Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
1.
PLoS One ; 18(3): e0281981, 2023.
Article in English | MEDLINE | ID: mdl-36913345

ABSTRACT

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.


Subject(s)
COVID-19 , MicroRNAs , Proscillaridin , Humans , COVID-19/diagnosis , COVID-19/genetics , Transcriptome , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Molecular Docking Simulation , Aurora Kinase A/genetics , MicroRNAs/genetics , Gene Regulatory Networks , Biomarkers , Genomics , COVID-19 Testing
2.
Pharmacol Res ; 189: 106693, 2023 03.
Article in English | MEDLINE | ID: mdl-36773710

ABSTRACT

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.


Subject(s)
Leiomyosarcoma , Proscillaridin , Uterine Neoplasms , Humans , Female , Animals , Mice , Leiomyosarcoma/drug therapy , Uncoupling Protein 2 , Proscillaridin/therapeutic use , Reactive Oxygen Species/metabolism , Uterine Neoplasms/drug therapy
3.
Molecules ; 27(18)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36144718

ABSTRACT

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.


Subject(s)
COVID-19 , Proscillaridin , Antiviral Agents/chemistry , Cysteine Endopeptidases/chemistry , Digitoxin , Digoxin , Drug Repositioning , Humans , Molecular Docking Simulation , Molecular Dynamics Simulation , Niclosamide , Ouabain , Papain/metabolism , RNA-Dependent RNA Polymerase , SARS-CoV-2
4.
Int J Mol Sci ; 23(13)2022 Jun 23.
Article in English | MEDLINE | ID: mdl-35805980

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Colonic Neoplasms , Proscillaridin , TNF-Related Apoptosis-Inducing Ligand , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Synergism , Humans , Proscillaridin/administration & dosage , Proscillaridin/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , TNF-Related Apoptosis-Inducing Ligand/pharmacology
5.
J Biomol Struct Dyn ; 40(8): 3681-3696, 2022 05.
Article in English | MEDLINE | ID: mdl-33228475

ABSTRACT

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.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , MicroRNAs , Proscillaridin , Biomarkers , COVID-19/genetics , Digoxin , Gene Expression Profiling , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Molecular Docking Simulation , SARS-CoV-2/genetics
6.
Cancer Chemother Pharmacol ; 88(5): 845-856, 2021 11.
Article in English | MEDLINE | ID: mdl-34331108

ABSTRACT

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.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Proscillaridin/pharmacology , Rhabdomyosarcoma, Embryonal/drug therapy , Acetylation/drug effects , Cell Line, Tumor , Cell Self Renewal/drug effects , Decitabine/administration & dosage , Drug Repositioning , Gene Expression Regulation, Neoplastic/drug effects , Histones/metabolism , Humans , Lysine/metabolism , Neoplasm Proteins , Promoter Regions, Genetic/drug effects , Proscillaridin/administration & dosage , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Rhabdomyosarcoma, Embryonal/genetics , Rhabdomyosarcoma, Embryonal/pathology
7.
Acta Biochim Biophys Sin (Shanghai) ; 53(1): 19-28, 2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33201987

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Mitochondria/drug effects , Proscillaridin/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Heterografts/drug effects , Humans , Liver Neoplasms/pathology , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Proscillaridin/therapeutic use , Reactive Oxygen Species/metabolism
8.
Front Immunol ; 11: 162, 2020.
Article in English | MEDLINE | ID: mdl-32174909

ABSTRACT

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.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Microglia/metabolism , NF-E2-Related Factor 2/metabolism , Proscillaridin/analogs & derivatives , Proscillaridin/administration & dosage , Signal Transduction/drug effects , Alzheimer Disease/pathology , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line, Transformed , Cytokines/metabolism , Female , Humans , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Transgenic , Peptide Fragments/pharmacology , Presenilin-1/genetics , Treatment Outcome
9.
Oxid Med Cell Longev ; 2020: 7698560, 2020.
Article in English | MEDLINE | ID: mdl-32104538

ABSTRACT

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.


Subject(s)
Alzheimer Disease/drug therapy , Cellular Senescence/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Oxidative Stress/drug effects , Sirtuin 3/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/toxicity , Cell Line, Tumor , Cellular Senescence/genetics , Humans , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Proton-Translocating ATPases/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/genetics , Oxygen/metabolism , Peptide Fragments/metabolism , Peptide Fragments/toxicity , Proscillaridin/analogs & derivatives , Reactive Oxygen Species/metabolism , Sirtuin 3/antagonists & inhibitors , Sirtuin 3/genetics , Superoxide Dismutase/metabolism , Up-Regulation
10.
Cell Cycle ; 19(5): 541-550, 2020 03.
Article in English | MEDLINE | ID: mdl-32009541

ABSTRACT

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.


Subject(s)
Disease Progression , Endoplasmic Reticulum Stress/drug effects , Proscillaridin/pharmacology , Proscillaridin/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Androgens , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Docetaxel/pharmacology , Humans , Male , Mice , Neoplasm Metastasis/drug therapy , Neoplasm Metastasis/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , Reactive Oxygen Species/antagonists & inhibitors , Xenograft Model Antitumor Assays
11.
Biochem Biophys Res Commun ; 521(4): 880-886, 2020 01 22.
Article in English | MEDLINE | ID: mdl-31708095

ABSTRACT

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.


Subject(s)
Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Proscillaridin/pharmacology , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice, Inbred BALB C , Proscillaridin/adverse effects , Xenograft Model Antitumor Assays , bcl-X Protein/genetics , bcl-X Protein/metabolism
12.
Nat Commun ; 10(1): 4731, 2019 10 21.
Article in English | MEDLINE | ID: mdl-31636264

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Cardiac Glycosides/pharmacology , Cellular Senescence/drug effects , Chondrocytes/drug effects , Fibroblasts/drug effects , A549 Cells , Animals , Antibiotics, Antineoplastic/pharmacology , Bleomycin/pharmacology , Breast Neoplasms , Cell Line, Tumor , Cell Membrane/drug effects , Digoxin/pharmacology , Female , Humans , Hydrogen-Ion Concentration/drug effects , Mice , Osteoarthritis , Ouabain/pharmacology , Proscillaridin/pharmacology , Pulmonary Fibrosis , Xenograft Model Antitumor Assays
13.
J Exp Clin Cancer Res ; 38(1): 251, 2019 Jun 13.
Article in English | MEDLINE | ID: mdl-31196146

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/adverse effects , Gene Expression/drug effects , Genes, myc , Heart Failure/etiology , Leukemia/genetics , Lysine/metabolism , Proscillaridin/adverse effects , Acetylation , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/genetics , Chromatin/metabolism , Dose-Response Relationship, Drug , Epigenesis, Genetic/drug effects , Gene Expression Profiling , Histones/metabolism , Humans , Leukemia/complications , Leukemia/drug therapy , Leukemia/metabolism , Models, Biological , Proscillaridin/therapeutic use , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
14.
Cell Death Dis ; 9(10): 984, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250248

ABSTRACT

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.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Glioblastoma/pathology , Glycogen Synthase Kinase 3 beta/metabolism , Microtubules/metabolism , Proscillaridin/pharmacology , Adenosine Triphosphatases/metabolism , Animals , Astrocytes/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Ion Pumps/metabolism , Mice , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/metabolism , Phosphorylation/drug effects , Polymerization/drug effects , Tubulin/metabolism
15.
Int J Med Sci ; 15(8): 832-839, 2018.
Article in English | MEDLINE | ID: mdl-30008594

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/pharmacology , Proscillaridin/pharmacology , Prostatic Neoplasms/drug therapy , STAT3 Transcription Factor/metabolism , Antibiotics, Antineoplastic/toxicity , Cell Line, Tumor , Cell Proliferation , Doxorubicin/toxicity , Humans , Male , STAT3 Transcription Factor/drug effects
16.
Cell Death Dis ; 9(6): 696, 2018 06 13.
Article in English | MEDLINE | ID: mdl-29899551

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Calcium/pharmacology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Proscillaridin/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Up-Regulation/drug effects , Acetyl-CoA Carboxylase/metabolism , Adenylate Kinase/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lung Neoplasms/metabolism , Models, Biological , Mutation/genetics , NF-kappa B/metabolism , Phosphorylation/drug effects , Proscillaridin/chemistry , Signal Transduction/drug effects , Sodium-Potassium-Exchanging ATPase/metabolism , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
17.
Oncogene ; 37(32): 4372-4384, 2018 08.
Article in English | MEDLINE | ID: mdl-29743592

ABSTRACT

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.


Subject(s)
Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Antineoplastic Agents/pharmacology , Cell Culture Techniques , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Humans , Phenotype , Proscillaridin/pharmacology , Signal Transduction/genetics
18.
Oxid Med Cell Longev ; 2018: 3853409, 2018.
Article in English | MEDLINE | ID: mdl-29576846

ABSTRACT

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.


Subject(s)
Adenocarcinoma/drug therapy , Endoplasmic Reticulum Stress/drug effects , Lung Neoplasms/drug therapy , Proscillaridin/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , A549 Cells , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , HEK293 Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Oxidative Stress/drug effects , STAT3 Transcription Factor/metabolism
19.
SLAS Discov ; 22(1): 67-76, 2017 01.
Article in English | MEDLINE | ID: mdl-27703082

ABSTRACT

Parkinson disease (PD) is a prevalent neurodegenerative disease characterized by selective degeneration of dopaminergic neurons in the substantia nigra, causing tremor and motor impairment. Parkin protein, whose mutants are the cause of Parkinson disease type 2 (PARK2), has been mechanistically linked to the regulation of apoptosis and the turnover of damaged mitochondria. Several studies have implicated aberrant mitochondria as a key contributor to the development of PD. In the attempt to discover new drugs, high-content cell-based assays are becoming more important to mimic the nature of biological processes and their diversifications in diseases and will be essential for lead identification and the optimization of therapeutic candidates. We have developed a novel fluorescence cell-based assay for high-content screening to find compounds that can promote the mitochondrial localization of Parkin without severe mitochondrial damage induction. In this work, this model was used to screen a library of 1280 compounds. After the screening campaign, the positive compounds were chosen for further testing, based on the strength of the initial response and lack of cytotoxicity. These results indicated that this Parkin cell-based assay is a robust (Z' > 0.5) and valid strategy to test potential candidates for preclinical studies.


Subject(s)
Biological Assay/methods , Drug Evaluation, Preclinical/methods , Parkinson Disease/drug therapy , Ubiquitin-Protein Ligases/metabolism , Cell Line, Tumor , Fluorescence , Humans , Mitochondria/metabolism , Parkinson Disease/metabolism , Proscillaridin/therapeutic use , Rhodamines
20.
Sci Rep ; 5: 17047, 2015 Nov 23.
Article in English | MEDLINE | ID: mdl-26592202

ABSTRACT

Sodium taurocholate cotransporting polypeptide (NTCP) has been reported as a functional receptor for hepatitis B virus (HBV) infection. However, HBV could not efficiently infect HepG2 cells expressing NTCP (NTCP-HepG2 cells) under adherent monolayer-cell conditions. In this study, NTCP was mainly detected in the basolateral membrane region, but not the apical site, of monolayer NTCP-HepG2 cells. We hypothesized that non-adherent cell conditions of infection would enhance HBV infectivity. Non-adherent NTCP-HepG2 cells were prepared by treatment with trypsin and EDTA, which did not degrade NTCP in the membrane fraction. HBV successfully infected NTCP-HepG2 cells at a viral dose 10 times lower in non-adherent phase than in adherent phase. Efficient infection of non-adherent NTCP-HepG2 cells with blood-borne or cell-culture-derived HBV was observed and was remarkably impaired in the presence of the myristoylated preS1 peptide. HBV could also efficiently infect HepaRG cells under non-adherent cell conditions. We screened several compounds using our culture system and identified proscillaridin A as a potent anti-HBV agent with an IC50 value of 7.2 nM. In conclusion, non-adherent host cell conditions of infection augmented HBV infectivity in an NTCP-dependent manner, thus providing a novel strategy to identify anti-HBV drugs and investigate the mechanism of HBV infection.


Subject(s)
Antiviral Agents/pharmacology , Organic Anion Transporters, Sodium-Dependent/genetics , Proscillaridin/pharmacology , Receptors, Virus/genetics , Symporters/genetics , Virus Internalization/drug effects , Bufanolides/pharmacology , Cell Adhesion , Digitoxin/pharmacology , Digoxin/pharmacology , Gene Expression , Hep G2 Cells , Hepatitis B virus/drug effects , Hepatitis B virus/physiology , High-Throughput Screening Assays , Humans , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Organic Anion Transporters, Sodium-Dependent/metabolism , Phthalazines/pharmacology , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/metabolism , Simvastatin/pharmacology , Strophanthins/pharmacology , Symporters/antagonists & inhibitors , Symporters/metabolism , Transgenes , Viral Envelope Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL