Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
1.
Pharmacol Res ; 189: 106693, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36773710

RESUMEN

Uterine leiomyosarcoma (ULMS) is a malignant stromal tumor arising from the myometrium with a poor prognosis and very limited response to current chemotherapy. This study aimed to identify novel targets for ULMS through a three-step screening process using a chemical library consisting of 1271 Food and Drug Administration-approved drugs. First, we evaluated their inhibitory effects on ULMS cells and identified four candidates: proscillaridin A, lanatoside C, floxuridine, and digoxin. Then, we subcutaneously or orthotopically transplanted SK-UT-1 cells into mice to establish mouse models. In vivo analyses showed that proscillaridin A and lanatoside C exerted a superior antitumor effect. The results of mRNA sequencing showed that uncoupling protein 2 (UCP2) was suppressed in the sirtuin signaling pathway, increasing reactive oxygen species (ROS) and inducing cell death. Moreover, the downregulation of UCP2 induced ROS and suppressed ULMS cell growth. Furthermore, analyses using clinical samples showed that UCP2 expression was significantly upregulated in ULMS tissues than in myoma tissues both at the RNA and protein levels. These findings suggested that UCP2 is a potential therapeutic target and can contribute to the development of novel therapeutic strategies in patients with ULMS.


Asunto(s)
Leiomiosarcoma , Proscilaridina , Neoplasias Uterinas , Humanos , Femenino , Animales , Ratones , Leiomiosarcoma/tratamiento farmacológico , Proteína Desacopladora 2 , Proscilaridina/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo , Neoplasias Uterinas/tratamiento farmacológico
2.
Int J Mol Sci ; 23(13)2022 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-35805980

RESUMEN

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a cytotoxic cytokine that induces cancer cell death by binding to TRAIL receptors. Because of its selective cytotoxicity toward cancer cells, TRAIL therapeutics, such as recombinant TRAIL and agonistic antibodies targeting TRAIL receptors, have garnered attention as promising cancer treatment agents. However, many cancer cells acquire resistance to TRAIL-induced cell death. To overcome this issue, we searched for agents to sensitize cancer cells to TRAIL-induced cell death by screening a small-molecule chemical library consisting of diverse compounds. We identified a cardiac glycoside, proscillaridin A, as the most effective TRAIL sensitizer in colon cancer cells. Proscillaridin A synergistically enhanced TRAIL-induced cell death in TRAIL-sensitive and -resistant colon cancer cells. Additionally, proscillaridin A enhanced cell death in cells treated with TRAIL and TRAIL sensitizer, the second mitochondria-derived activator of caspase mimetic. Proscillaridin A upregulated TRAIL receptor expression, while downregulating the levels of the anti-cell death molecules, cellular FADD-like IL-1ß converting enzyme-like inhibitor protein and Mcl1, in a cell type-dependent manner. Furthermore, proscillaridin A enhanced TRAIL-induced cell death partly via O-glycosylation. Taken together, our findings suggest that proscillaridin A is a promising agent that enhances the anti-cancer efficacy of TRAIL therapeutics.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias del Colon , Proscilaridina , Ligando Inductor de Apoptosis Relacionado con TNF , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Sinergismo Farmacológico , Humanos , Proscilaridina/administración & dosificación , Proscilaridina/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
3.
Molecules ; 27(18)2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36144718

RESUMEN

The SARS-CoV-2 targets were evaluated for a set of FDA-approved drugs using a combination of drug repositioning and rigorous computational modeling methodologies such as molecular docking and molecular dynamics (MD) simulations followed by binding free energy calculations. Six FDA-approved drugs including, Ouabain, Digitoxin, Digoxin, Proscillaridin, Salinomycin and Niclosamide with promising anti-SARS-CoV-2 activity were screened in silico against four SARS-CoV-2 proteins-papain-like protease (PLpro), RNA-dependent RNA polymerase (RdRp), SARS-CoV-2 main protease (Mpro), and adaptor-associated kinase 1 (AAK1)-in an attempt to define their promising targets. The applied computational techniques suggest that all the tested drugs exhibited excellent binding patterns with higher scores and stable complexes compared to the native protein cocrystallized inhibitors. Ouabain was suggested to act as a dual inhibitor for both PLpro and Mpro enzymes, while Digitoxin bonded perfectly to RdRp. In addition, Salinomycin targeted PLpro. Particularly, Niclosamide was found to target AAK1 with greater affinity compared to the reference drug. Our study provides comprehensive molecular-level insights for identifying or designing novel anti-COVID-19 drugs.


Asunto(s)
COVID-19 , Proscilaridina , Antivirales/química , Cisteína Endopeptidasas/química , Digitoxina , Digoxina , Reposicionamiento de Medicamentos , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Niclosamida , Ouabaína , Papaína/metabolismo , ARN Polimerasa Dependiente del ARN , SARS-CoV-2
4.
Acta Biochim Biophys Sin (Shanghai) ; 53(1): 19-28, 2021 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-33201987

RESUMEN

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths globally. At present, drug options for systemic treatment of HCC are very limited. There is an urgent need to develop additional effective drugs for HCC treatment. In the present study, we found that proscillaridin A (ProA), a cardiac glycoside, exerted a strong anticancer effect on multiple HCC cell lines. ProA significantly inhibited the cell proliferation, migration, and invasion of HCC cells. ProA also had a marked inhibitory effect on the progression of HCC in the MHCC97H xenograft nude mouse model. ProA-mediated suppression of HCC was closely related to cell apoptosis. ProA-treated HCC cells displayed significant mitochondrial damage and elevated reactive oxygen species production, resulting in profound cell apoptosis. Meanwhile, ProA also played a role in autophagy induction in HCC cells. Defects in autophagy partially relieved ProA's anticancer effect in HCC cells. Our findings demonstrate that ProA can effectively inhibit HCC progression and may serve as a potential therapeutic agent for HCC treatment.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Mitocondrias/efectos de los fármacos , Proscilaridina/farmacología , Animales , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Ciclo Celular/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Xenoinjertos/efectos de los fármacos , Humanos , Neoplasias Hepáticas/patología , Ratones Endogámicos NOD , Ratones SCID , Invasividad Neoplásica , Proscilaridina/uso terapéutico , Especies Reactivas de Oxígeno/metabolismo
5.
Biochem Biophys Res Commun ; 521(4): 880-886, 2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31708095

RESUMEN

The side effects of chemotherapy, drug resistance, and tumor metastasis hinder the development of treatment for osteosarcoma, leading to poor prognosis of patients with the disease. Proscillaridin A, a kind of cardiac glycoside, has been proven to have anti-proliferative properties in many malignant tumors, but the efficacy of the drug in treating osteosarcoma is unclear. In the present study, we assessed the effects of Proscillaridin A on osteosarcoma and investigated its underlying action mechanism. The cell cytotoxicity assay showed that Proscillaridin A significantly inhibited the proliferation of 143B cells in a dose- and time-dependent manner. Also, flow cytometry and invasion assay revealed that Proscillaridin A induced apoptosis and reduced 143B cell motility. Western blotting and PCR were used to detect the expressions of Bcl-xl and MMP2 and showed that mRNA/protein expression levels decreased significantly in Proscillaridin A-treated osteosarcoma cells. Using a mouse xenograft model, we found that Proscillaridin A treatment significantly inhibited tumor growth and lung metastasis in vivo and decreased the expression levels of Bcl-xl and MMP2. No noticeable side effect was observed in the liver, kidney, and hematological functions. Conclusively, Proscillaridin A suppressed proliferation, induced apoptosis, and inhibited 143B cell metastasis in vitro and in vivo, and these effects could be mediated by downregulating the expressions of Bcl-xl and MMP2.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/patología , Proscilaridina/farmacología , Animales , Antineoplásicos/efectos adversos , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Masculino , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones Endogámicos BALB C , Proscilaridina/efectos adversos , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
6.
Int J Med Sci ; 15(8): 832-839, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30008594

RESUMEN

Cardiac glycosides are natural compounds used for the treatment of congestive heart failure and cardiac arrhythmias. Recently, they have been reported to exhibit anticancer activity. Proscillaridin A (PSN-A), a cardiac glycoside constituent of Urginea maritima has been shown to exhibit anticancer activity. However, the cellular targets and anticancer mechanism of PSN-A in various cancers including prostate cancer remain largely unexplored. In the present study, we have shown that PSN-A inhibits proliferation and induces apoptosis in prostate cancer cells in a dose-dependent manner. Further mechanistic study have shown that anticancer activity of PSN-A in prostate cancer cells is associated with ROS generation, Bcl-2 family proteins modulation, mitochondrial membrane potential disruption and ultimately activation of caspase-3 and cleavage of PARP. Moreover, we found that PSN-A inhibits JAK2/STAT3 signaling and augments doxorubicin toxicity in prostate cancer cells. Of note, LNCaP cells were found to be more sensitive to PSN-A treatment as compared to DU145 cells. Taken together, the data provided first evidence of the anticancer activity and possible molecular mechanism of PSN-A in prostate cancer. Further study is needed to develop PSN-A into a potential lead compound for the treatment of prostate cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Proscilaridina/farmacología , Neoplasias de la Próstata/tratamiento farmacológico , Factor de Transcripción STAT3/metabolismo , Antibióticos Antineoplásicos/toxicidad , Línea Celular Tumoral , Proliferación Celular , Doxorrubicina/toxicidad , Humanos , Masculino , Factor de Transcripción STAT3/efectos de los fármacos
7.
PLoS One ; 18(3): e0281981, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36913345

RESUMEN

The pandemic of COVID-19 is a severe threat to human life and the global economy. Despite the success of vaccination efforts in reducing the spread of the virus, the situation remains largely uncontrolled due to the random mutation in the RNA sequence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which demands different variants of effective drugs. Disease-causing gene-mediated proteins are usually used as receptors to explore effective drug molecules. In this study, we analyzed two different RNA-Seq and one microarray gene expression profile datasets by integrating EdgeR, LIMMA, weighted gene co-expression network and robust rank aggregation approaches, which revealed SARS-CoV-2 infection causing eight hub-genes (HubGs) including HubGs; REL, AURKA, AURKB, FBXL3, OAS1, STAT4, MMP2 and IL6 as the host genomic biomarkers. Gene Ontology and pathway enrichment analyses of HubGs significantly enriched some crucial biological processes, molecular functions, cellular components and signaling pathways that are associated with the mechanisms of SARS-CoV-2 infections. Regulatory network analysis identified top-ranked 5 TFs (SRF, PBX1, MEIS1, ESR1 and MYC) and 5 miRNAs (hsa-miR-106b-5p, hsa-miR-20b-5p, hsa-miR-93-5p, hsa-miR-106a-5p and hsa-miR-20a-5p) as the key transcriptional and post-transcriptional regulators of HubGs. Then, we conducted a molecular docking analysis to determine potential drug candidates that could interact with HubGs-mediated receptors. This analysis resulted in the identification of top-ranked ten drug agents, including Nilotinib, Tegobuvir, Digoxin, Proscillaridin, Olysio, Simeprevir, Hesperidin, Oleanolic Acid, Naltrindole and Danoprevir. Finally, we investigated the binding stability of the top-ranked three drug molecules Nilotinib, Tegobuvir and Proscillaridin with the three top-ranked proposed receptors (AURKA, AURKB, OAS1) by using 100 ns MD-based MM-PBSA simulations and observed their stable performance. Therefore, the findings of this study might be useful resources for diagnosis and therapies of SARS-CoV-2 infections.


Asunto(s)
COVID-19 , MicroARNs , Proscilaridina , Humanos , COVID-19/diagnóstico , COVID-19/genética , Transcriptoma , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Simulación del Acoplamiento Molecular , Aurora Quinasa A/genética , MicroARNs/genética , Redes Reguladoras de Genes , Biomarcadores , Genómica , Prueba de COVID-19
8.
J Biomol Struct Dyn ; 40(8): 3681-3696, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-33228475

RESUMEN

The coronavirus disease, caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is a global health crisis that is being endured with an increased alarm of transmission each day. Though the pandemic has activated innumerable research attention to decipher an antidote, fundamental understanding of the molecular mechanisms is necessary to halt the disease progression. The study focused on comparison of the COVID-19 infected lung tissue gene expression datasets -GSE155241 and GSE150316 with the GEO2R-limma package. The significant up- and downregulated genes were annotated. Further evaluation of the enriched pathways, transcription factors, kinases, noncoding RNAs and drug perturbations revealed the significant molecular mechanisms of the host response. The results revealed a surge in mitochondrial respiration, cytokines, neurodegenerative mechanisms and deprived oxygen, iron, copper, and glucose transport. Hijack of ubiquitination by SARS-CoV-2, hox gene differentiation, histone modification, and miRNA biogenesis were the notable molecular mechanisms inferred. Long non-coding RNAs such as C058791.1, TTTY15 and TPTEP1 were predicted to be efficient in regulating the disease mechanisms. Drugs-F-1566-0341, Digoxin, Proscillaridin and Linifanib that reverse the gene expression signatures were predicted from drug perturbations analysis. The binding efficiency and interaction of proscillaridin and digoxin as obtained from the molecular docking studies confirmed their therapeutic potential. Two overlapping upregulated genes MDH1, SGCE and one downregulated gene PFKFB3 were appraised as potential biomarkers candidates. The upregulation of PGM5, ISLR and ANK2 as measured from their expressions in normal lungs affirmed their possible prognostic biomarker competence. The study explored significant insights for better diagnosis, and therapeutic options for COVID-19. Communicated by Ramaswamy H. Sarma.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , MicroARNs , Proscilaridina , Biomarcadores , COVID-19/genética , Digoxina , Perfilación de la Expresión Génica , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Simulación del Acoplamiento Molecular , SARS-CoV-2/genética
9.
Cancer Chemother Pharmacol ; 88(5): 845-856, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34331108

RESUMEN

PURPOSE: Embryonal rhabdomyosarcoma (eRMS) is the most common type of rhabdomyosarcoma in children. eRMS is characterized by malignant skeletal muscle cells driven by hyperactivation of several oncogenic pathways including the MYC pathway. Targeting MYC in cancer has been extremely challenging. Recently, we have demonstrated that the heart failure drug, proscillaridin A, produced anticancer effects with specificity toward MYC expressing leukemia cells. We also reported that decitabine, a hypomethylating drug, synergizes with proscillaridin A in colon cancer cells. Here, we investigated whether proscillaridin A exhibits epigenetic and anticancer activity against eRMS RD cells, overexpressing MYC oncogene, and its combination with decitabine. METHODS: We investigated the anticancer effects of proscillaridin A in eRMS RD cells in vitro. In response to drug treatment, we measured growth inhibition, cell cycle arrest, loss of clonogenicity and self-renewal capacity. We further evaluated the impact of proscillaridin A on MYC expression and its downstream transcriptomic effects by RNA sequencing. Then, we measured protein expression of epigenetic regulators and their associated chromatin post-translational modifications in response to drug treatment. Chromatin immunoprecipitation sequencing data sets were coupled with transcriptomic results to pinpoint the impact of proscillaridin A on gene pathways associated with specific chromatin modifications. Lastly, we evaluated the effect of the combination of proscillaridin A and the DNA demethylating drug decitabine on eRMS RD cell growth and clonogenic potential. RESULTS: Clinically relevant concentration of proscillaridin A (5 nM) produced growth inhibition, cell cycle arrest and loss of clonogenicity in eRMS RD cells. Proscillaridin A produced a significant downregulation of MYC protein expression and inhibition of oncogenic transcriptional programs controlled by MYC, involved in cell replication. Interestingly, significant reduction in total histone 3 acetylation and on specific lysine residues (lysine 9, 14, 18, and 27 on histone 3) was associated with significant protein downregulation of a series of lysine acetyltransferases (KAT3A, KAT3B, KAT2A, KAT2B, and KAT5). In addition, proscillaridin A produced synergistic growth inhibition and loss of clonogenicity when combined with the approved DNA demethylating drug decitabine. CONCLUSION: Proscillaridin A produces anticancer and epigenetic effects in the low nanomolar range and its combination with decitabine warrants further investigation for the treatment of eRMS.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proscilaridina/farmacología , Rabdomiosarcoma Embrionario/tratamiento farmacológico , Acetilación/efectos de los fármacos , Línea Celular Tumoral , Autorrenovación de las Células/efectos de los fármacos , Decitabina/administración & dosificación , Reposicionamiento de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histonas/metabolismo , Humanos , Lisina/metabolismo , Proteínas de Neoplasias , Regiones Promotoras Genéticas/efectos de los fármacos , Proscilaridina/administración & dosificación , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Rabdomiosarcoma Embrionario/genética , Rabdomiosarcoma Embrionario/patología
10.
Acta Pol Pharm ; 67(6): 630-4, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21229879

RESUMEN

Two modified glycosides--digoxin and proscillaridin A conjugated to a generation 3 of polyamidoamine dendrimer (G3 PAMAM-NH2) were evaluated as DNA topoisomerase II inhibitors. The ability of these compounds (PAMAM-Dig and PAMAM-Prosc) to inhibit topoisomerase I and II activity was quantified by measuring the action on supercoiled DNA substrate as a function of increasing concentration of the test compounds by the use of agarose gel electrophoresis. The obtained results suggest that a conjugation of the modified glycosides with G3 PAMAM-NH2 significantly improved the ability of the parent compounds to an inhibition of DNA topoisomerases.


Asunto(s)
ADN-Topoisomerasas de Tipo I , Proteínas de Unión al ADN/antagonistas & inhibidores , Dendrímeros/farmacología , Digoxina/farmacología , Proscilaridina/farmacología , Inhibidores de Topoisomerasa/farmacología , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , ADN-Topoisomerasas de Tipo I/química , ADN-Topoisomerasas de Tipo I/metabolismo , ADN-Topoisomerasas de Tipo II/química , ADN-Topoisomerasas de Tipo II/metabolismo , ADN Superhelicoidal/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Dendrímeros/química , Digoxina/química , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Agar , Humanos , Estructura Molecular , Proscilaridina/química , Relación Estructura-Actividad , Inhibidores de Topoisomerasa/química
11.
Front Immunol ; 11: 162, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32174909

RESUMEN

Neuroinflammation induced by overactivated glia cells is believed to be a major hallmark of Alzheimer's disease (AD) and a hopeful target against AD. A rhamnoside PL201 was previously reported to promote neurogenesis and ameliorate AD, and in this study, we revealed that PL201 also significantly reduced accumulation of the activated microglia and proinflammatory cytokines in APP/PS1 mice. In vitro, PL201 consistently suppressed the microglia induction of proinflammatory cytokines after stimulation with lipopolysaccharides and Aß42. Further mechanistic studies demonstrated that PL201 considerably enhanced the expression level and the nuclear translocation of Nrf2, a key regulator of neuroinflammation. Moreover, PL201 effectively stimulated Nrf2 signaling cascade, including upregulation of HO-1 and downregulation of NF-κB pathway. Thus, our findings indicated the anti-neuroinflammatory effect by PL201 in vivo and suggested that PL201 or the like, with multiple functions such as neurogenesis, mitochondria maintenance, and anti-neuroinflammation, could be a promising candidate in AD treatment.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Microglía/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , Proscilaridina/análogos & derivados , Proscilaridina/administración & dosificación , Transducción de Señal/efectos de los fármacos , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/farmacología , Precursor de Proteína beta-Amiloide/genética , Animales , Línea Celular Transformada , Citocinas/metabolismo , Femenino , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/farmacología , Presenilina-1/genética , Resultado del Tratamiento
12.
Cell Cycle ; 19(5): 541-550, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32009541

RESUMEN

Prostate cancer (PCa) is the second commonly diagnosed malignancy in men over the world. Although androgen deprivation therapy for advanced PCa patients has significantly improved their survival, the majority of these patients eventually develop castration-resistant prostate cancer (CRPC). Proscillaridin A (Pro A), a cardiac glycoside that is clinically used to treat various heart failure diseases, has been reported to have anticancer activity in several cancers. However, whether Pro A exerts an inhibitory effect on PCa progression remains unknown. In this study, we determined possible antitumor effects of Pro A on PCa cells and demonstrated the following: firstly, Pro A selectively inhibited androgen-independent PCa (including PC3 and DU145) cell growth and induced cell apoptosis in vitro; secondly, Pro A significantly decreased cell motility and invasion of androgen-independent PCa cells; thirdly, Pro A enhanced the sensitivity of PCa cells to docetaxel; fourthly, Pro A significantly inhibited the growth of PCa xenografts in vivo and patient-derived organoids (PDO). In addition, RNA-sequencing analysis revealed that the antitumor effects of Pro A on androgen-independent PCa appeared to be achieved via driving the activation of endoplasmic reticulum stress. The antitumor effects of Pro A could be ameliorated by reactive oxygen species scavenger and ER stress inhibitors. Therefore, these data suggest that Pro A may provide a potential therapeutic option for the treatment of PCa, particularly CRPC.


Asunto(s)
Progresión de la Enfermedad , Estrés del Retículo Endoplásmico/efectos de los fármacos , Proscilaridina/farmacología , Proscilaridina/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Andrógenos , Animales , Apoptosis/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Docetaxel/farmacología , Humanos , Masculino , Ratones , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/patología , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Oxid Med Cell Longev ; 2020: 7698560, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32104538

RESUMEN

Oxidative stress-induced mitochondrial dysfunction and cell senescence are considered critical contributors to Alzheimer's disease (AD), and oxidant/antioxidant imbalance has been a therapeutic target in AD. SIRT3 is a mitochondrial protein regulating metabolic enzyme activity by deacetylation and its downregulation is associated with AD pathology. In the present study, we showed that a newly synthesized rhamnoside derivative PL171 inhibited the generation of reactive oxidant species (ROS) induced by amyloid-ß 42 oligomers (Aß 42O), major AD pathological proteins. Moreover, the reduction of mitochondrial membrane potential (MMP) and the impairment of mitochondrial oxygen consumption triggered by Aß 42O were also prevented by PL171. Further experiments demonstrated that PL171 reduced the acetylation of mitochondrial proteins, and particularly the acetylation of manganese superoxide dismutase (MnSOD) and oligomycin-sensitivity-conferring protein (OSCP), two mitochondrial SIRT3 substrates, was suppressed by PL171. Mechanism studies revealed that PL171 upregulated SIRT3 and its upstream peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) under basal and Aß 42O-treated conditions. The inhibition of SIRT3 activity could eliminate the protective effects of PL171. Further, long-term treatment with Aß 42O increased the number of senescent neuronal cell, which was also alleviated by PL171 in a SIRT3-dependent manner. Taken together, our results indicated that PL171 rescued Aß 42O-induced oxidative stress, mitochondrial dysfunction, and cell senescence via upregulating SIRT3 and might be a potential drug candidate against AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Senescencia Celular/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Sirtuina 3/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Línea Celular Tumoral , Senescencia Celular/genética , Humanos , Mitocondrias/metabolismo , Mitocondrias/patología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Estrés Oxidativo/genética , Oxígeno/metabolismo , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/toxicidad , Proscilaridina/análogos & derivados , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 3/antagonistas & inhibidores , Sirtuina 3/genética , Superóxido Dismutasa/metabolismo , Regulación hacia Arriba
14.
J Nat Prod ; 72(11): 1969-74, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19894733

RESUMEN

Cardiac glycosides have been reported to exhibit cytotoxic activity against several different cancer types, but studies against colorectal cancer are lacking. In a screening procedure aimed at identifying natural products with activity against colon cancer, several cardiac glycosides were shown to be of interest, and five of these were further evaluated in different colorectal cancer cell lines and primary cells from patients. Convallatoxin (1), oleandrin (4), and proscillaridin A (5) were identified as the most potent compounds (submicromolar IC50 values), and digitoxin (2) and digoxin (3), which are used in cardiac disease, exhibited somewhat lower activity (IC50 values 0.27-4.1 microM). Selected cardiac glycosides were tested in combination with four clinically relevant cytotoxic drugs (5-fluorouracil, oxaliplatin, cisplatin, irinotecan). The combination of 2 and oxaliplatin exhibited synergism including the otherwise highly drug-resistant HT29 cell line. A ChemGPS-NP application comparing modes of action of anticancer drugs identified cardiac glycosides as a separate cluster. These findings demonstrate that such substances may exhibit significant activity against colorectal cancer cell lines, by mechanisms disparate from currently used anticancer drugs, but at concentrations generally considered not achievable in patient plasma.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Camptotecina/análogos & derivados , Camptotecina/farmacología , Cardenólidos/sangre , Cardenólidos/química , Cardenólidos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Digitoxina/sangre , Digitoxina/química , Digitoxina/farmacología , Digoxina/sangre , Digoxina/química , Digoxina/farmacología , Ensayos de Selección de Medicamentos Antitumorales , Células HT29 , Humanos , Irinotecán , FN-kappa B/efectos de los fármacos , Proscilaridina/sangre , Proscilaridina/química , Proscilaridina/farmacología , Estrofantinas/sangre , Estrofantinas/química , Estrofantinas/farmacología
15.
J Exp Clin Cancer Res ; 38(1): 251, 2019 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-31196146

RESUMEN

BACKGROUND: Cardiac glycosides are approved for the treatment of heart failure as Na+/K+ pump inhibitors. Their repurposing in oncology is currently investigated in preclinical and clinical studies. However, the identification of a specific cancer type defined by a molecular signature to design targeted clinical trials with cardiac glycosides remains to be characterized. Here, we demonstrate that cardiac glycoside proscillaridin A specifically targets MYC overexpressing leukemia cells and leukemia stem cells by causing MYC degradation, epigenetic reprogramming and leukemia differentiation through loss of lysine acetylation. METHODS: Proscillaridin A anticancer activity was investigated against a panel of human leukemia and solid tumor cell lines with different MYC expression levels, overexpression in vitro systems and leukemia stem cells. RNA-sequencing and differentiation studies were used to characterize transcriptional and phenotypic changes. Drug-induced epigenetic changes were studied by chromatin post-translational modification analysis, expression of chromatin regulators, chromatin immunoprecipitation, and mass-spectrometry. RESULTS: At a clinically relevant dose, proscillaridin A rapidly altered MYC protein half-life causing MYC degradation and growth inhibition. Transcriptomic profile of leukemic cells after treatment showed a downregulation of genes involved in MYC pathways, cell replication and an upregulation of hematopoietic differentiation genes. Functional studies confirmed cell cycle inhibition and the onset of leukemia differentiation even after drug removal. Proscillaridin A induced a significant loss of lysine acetylation in histone H3 (at lysine 9, 14, 18 and 27) and in non-histone proteins such as MYC itself, MYC target proteins, and a series of histone acetylation regulators. Global loss of acetylation correlated with the rapid downregulation of histone acetyltransferases. Importantly, proscillaridin A demonstrated anticancer activity against lymphoid and myeloid stem cell populations characterized by MYC overexpression. CONCLUSION: Overall, these results strongly support the repurposing of proscillaridin A in MYC overexpressing leukemia.


Asunto(s)
Antineoplásicos/efectos adversos , Expresión Génica/efectos de los fármacos , Genes myc , Insuficiencia Cardíaca/etiología , Leucemia/genética , Lisina/metabolismo , Proscilaridina/efectos adversos , Acetilación , Antineoplásicos/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cromatina/genética , Cromatina/metabolismo , Relación Dosis-Respuesta a Droga , Epigénesis Genética/efectos de los fármacos , Perfilación de la Expresión Génica , Histonas/metabolismo , Humanos , Leucemia/complicaciones , Leucemia/tratamiento farmacológico , Leucemia/metabolismo , Modelos Biológicos , Proscilaridina/uso terapéutico , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
16.
Nat Commun ; 10(1): 4731, 2019 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-31636264

RESUMEN

Compounds with specific cytotoxic activity in senescent cells, or senolytics, support the causal involvement of senescence in aging and offer therapeutic interventions. Here we report the identification of Cardiac Glycosides (CGs) as a family of compounds with senolytic activity. CGs, by targeting the Na+/K+ATPase pump, cause a disbalanced electrochemical gradient within the cell causing depolarization and acidification. Senescent cells present a slightly depolarized plasma membrane and higher concentrations of H+, making them more susceptible to the action of CGs. These vulnerabilities can be exploited for therapeutic purposes as evidenced by the in vivo eradication of tumors xenografted in mice after treatment with the combination of a senogenic and a senolytic drug. The senolytic effect of CGs is also effective in the elimination of senescence-induced lung fibrosis. This experimental approach allows the identification of compounds with senolytic activity that could potentially be used to develop effective treatments against age-related diseases.


Asunto(s)
Apoptosis/efectos de los fármacos , Glicósidos Cardíacos/farmacología , Senescencia Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Células A549 , Animales , Antibióticos Antineoplásicos/farmacología , Bleomicina/farmacología , Neoplasias de la Mama , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Digoxina/farmacología , Femenino , Humanos , Concentración de Iones de Hidrógeno/efectos de los fármacos , Ratones , Osteoartritis , Ouabaína/farmacología , Proscilaridina/farmacología , Fibrosis Pulmonar , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Oncogene ; 37(32): 4372-4384, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29743592

RESUMEN

The RAS proteins are the most frequently mutated oncogenes in cancer, with highest frequency found in pancreatic, lung, and colon tumors. Moreover, the activity of RAS is required for the proliferation and/or survival of these tumor cells and thus represents a high-value target for therapeutic development. Direct targeting of RAS has proven challenging for multiple reasons stemming from the biology of the protein, the complexity of downstream effector pathways and upstream regulatory networks. Thus, significant efforts have been directed at identifying downstream targets on which RAS is dependent. These efforts have proven challenging, in part due to confounding factors such as reliance on two-dimensional adherent monolayer cell cultures that inadequately recapitulate the physiologic context to which cells are exposed in vivo. To overcome these issues, we implemented a high-throughput screening (HTS) approach using a spheroid-based 3-dimensional culture format, thought to more closely reflect conditions experienced by cells in vivo. Using isogenic cell pairs, differing in the status of KRAS, we identified Proscillaridin A as a selective inhibitor of cells harboring the oncogenic KRasG12V allele. Significantly, the identification of Proscillaridin A was facilitated by the 3D screening platform and would not have been discovered employing standard 2D culturing methods.


Asunto(s)
Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Antineoplásicos/farmacología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales/métodos , Humanos , Fenotipo , Proscilaridina/farmacología , Transducción de Señal/genética
18.
Cell Death Dis ; 9(6): 696, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29899551

RESUMEN

Non-small-cell lung cancer (NSCLC) is the predominant histological type of lung cancer and is characterized by the highest mortality and incidence rates among these types of malignancies. Cardiac glycosides, a class of natural products, have been identified as a potential type of chemotherapeutic agent. This study aims to investigate the anti-cancer effects and the mechanisms of action of Proscillaridin A (P.A) in NSCLC cells. In vitro sodium-potassium pump (Na+/K+ ATPase) enzyme assays indicated that P.A is a direct Na+/K+ ATPase inhibitor. P.A showed potent cytotoxic effects in NSCLC cells at nanomolar levels. Treatment mechanism studies indicated that P.A elevated Ca2+ levels, activated the AMPK pathway and downregulated phosphorylation of ACC and mTOR. Subsequently, P.A increased death receptor 4 (DR4) expression and downregulated NF-κB. Interestingly, P.A selectively suppressed EGFR activation in EGFR mutant cells but not in EGFR wild-type cells. In vivo, P.A significantly suppressed tumor growth in nude mice compared to vehicle-treated mice. Compared with the Afatinib treatment group, P.A displayed less pharmaceutical toxicity, as the body weight of mice treated with P.A did not decrease as much as those treated with Afatinib. Consistent changes in protein levels were obtained from western blotting analysis of tumors and cell lines. Immunohistochemistry analysis of the tumors from P.A-treated mice showed a significant suppression of EGFR phosphorylation (Tyr 1173) and reduction of the cell proliferation marker Ki-67. Taken together, our results suggest that P.A is a promising anti-cancer therapeutic candidate for NSCLC.


Asunto(s)
Apoptosis/efectos de los fármacos , Calcio/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Proscilaridina/farmacología , Receptores del Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Acetil-CoA Carboxilasa/metabolismo , Adenilato Quinasa/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Pulmonares/metabolismo , Modelos Biológicos , Mutación/genética , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Proscilaridina/química , Transducción de Señal/efectos de los fármacos , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Ensayo de Tumor de Célula Madre , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cell Death Dis ; 9(10): 984, 2018 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-30250248

RESUMEN

Glioblastoma (GBM) is characterized by highly aggressive growth and invasive behavior. Due to the highly lethal nature of GBM, new therapies are urgently needed and repositioning of existing drugs is a promising approach. We have previously shown the activity of Proscillaridin A (ProA), a cardiac glycoside inhibitor of the Na(+)/K(+) ATPase (NKA) pump, against proliferation and migration of GBM cell lines. ProA inhibited tumor growth in vivo and increased mice survival after orthotopic grafting of GBM cells. This study aims to decipher the mechanism of action of ProA in GBM tumor and stem-like cells. ProA displayed cytotoxic activity on tumor and stem-like cells grown in 2D and 3D culture, but not on healthy cells as astrocytes or oligodendrocytes. Even at sub-cytotoxic concentration, ProA impaired cell migration and disturbed EB1 accumulation at microtubule (MT) plus-ends and MT dynamics instability. ProA activates GSK3ß downstream of NKA inhibition, leading to EB1 phosphorylation on S155 and T166, EB1 comet length shortening and MT dynamics alteration, and finally inhibition of cell migration and cytotoxicity. Similar results were observed with digoxin. Therefore, we disclosed here a novel pathway by which ProA and digoxin modulate MT-governed functions in GBM tumor and stem-like cells. Altogether, our results support ProA and digoxin as potent candidates for drug repositioning in GBM.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/patología , Glioblastoma/patología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Microtúbulos/metabolismo , Proscilaridina/farmacología , Adenosina Trifosfatasas/metabolismo , Animales , Astrocitos/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Bombas Iónicas/metabolismo , Ratones , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/metabolismo , Fosforilación/efectos de los fármacos , Polimerizacion/efectos de los fármacos , Tubulina (Proteína)/metabolismo
20.
Oxid Med Cell Longev ; 2018: 3853409, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29576846

RESUMEN

Cardiac glycosides are natural compounds used for the treatment of cardiovascular disorders. Although originally prescribed for cardiovascular diseases, more recently, they have been rediscovered for their potential use in the treatment of cancer. Proscillaridin A (PSD-A), a cardiac glycoside component of Urginea maritima, has been reported to exhibit anticancer activity. However, the cellular targets and anticancer mechanism of PSD-A in various cancers including lung cancer remain largely unexplored. In the present study, we found that PSD-A inhibits growth and induces apoptosis in A549 lung adenocarcinoma cells. The anticancer activity of PSD-A was found to be associated with the activation of JNK, induction of ER stress, mitochondrial dysfunction, and inhibition of STAT3 activation. PSD-A induces oxidative stress as evidenced from ROS generation, GSH depletion, and decreased activity of TrxR1. PSD-A-mediated ER stress was verified by increased phosphorylation of eIF2α and expression of its downstream effector proteins ATF4, CHOP, and caspases-4. PSD-A triggered apoptosis by inducing JNK (1/2) activation, increasing bax/bcl-2 ratio, dissipating mitochondrial membrane potential, and inducing cleavage of caspases and PARP. Further study revealed that PSD-A inhibits both constitutive and inducible STAT3 activations and decreases STAT3 DNA-binding activity. Moreover, PSD-A-mediated inhibition of STAT3 activation was found to be associated with increased SHP-1 expression, decreased phosphorylation of Src, and binding of PSD-A with STAT3 SH2 domain. Finally, STAT3 knockdown by shRNA inhibited growth and enhanced apoptotic efficacy of PSD-A. Taken together, the data suggest that PSD-A could be developed into a potential therapeutic agent against lung adenocarcinoma.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Proscilaridina/farmacología , Factor de Transcripción STAT3/antagonistas & inhibidores , Células A549 , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Células HEK293 , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Estrés Oxidativo/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda