negatively regulated the activity of ß-catenin by promoting the phosphorylation of GSK3ß. In addition, the intranuclear proteins, including MMP2, MMP9, and C-MYC were significantly inhibited by TRC. The in vivo xenograft models confirmed that TRC effectually prevented the tumor growth through upregulating WWOX. Therefore, the treatment of TRC may be a potential solution of RGF resistance and promising therapeutic method in malignant HCC.


Asunto(s)
Carcinoma Hepatocelular , Medicamentos Herbarios Chinos/farmacología , Neoplasias Hepáticas , Compuestos de Fenilurea/farmacología , Piridinas/farmacología , Proteínas Supresoras de Tumor/metabolismo , Oxidorreductasa que Contiene Dominios WW/metabolismo , Animales , Apoptosis , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Glucógeno Sintasa Quinasa 3 , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Chemistry ; 26(57): 13031-13038, 2020 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-32428362

RESUMEN

Theranostic agents, taking the advantages of both imaging and therapeutic functions, are anticipated to be key components in the development of personalized medicine in which the therapeutic response can be real-time monitored. Herein, three metallacycles with pendent adamantane groups are prepared by coordination-driven self-assembly of PtII ligands with anticancer activities and tetraphenylethylene derivatives with emission. ß-Cyclodextrin, which shows good host-guest interactions with adamantane moieties, was added to form amphiphilic supramolecular nanoparticles with the aim to enhance the aqueous solubilities and bioactivities of these metallacycles. Moreover, when rhodamine-modified ß-cyclodextrin was used as the carrier, the release of the metallacycles from the nanoparticles could be monitored in situ through the fluorescence changes owing to the efficient fluorescence resonance energy transfer from the metallacycles to rhodamine-modified ß-cyclodextrin. In vitro and in vivo studies showed that these nanoparticles not only served as cell imaging contrast agents but also displayed improved anticancer activities, allowing them to serve as potential candidates for cancer theranostics. This study provides a simple and efficient method to prepare theranostic agents by hierarchical supramolecular self-assembly, which will pave the way for image-guided cancer therapy, targeted cancer therapy, and related biomedical fields.


Asunto(s)
Nanopartículas , Neoplasias , Humanos , Neoplasias/tratamiento farmacológico , Medicina de Precisión , Rodaminas , beta-Ciclodextrinas
5.
Pharmacol Res ; 158: 104868, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32407961

RESUMEN

Hepatocellular carcinoma (HCC) is a lethal malignancy with limited treatment options. The tyrosine kinase receptor EphB4 promotes oncogenesis and tumor development and progression. Its inhibition is regarded as an effective strategy for the treatment of solid tumors. In the present study, we identified cantharidin as a novel EphB4 inhibitor for HCC treatment and evaluated the underlying molecular pharmacological mechanisms of action. We observed increased expression levels of EphB4 in HCC patients and a positive correlation between EphB4 and p-JAK2 levels in HCC patient samples. Knockdown of EphB4 using small interfering RNA decreased the expression levels of p-JAK2 and p-STAT3 in HCC cells, suggesting JAK2/STAT3 being a novel downstream signaling target of EphB4. Cell viability experiments revealed that the anti-cancer effect of cantharidin was positively correlated with EphB4 expression levels in HCC cell lines. We confirmed the potent antiproliferative activity of cantharidin on HepG2 cells with high expression of EphB4 and tumor xenograft. Molecular docking assay, immunoblotting assay and quantitative reverse transcription PCR assay indicated that cantharidin bound to EphB4, and thereby resulted in EphB4 suppression at mRNA and protein levels. Hep3B and SMMC-7721 cells were with low expression of EphB4. In EphB4-/HepG2, EphB4+/HepG2, and EphB4+/Hep3B cells, EphB4 knockdown alleviated the cantharidin-induced decrease in cell viability and colony formation ability and increase in apoptosis in HepG2 cells, while its overexpression exacerbated these effects in Hep3B cells and increased the apoptosis of HepG2 cells. In nude mouse models, cantharidin suppressed tumor growth more effectively in EphB4+/SMMC-7721 xenografts than in wild-type SMMC-7721 xenografts. Underlying mechanistic study showed that by targeting EphB4, cantharidin blocked a novel target, the downstream JAK2/STAT3 pathway, and the previously known target, the PI3K/Akt signaling, resulting in intrinsic apoptosis. These results indicated that cantharidin may be a potential candidate for HCC treatment by regulating the EphB4 signaling pathway.


Asunto(s)
Cantaridina/metabolismo , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/metabolismo , Receptor EphB4/antagonistas & inhibidores , Receptor EphB4/metabolismo , Animales , Cantaridina/farmacología , Cantaridina/uso terapéutico , Carcinoma Hepatocelular/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Células Hep G2 , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Neoplasias Hepáticas/tratamiento farmacológico , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor EphB4/química , Factor de Transcripción STAT3/antagonistas & inhibidores , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Resultado del Tratamiento , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
6.
Langmuir ; 35(24): 8045-8051, 2019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31140813

RESUMEN

The realization of multistage-controlled drug delivery at the cell level through the morphology transitions of supramolecular self-assemblies (SSA) is still a challenge. Herein, successive morphology transitions of SSA with pH responsiveness were successfully achieved through the subsequent action of ultrasound and redox stimuli. Specifically, we first prepared noncovalently PEGylated spherical self-assemblies formed by host-guest-conjugated amphiphilic ß-CD dimers. The functionalized PEG could be associated/disassociated onto the spherical self-assemblies by adjusting pH values of solutions. They could reassemble into branched self-assemblies induced by ultrasonication. Such branched self-assemblies could be further dissociated into second spherical self-assemblies under a redox stimulus. This morphology transition process was used to conduct triple-controlled targeted drug delivery and release in cancer cells. This work will be beneficial for the design of smart SSA for controlled release in vivo.

7.
Phytother Res ; 33(3): 745-755, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30565332

RESUMEN

Hepatocellular carcinoma (HCC) is a biologically complex disease. Combination chemotherapy is a good strategy after surgery treatment. In this study, we report that berberine combined with HMQ1611 (BCH) had a good synergistic effect on the HCC. Our findings concluded that BCH showed good inhibition on the HCC proliferation and colony formation, which attributed to cell cycle arrest by BCH at G1 phase through impairing the expression of cyclinD1, cyclinE, and cdc2 and downregulated the phosphorylation of Akt, mTOR, and ERK. Moreover, BCH negatively regulated Wnt signaling pathway by upregulating the Axin and inhibiting the nuclear translocation of ß-catenin. BCH suppressed the phosphorylation of LRP5/6, GSK3ß, the expression of Wnt5a, Frizzled8, CK1, and APC, as well as the nucleus protein included MMP2, MMP3, MMP9, and c-myc. The above data of Wnt signaling regulators contributed to inhibition by BCH on cell migration. In vivo studies, BCH significantly suppressed the growth of SMMC-7721 xenograft tumors through downregulating Ki67 and ß-catenin, as well as upregulating Axin and p-ß-catenin. In conclusion, the results revealed that BCH exhibited potential antitumor activities against human liver cancer in vitro and in vivo, and the potential mechanism underlying these activities depended on the inhibition of the Wnt/ß-catenin signaling pathway.


Asunto(s)
Acetanilidas/farmacología , Benzamidas/farmacología , Berberina/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Vía de Señalización Wnt/efectos de los fármacos , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos BALB C , beta Catenina/fisiología
8.
Mikrochim Acta ; 186(7): 407, 2019 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-31183562

RESUMEN

This paper presents a method for the preparation of a graphene-based hybrid composite film by electrodeposition of reduced graphene oxide and overoxidized electropolymerized polypyrrole onto a glassy carbon electrode (GCE) using cyclic voltammetry. The morphology of the hybrid composite film was characterized by scanning electron microscopy. The electrochemical activity of the modified GCE was studied by cyclic voltammetry using the negatively charged redox probe Fe(CN)63- and the positively charged redox probe Ru(NH3)63+. The modified GCE displays excellent electrocatalytic activity for dopamine (DA) and uric acid (UA), but electrostatically repulses ascorbate anion under physiological pH conditions. The voltammetric response to DA is linear in the 2.0 µM to 160 µM concentration range even in the presence of 1.0 mM ascorbic acid and 0.1 mM of UA. The detection limit is 0.5 µM. The amperometric response to DA (best measured at 0.22 V vs. Ag/AgCl) extends from 0.4 µM to 517 µM and has a 0.2 µM detection limit. Graphical abstract Schematic presentation of the fabrication of a glassy carbon electrode modified with reduced graphene oxide and overoxidized electropolymerized polypyrrole, and its application to the determination of dopamine in the presence of ascorbic acid and uric acid.

9.
J Cell Mol Med ; 22(5): 2955-2959, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29516635

RESUMEN

Looking for novel, effective and less toxic therapies for cervical cancer is of significant importance. In this study, we reported that HMQ-T-F2(F2) significantly inhibited cell proliferation and transplantable tumour growth. Mechanistically, HMQ-T-F2 inhibited HeLa cell growth through repressing the expression and nuclear translocation of ß-catenin, enhancing Axin expression, as well as downregulating the Wnt downstream targeted proteins. Knock-down of a checkpoint ß-catenin by siRNA significantly attenuated HeLa cell proliferation. Furthermore, XAV939, an inhibitor of ß-catenin, was used to treat HeLa cells and the results demonstrated that HMQ-T-F2 inhibited proliferation and migration via the inhibition of the Wnt/ß-catenin pathway.


Asunto(s)
Tiourea/farmacología , Regulación hacia Arriba/efectos de los fármacos , Neoplasias del Cuello Uterino/genética , Animales , Proteína Axina/genética , Proteína Axina/metabolismo , Proliferación Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Ratones , Activación Transcripcional/genética , Neoplasias del Cuello Uterino/patología , beta Catenina/metabolismo
10.
J Cell Mol Med ; 22(11): 5231-5243, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30589500

RESUMEN

Hepatocellular carcinoma (HCC) is a highly prevalent cancer worldwide and it is necessary to discover and develop novel preventive strategies and therapeutic approaches for HCC. Herein, we report that EphrinB2 expression is correlated with liver cancer progression. Moreover, by using phosphorylated proteomics array, we reveal a pro-apoptosis protein whose phosphorylation and activation levels are up-regulated upon EphrinB2 knockdown. These results suggest that EphrinB2 may act as an anti-apoptotic protein in liver cancer cells. We also explored the therapeutic potential of HMQ-T-B10 (B10), which was designed and synthesized in our laboratory, for HCC and its underlying mechanisms in vitro and in vivo. Our data demonstrate that B10 could bind EphrinB2 and show inhibitory activity on human liver cancer cells. Moreover, induction of human liver cancer cell apoptosis by B10 could be augmented upon EphrinB2 knockdown. B10 inhibited HCC cell growth and induced HCC cell apoptosis by repressing the EphrinB2 and VEGFR2 signalling pathway. Growth of xenograft tumours derived from Hep3B in nude mice was also significantly inhibited by B10. Collectively, these findings highlight the potential molecular mechanisms of B10 and its potential as an effective antitumour agent for HCC.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Efrina-B2/genética , Neoplasias Hepáticas/tratamiento farmacológico , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/síntesis química , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
J Mol Recognit ; 31(6): e2701, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29363191

RESUMEN

The binding property between a ligand and its receptor is very important for numerous biological processes. In this study, we developed a high epidermal growth factor receptor (EGFR)-expression cell membrane chromatography (CMC) method to investigate the binding characteristics between EGFR and the ligands gefitinib, erlotinib, canertinib, afatinib, and vandetanib. Competitive binding analysis using gefitinib as the marker was used to investigate the interactions that occurred at specific binding sites on EGFR. The ability of displacement was measured from the HEK293-EGFR/CMC column on the binding sites occupied by gefitinib for these ligands, which revealed the following order: gefitinib (KD, 8.49 ± 0.11 × 10-7  M) > erlotinib (KD, 1.07 ± 0.02 × 10-6  M) > canertinib (KD, 1.41 ± 0.07 × 10-6  M) > afatinib (KD, 1.80 ± 0.12 × 10-6  M) > vandetanib (KD, 1.99 ± 0.03 × 10-6  M). This order corresponded with the values estimated by frontal displacement analysis and the scores obtained with molecular docking. Furthermore, thermodynamic analysis indicated that the hydrogen bond or Van der Waals force was the main interaction force in the process of EGFR binding to all 5 ligands. Overall, these results demonstrate that a CMC method could be an effective tool to investigate the binding characteristics between ligands and receptors.


Asunto(s)
Membrana Celular/química , Unión Proteica/genética , Inhibidores de Proteínas Quinasas/química , Afatinib/química , Membrana Celular/genética , Cromatografía , Receptores ErbB/química , Receptores ErbB/genética , Clorhidrato de Erlotinib/química , Gefitinib/química , Células HEK293 , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Morfolinas/química , Piperidinas/química , Quinazolinas/química
12.
Phytother Res ; 31(9): 1392-1399, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28703366

RESUMEN

TPD7, a novel biphenyl urea taspine derivative, and berberine have presented inhibition on VEGFR2 that can be regulated by ephrin-B2 reverse signaling through interactions with the PDZ domain. The purpose of this study is to investigate the inhibitory effect of the combination of TPD7 and berberine (TAB) on T-cell acute lymphoblastic leukemia cell growth. TPD7 and berberine together synergistically inhibited the proliferation of Jurkat cells. Also, the combination of TAB induced G1 -phase cell-cycle arrest by downregulating the level of cyclin D1, cyclin E, and CDC2. Furthermore, the combination of TAB significantly enhanced apoptosis in Jurkat cells, and the apoptosis most likely resulted from the modulation of the level of Bcl-2 family members. Most importantly, the concomitant treatment simultaneously regulated the ephrin-B2 and VEGFR2 signaling, as well as modulated the MEK/ERK and PTEN/PI3K/AKT/mTOR signaling. Therefore, the combination treatment of TAB may be a promising therapeutic method in treating T-cell acute lymphoblastic leukemia. Copyright © 2017 John Wiley & Sons, Ltd.


Asunto(s)
Berberina/farmacología , Carbanilidas/farmacología , Efrina-B2/metabolismo , Hidroxilaminas/farmacología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Células Jurkat , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico
13.
Zhong Yao Cai ; 37(12): 2204-6, 2014 Dec.
Artículo en Zh | MEDLINE | ID: mdl-26080503

RESUMEN

OBJECTIVE: To investigate the secondary metabolites from Penicillium raistrickii. METHODS: Compounds were isolated and purified by normal and reverse phase silica gel, Sephadex LH-20 gel column chromatography and RP-HPLC. Their structures were established by means of spectral techniques and physicochemical properties. RESULTS: Twelve compounds were identified as pestafolide A(II), 3-methoxy-4-methyl-2,4-dien-pentanoic acid (2),p-hydroxy phenylacetamide (3),2-(2-hydroxy propanamido) benzamide (4), nicotinic acid (5), thymine (6), uracil (7) cyclo (Gly-Ala) (8), (22E,24R)-3ß,5α,9α-trihydroxy ergosta-7,22-diene-6-one (9), cerevisterol (10), ergosterol (11) and ergosterol peroxide (12). CONCLUSION: All compounds are isolated from Penicillium raistrickii for the first time.


Asunto(s)
Penicillium/química , Benzamidas/química , Benzamidas/aislamiento & purificación , Dipéptidos/química , Dipéptidos/aislamiento & purificación , Ergosterol/análogos & derivados , Ergosterol/química , Ergosterol/aislamiento & purificación , Niacina/química , Niacina/aislamiento & purificación , Fitosteroles/química , Fitosteroles/aislamiento & purificación , Metabolismo Secundario
14.
Toxicon ; 246: 107795, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-38849008

RESUMEN

Amphotericin B (AmB) induced liver and kidney injury is often responsible for hepatic and renal dysfunction. Therefore, the protection strategy on liver and renal functions in patients treated with AmB should be emphasized. In this paper, diammonium glycyrrhizinate (DG) and piperazine ferulate (PF) were taken as the research object to study its hepatoprotective and neuroprotective effect on AmB-induced liver and kidney damage in vitro and in vivo. The microplate method and ELISA kits were employed for the biochemical detection in the serum and urine of mice. Flow cytometric analysis and western blotting analysis were conducted to study the mechanism of DG and PF. Our results confirmed the prevention capacity of DG and PF on AmB-induced liver and kidney injury through the alleviation of pathological changes and enzyme reducing action. Furthermore, DG and PF suppressed ROS-mediated mitochondrial apoptosis in AmB-treated mice and cells through Caspase pathway and Caspase-independent AIF pathway. In summary, DG and PF could protect AmB-induced hepatotoxicity and nephrotoxicity by disrupting oxidative stress and apoptosis.


Asunto(s)
Anfotericina B , Apoptosis , Enfermedad Hepática Inducida por Sustancias y Drogas , Ácido Glicirrínico , Fármacos Neuroprotectores , Animales , Apoptosis/efectos de los fármacos , Ratones , Ácido Glicirrínico/farmacología , Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Anfotericina B/toxicidad , Masculino , Hígado/efectos de los fármacos , Hígado/patología , Riñón/efectos de los fármacos , Riñón/patología , Estrés Oxidativo/efectos de los fármacos , Piperazinas/farmacología , Piperazina/farmacología , Sustancias Protectoras/farmacología
15.
J Adv Res ; 58: 193-210, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37271476

RESUMEN

INTRODUCTION: Tumor-associated calcium signal transducer 2 (Trop2) has been used as a transport gate for cytotoxic agents into cells in antibody-drug conjugate designs because of its expression in a wide range of solid tumors. However, the specific role of Trop2 itself in breast cancer progression remains unclear and small molecules targeting Trop2 have not yet been reported. OBJECTIVES: To screen small molecules targeting Trop2, and to reveal its pharmacological effects and the molecular mechanisms of action. METHODS: Small molecule targeting Trop2 was identified by cell membrane chromatography, and validated by cellular thermal shift assay and point mutation analyses. We investigated the pharmacological effects of Trop2 inhibitor using RNA-seq, human foreskin fibroblast (HFF)-derived extracellular matrix (ECM), Matrigel drop invasion assays, colony-forming assay, xenograft tumor model, 4T1 orthotopic metastasis model and 4T1 experimental metastasis model. The molecular mechanism was determined using immunoprecipitation, mass spectrometry, immunofluorescence, immunohistochemistry and Western blotting. RESULTS: Here we identified Bruceine D (BD) as the inhibitor of Trop2, and demonstrated anti-metastasis effects of BD in breast cancer. Notably, Lys307 and Glu310 residues of Trop2 acted as critical sites for BD binding. Mechanistically, BD suppressed Trop2-induced cancer metastasis by blocking the formation of Trop2/ß-catenin positive loop, in which the Trop2/ß-catenin complex prevented ß-catenin from being degraded via the ubiquitin-proteosome pathway. Destabilized ß-catenin caused by BD reduced nucleus translocation, leading to the reduction of transcription of Trop2, the reversal of epithelial-mesenchymal transition (EMT) process, and the inhibition of ECM remodeling, further inhibiting cancer metastasis. Additionally, the inhibitory effects of BD on lung metastatic colonization and the beneficial effects of BD on prolongation of survival were validated in 4T1 experimental metastasis model. CONCLUSIONS: These results support the tumor-promoting role of Trop2 in breast cancer by stabilizing ß-catenin in Trop2/ß-catenin positive loop, and suggest Bruceine D as a promising candidate for Trop2 inhibition.


Asunto(s)
Neoplasias de la Mama , Cuassinas , Animales , Humanos , Femenino , Neoplasias de la Mama/patología , Transducción de Señal , Línea Celular Tumoral , beta Catenina/genética , beta Catenina/metabolismo , Retroalimentación , Modelos Animales de Enfermedad
16.
Natl Sci Rev ; 11(5): nwae113, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38698903

RESUMEN

Precise and efficient therapy of malignant tumors is always a challenge. Herein, gold nanoclusters co-modified by aggregation-induced-emission (AIE) molecules, copper ion chelator (acylthiourea) and tumor-targeting agent (folic acid) were fabricated to perform AIE-guided and tumor-specific synergistic therapy with great spatio-temporal controllability for the targeted elimination and metastasis inhibition of malignant tumors. During therapy, the functional gold nanoclusters (AuNTF) would rapidly accumulate in the tumor tissue due to the enhanced permeability and retention effect as well as folic acid-mediated tumor targeting, which was followed by endocytosis by tumor cells. After that, the overexpressed copper ions in the tumor cells would trigger the aggregation of these intracellular AuNTF via a chelation process that not only generated the photothermal agent in situ to perform the tumor-specific photothermal therapy damaging the primary tumor, but also led to the copper deficiency of tumor cells to inhibit its metastasis. Moreover, the copper ions were reduced to cuprous ions along with the chelation, which further catalysed the excess H2O2 in the tumor cells to produce cytotoxic reactive oxygen species, resulting in additional chemodynamic therapy for enhanced antitumor efficiency. The aggregation of AuNTF also activated the AIE molecules to present fluorescence, which not only imaged the therapeutic area for real-time monitoring of this tumor-specific synergistic therapy, but also allowed us to perform near-infrared radiation at the correct time point and location to achieve optimal photothermal therapy. Both in vitro and in vivo results revealed the strong tumor elimination, effective metastasis inhibition and high survival rate of tumor-bearing mice after treatment using the AuNTF nanoclusters, indicating that this AIE-guided and tumor-specific synergistic strategy could offer a promising approach for tumor therapy.

17.
Nanomicro Lett ; 16(1): 226, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38916749

RESUMEN

The development of supramolecular hosts which can efficiently encapsulate photosensitizers to improve the photodynamic efficacy holds great promise for cancer therapy. Here, we report two perylene diimide-based metallacages that can form stable host-guest complexes with planar conjugated molecules including polycyclic aromatic hydrocarbons and photosensitizers (hypocrellin A). Such host-guest complexation not only prevents the aggregation of photosensitizers in aqueous environments, but also offers fluorescence resonance energy transfer (FRET) from the metallacage to the photosensitizers to further improve the singlet oxygen generation (ΦΔ = 0.66). The complexes are further assembled with amphiphilic polymers, forming nanoparticles with improved stability for anticancer study. Both in vitro and in vivo studies indicate that the nanoparticles display excellent anticancer activities upon light irradiation, showing great potential for cancer photodynamic therapy. This study provides a straightforward and effective approach for enhancing the photosensitivity of conventional photosensitizers via host-guest complexation-based FRET, which will open a new avenue for host-guest chemistry-based supramolecular theranostics.

18.
Bioact Mater ; 34: 354-365, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38269307

RESUMEN

Tumor microenvironment (TME), as the "soil" of tumor growth and metastasis, exhibits significant differences from normal physiological conditions. However, how to manipulate the distinctions to achieve the accurate therapy of primary and metastatic tumors is still a challenge. Herein, an innovative nanoreactor (AH@MBTF) is developed to utilize the apparent differences (copper concentration and H2O2 level) between tumor cells and normal cells to eliminate primary tumor based on H2O2-dependent photothermal-chemodynamic therapy and suppress metastatic tumor through copper complexation. This nanoreactor is constructed using functionalized MSN incorporating benzoyl thiourea (BTU), triphenylphosphine (TPP), and folic acid (FA), while being co-loaded with horseradish peroxidase (HRP) and its substrate ABTS. During therapy, the BTU moieties on AH@MBTF could capture excessive copper (highly correlated with tumor metastasis), presenting exceptional anti-metastasis activity. Simultaneously, the complexation between BTU and copper triggers the formation of cuprous ions, which further react with H2O2 to generate cytotoxic hydroxyl radical (•OH), inhibiting tumor growth via chemodynamic therapy. Additionally, the stepwise targeting of FA and TPP guides AH@MBTF to accurately accumulate in tumor mitochondria, containing abnormally high levels of H2O2. As a catalyst, HRP mediates the oxidation reaction between ABTS and H2O2 to yield activated ABTS•+. Upon 808 nm laser irradiation, the activated ABTS•+ performs tumor-specific photothermal therapy, achieving the ablation of primary tumor by raising the tissue temperature. Collectively, this intelligent nanoreactor possesses profound potential in inhibiting tumor progression and metastasis.

19.
Acta Biomater ; 166: 485-495, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37121369

RESUMEN

The excessive copper in tumor cells is crucial for the growth and metastasis of malignant tumor. Herein, we fabricated a nanohybrid to capture, convert and utilize the overexpressed copper in tumor cells, which was expected to achieve copper dependent photothermal damage of primary tumor and copper-deficiency induced metastasis inhibition, generating accurate and effective tumor treatment. The nanohybrid consistsed of 3-azidopropylamine, 4-ethynylaniline and N-aminoethyl-N'-benzoylthiourea (BTU) co-modified gold nanoparticles (AuNPs). During therapy, the BTU segment would specifically chelate with copper in tumor cells after endocytosis to reduce the intracellular copper content, causing copper-deficiency to inhibit the vascularization and tumor migration. Meanwhile, the copper was also rapidly converted to be cuprous by BTU, which further catalyzed the click reaction between azido and alkynyl on the surface of AuNPs, resulting in on-demand aggregation of these AuNPs. This process not only in situ generated the photothermal agent in tumor cells to achieve accurate therapy avoiding unexpected damage, but also enhanced its retention time for sustained photothermal therapy. Both in vitro and in vivo results exhibited the strong tumor inhibition and high survival rate of tumor-bearing mice after application of our nanohybrid, indicating that this synergistic therapy could offer a promising approach for malignant tumor treatment. STATEMENT OF SIGNIFICANCE: The distinctive excessive copper in tumor cells is crucial for the growth and metastasis of tumor. Therefore, we fabricated intelligent gold nanoparticles to simultaneously response and reverse this tumorigenic physiological microenvironment for the synergistic therapy of malignant tumor. In this study, for the first time we converted and utilized the overexpressed Cu2+ in tumor cells to trigger intracellular click chemistry for tumor-specific photothermal therapy, resulting in accurate damage of primary tumor. Moreover, we effectively manipulated the content of Cu2+ in tumor cells to suppress the migration and vascularization of malignant tumor, resulting in effective metastasis inhibition.


Asunto(s)
Nanopartículas del Metal , Nanopartículas , Neoplasias , Animales , Ratones , Oro/farmacología , Oro/química , Cobre/farmacología , Cobre/química , Terapia Fototérmica , Nanopartículas del Metal/uso terapéutico , Nanopartículas del Metal/química , Química Clic , Nanopartículas/química , Neoplasias/patología , Línea Celular Tumoral , Microambiente Tumoral
20.
J Pharm Pharmacol ; 75(10): 1294-1309, 2023 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-37463100

RESUMEN

OBJECTIVES: Pancreatic cancer (PC) is a very lethal malignancy with a scarcity of treatment options. Although erlotinib- and gemcitabine-based treatments have been approved for PC, their effectiveness is limited. The present study is aimed at exploring the molecular and epigenetic mechanisms of anticancer activities of homoharringtonine (HHT) and its interaction with erlotinib to develop a potential therapeutic strategy for PC. METHODS: The RT-qPCR, western blotting, immunofluorescence and expression-vectors and oligonucleotide transfection were employed to determine the expression characteristics of onco-factors. Anticancer activities were determined by MTT, colony forming, and flowcytometric analysis. Dual luciferase assay was conducted to confirm putative target of miR-130b-3p. In-vivo experiments were followed by immunohistochemical assay. KEY FINDINGS: The EphB4/JAK2/STAT3 pathway drives the growth and proliferation of PC through induction of prosurvival factors and cell cycle mediators. HHT directly and epigenetically via miR-130b-3p targets EphB4, leading to downregulation of JAK2/STAT3 pathway. The inactivation of STAT3 results in diminution of antiapoptotic factors and cell cycle mediators. HHT also enhances the anticancer activity of erlotinib. CONCLUSIONS: HHT demonstrates potential anticancer activities in PC by downregulating EphB4/JAK2/STAT3 signalling. HHT also produces synergistic effects with erlotinib.


Asunto(s)
MicroARNs , Neoplasias Pancreáticas , Humanos , Homoharringtonina/farmacología , MicroARNs/metabolismo , Clorhidrato de Erlotinib/farmacología , Línea Celular Tumoral , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Proliferación Celular , Janus Quinasa 2/metabolismo , Janus Quinasa 2/farmacología , Factor de Transcripción STAT3/metabolismo , Neoplasias Pancreáticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA