Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 513
Filter
1.
Clin Exp Pharmacol Physiol ; 46(6): 575-586, 2019 06.
Article in English | MEDLINE | ID: mdl-30854687

ABSTRACT

It has been established that lanatoside C, a FDA-approved cardiac glycoside, reduces proliferation of cancer cell lines. The proliferation of fibroblasts is critical to the pathogenesis of pulmonary fibrosis (PF), a progressive and fatal fibrotic lung disease lacking effective treatment. In this study we have investigated the impact of lanatoside C on a bleomycin (BLM)-induced mouse model of PF and through the evaluation of fibroblast proliferation and activation in vitro. We evaluated explanted lung tissue by histological staining, western blot analysis, qRT-PCR and survival analysis, demonstrating that lanatoside C was able to protect mice against BLM-induced pulmonary fibrosis. The proliferation of cultured pulmonary fibroblasts isolated from BLM-induced PF mice was suppressed by lanatoside C, as hypothesized, through the induction of cell apoptosis and cell cycle arrest at the G2/M phase. The Akt signalling pathway was involved in this process. Interestingly, the production of α-SMA, fibronectin, and collagen I and III in response to TGF-ß1 in healthy mouse fibroblasts was suppressed following lanatoside C administration by inhibition of TGF-ß1/Smad signalling. In addition, TGF-ß1-induced migration in lung fibroblasts was also impeded after lanatoside C treatment. Together, our data revealed that lanatoside C alleviated BLM-induced pulmonary fibrosis in mice via attenuation of growth and differentiation of fibroblasts, suggesting that it has potential as a candidate therapy for PF patients.


Subject(s)
Bleomycin/pharmacology , Cell Differentiation/drug effects , Fibroblasts/drug effects , Fibroblasts/pathology , Lanatosides/pharmacology , Pulmonary Fibrosis/pathology , Pulmonary Fibrosis/prevention & control , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Cyclin D1/metabolism , Cyclin E/metabolism , Cytoprotection/drug effects , Down-Regulation/drug effects , Fibroblasts/metabolism , Forkhead Box Protein O1/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
2.
Mol Vis ; 23: 346-355, 2017.
Article in English | MEDLINE | ID: mdl-28680263

ABSTRACT

PURPOSE: This study investigated the effect of cedilanid on retinal neovascularization in a mouse model of oxygen-induced retinopathy. METHODS: Seven-day-old C57BL/6 mice were exposed to 75% ± 1% oxygen for 5 days and were then returned to room air to induce retinal neovascularization. Cedilanid (0.025-0.2 µg) was intravitreally injected into the left eye of each mouse on postnatal day 12 (P12) and P15. PBS was intravitreally injected into the right eye as a control. Retinal neovascularization was evaluated with isolectin GS-IB4 staining of the retinal blood vessels. The function of reestablishment blood vessels was evaluated with angiography with the injection of fluorescein isothiocyanate (FITC)-dextran followed by isolectin GS-IB4 staining. Real time (RT)-PCR and western blot were used to examine the mRNA and protein expression of hypoxia inducible factor 1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF), respectively. RESULTS: Retinal neovascular areas and obliterative areas were statistically significantly smaller in the eyes injected with cedilanid (0.05 µg, 0.1 µg, and 0.2 µg) compared with the control eyes. The inhibitory effect of cedilanid was observed in a dose-dependent manner. In addition, the retinal neovascular areas and the obliterative areas in the eyes injected with 0.2 µg cedilanid on P12 were statistically significantly smaller than those in the eyes injected with the same dose of cedilanid on P15. Cedilanid promoted the circulative function of reestablished blood vessels in the obliterative areas. Cedilanid inhibited the expression of HIF-1α and VEGF in mice treated with hyperoxia. CONCLUSIONS: Cedilanid inhibits retinal neovascularization in a mouse model of oxygen-induced retinopathy. Early treatment with cedilanid produces better inhibition of retinal neovascularization. Cedilanid may be a potential treatment of neovascular diseases.


Subject(s)
Lanatosides/therapeutic use , Retinal Neovascularization/drug therapy , Animals , Animals, Newborn , Disease Models, Animal , Dose-Response Relationship, Drug , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred C57BL , Oxygen , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Time Factors , Vascular Endothelial Growth Factor A/metabolism
3.
Stem Cells ; 32(8): 2021-32, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24801379

ABSTRACT

The tumor-tropic properties of neural stem cells (NSCs) have been shown to serve as a novel strategy to deliver therapeutic genes to tumors. Recently, we have reported that the cardiac glycoside lanatoside C (Lan C) sensitizes glioma cells to the anticancer agent tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Here, we engineered an FDA-approved human NSC line to synthesize and secrete TRAIL and the Gaussia luciferase (Gluc) blood reporter. We showed that upon systemic injection, these cells selectively migrate toward tumors in the mice brain across the blood-brain barrier, target invasive glioma stem-like cells, and induce tumor regression when combined with Lan C. Gluc blood assay revealed that 30% of NSCs survived 1 day postsystemic injection and around 0.5% of these cells remained viable after 5 weeks in glioma-bearing mice. This study demonstrates the potential of systemic injection of NSCs to deliver anticancer agents, such as TRAIL, which yields glioma regression when combined with Lan C.


Subject(s)
Brain Neoplasms/pathology , Genetic Therapy/methods , Glioblastoma/pathology , Neural Stem Cells/transplantation , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , Animals , Apoptosis/genetics , Cardiac Glycosides/pharmacology , Cell Line, Tumor , Cell Movement/physiology , Coculture Techniques , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Lanatosides/pharmacology , Mice , Mice, Nude , TNF-Related Apoptosis-Inducing Ligand/genetics , Transfection , Xenograft Model Antitumor Assays
4.
Lung ; 192(1): 87-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24241792

ABSTRACT

BACKGROUND: The aim of this study was to investigate the gene expression profile of chronic obstructive pulmonary disease (COPD) patients and non-COPD patients. METHODS: Microarray raw data (GSE29133) was downloaded from Gene Expression Omnibus, including three COPD samples and three normal controls. Gene expression profiling was performed using Affymetrix human genome u133 plus 2.0 GeneChip. Differentially expressed genes were identified by Student's t test and genes with p < 0.05 were considered significantly changed. Up- and downregulated genes were submitted to the molecular signatures database (MSigDB) to search for a possible association with other previously published gene expression signatures. Furthermore, we constructed a COPD protein-protein interaction (PPI) network and used the connectivity map (cMap) to query for potential drugs for COPD. RESULTS: A total of 680 upregulated genes and 530 downregulated genes in COPD were identified. The MSigDB investigation found that upregulated genes were highly similar to gene signatures that respond to interferon and downregulated genes were similar to erythroid progenitor cells from fetal livers of E13.5 embryos with KLF1 knocked out. A PPI network consisting of 814 gene/proteins and 2,613 interactions was identified by Search Tool for the Retrieval of Interacting Genes. The cMap predicted helveticoside, disulfiram, and lanatoside C as the top three possible drugs that could perhaps treat COPD. CONCLUSION: Comprehensive analysis of the gene expression profile for COPD versus control reveals helveticoside, disulfiram, and lanatoside C as potential molecular targets in COPD. This evidence provides a new breakthrough in the medical treatment of patients with COPD.


Subject(s)
Protein Interaction Mapping , Protein Interaction Maps , Proteins/metabolism , Pulmonary Disease, Chronic Obstructive/metabolism , Case-Control Studies , Data Mining , Databases, Genetic , Digitalis Glycosides/therapeutic use , Disulfiram/therapeutic use , Drug Design , Gene Expression Profiling/methods , Gene Expression Regulation , Genetic Predisposition to Disease , Humans , Lanatosides/therapeutic use , Molecular Targeted Therapy , Oligonucleotide Array Sequence Analysis , Phenotype , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/genetics , Strophanthins/therapeutic use
5.
Phytomedicine ; 124: 155308, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38185069

ABSTRACT

BACKGROUND: In the past decades, extensive research has been conducted to identify new drug targets for the treatment of Herpes simplex virus type 1 (HSV-1) infections. However, the emergence of drug-resistant HSV-1 strains remains a major challenge. This necessitates the identification of new drugs with novel mechanisms of action. Lanatoside C (LanC), a cardiac glycoside (CG) approved by the US Food and Drug Administration (FDA), has demonstrated anticancer and antiviral properties. Nevertheless, its potential as an agent against HSV-1 infections and the underlying mechanism of action are currently unknown. PURPOSE: This study aimed to investigate the antiviral activity of LanC against HSV-1 and elucidate its molecular mechanisms. METHODS: The in vitro antiviral activity of LanC was assessed by examining the levels of viral genes, proteins, and virus titers in HSV-1-infected ARPE-19 and Vero cells. Immunofluorescence (IF) analysis was performed to determine the intracellular distribution of NRF2. Additionally, an in vivo mouse model of HSV-1 infection was developed to evaluate the antiviral activity of LanC, using indicators such as intraepidermal nerve fibers (IENFs) loss and viral gene inhibition. RESULTS: Our findings demonstrate that LanC significantly inhibits HSV-1 replication both in vitro and in vivo. The antiviral effect of LanC is mediated by the perinuclear translocation of NRF2. CONCLUSIONS: LanC exhibits anti-HSV-1 effects in viral infections, which are associated with the intracellular translocation of NRF2. These findings suggest that LanC has the potential to serve as a novel NRF2 modulator in the treatment of viral diseases.


Subject(s)
Herpesvirus 1, Human , Lanatosides , Chlorocebus aethiops , Animals , Mice , Vero Cells , NF-E2-Related Factor 2 , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Virus Replication
6.
J Org Chem ; 77(18): 7850-7, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22870937

ABSTRACT

Acylation of lanatoside C in the presence of organocatalyst 5 gave the C(4'''')-O-acylate in up to 90% regioselectivity (catalyst-controlled regioselectivity). Various functionalized acyl groups can be introduced at the C(4'''')-OH by a mixed anhydride method in the presence of 5 or the related organocatalyst. On the other hand, DMAP-catalyzed acylation of lanatoside C gave the C(3'''')-O-acylate in up to 97% regioselectivity (substrate-controlled regioselectivity). Thus, diverse regioselective introduction of acyl groups among eight free hydroxy groups of lanatoside C was achieved.


Subject(s)
Cardiac Glycosides/chemistry , Lanatosides/chemistry , Lanatosides/chemical synthesis , Acylation , Catalysis , Stereoisomerism
7.
Sci Rep ; 11(1): 12871, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145369

ABSTRACT

Lanatoside C has a promising anti-tumor activity and is a potential candidate for radiosensitizers. In this study, we have investigated the therapeutic efficacy of the combination of 131I-trastuzumab and lanatoside C for inhibition of human epidermal growth factor receptor 2 (HER2) positive tumor progression in NCI-N87 xenograft model. The combination treatment (131I-trastuzumab and lanatoside C) showed highest cytotoxicity when compared to non-treated control or trastuzumab alone or 131I alone or 131I-trastuzumab alone in vitro. Biodistribution studies using 131I-trastuzumab or combination of 131I-trastuzumab and lanatoside C showed tumor uptake in BALB/c nude mice bearing HER2 positive NCI-N87 tumor xenograft model. The higher tumor uptake was observed in 131I-trastuzumab (19.40 ± 0.04% ID/g) than in the combination of 131I-trastuzumab and lanatoside C (14.02 ± 0.02% ID/g) at 24 h post-injection. Most importantly, an antitumor effect was observed in mice that received the combination of 131I-trastuzumab and lanatoside C (p = 0.009) when compared to control. In addition, mice received lanatoside C alone (p = 0.085) or 131I-trastuzumab alone (p = 0.160) did not significantly inhibit tumor progression compared with control. Taken together, our data suggest that combination of 131I-trastuzumab and lanatoside C might be a potential synergistic treatment for radioimmunotherapy to control the HER2 positive tumor.


Subject(s)
Iodine Radioisotopes/administration & dosage , Lanatosides/pharmacology , Neoplasms/etiology , Neoplasms/therapy , Radioimmunotherapy , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Animals , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Humans , Iodine Radioisotopes/chemistry , Lanatosides/chemistry , Mice , Neoplasms/metabolism , Radioimmunotherapy/methods , Receptor, ErbB-2/genetics , Tissue Distribution , Trastuzumab/chemistry , Xenograft Model Antitumor Assays
8.
Neoplasia ; 23(12): 1213-1226, 2021 12.
Article in English | MEDLINE | ID: mdl-34768108

ABSTRACT

The 78 kilodalton glucose-regulated protein (GRP78) is a major endoplasmic reticulum (ER) molecular chaperone with antiapoptotic properties and a key regulator of the unfolded protein response (UPR). ER-stress induction of GRP78 in cancer cells represents a major pro-survival branch of the UPR. Pancreatic ductal adenocarcinoma (PDAC) remains a highly lethal disease and high level of GRP78 is associated with aggressive disease and poor survival. Recently, we reported that PDAC exhibited high level of ER stress and that GRP78 haploinsufficiency is sufficient to suppress pancreatic tumorigenesis in mice, suggesting the utility of inhibitors of GRP78 expression in combating pancreatic cancer. Screening of clinically relevant compound libraries revealed that cardiac glycosides (CGs) can inhibit ER-stress induction of GRP78 in pancreatic and other types of human cancers. Using the FDA-approved CG compound Lanatoside C (LanC) and human pancreatic cancer cell lines as model systems, we discovered that LanC preferably suppressed ER stress induction of GRP78 and to a lesser extent GRP94. The suppression is at the post-transcriptional level and dependent on the Na+/K+-ATPase ion pump. Overexpression of GRP78 mitigates apoptotic activities of LanC in ER stressed cells. Our study revealed a new function of CGs as inhibitor of stress induction of GRP78, and that this suppression at least in part contributes to the apoptotic activities of CGs in human pancreatic cancer cells in vitro. These findings support further investigation into CGs as potential antineoplastic agents for pancreatic and other cancers which depend on GRP78 for growth and survival.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Endoplasmic Reticulum Chaperone BiP/drug effects , Endoplasmic Reticulum Stress/drug effects , Lanatosides/pharmacology , Pancreatic Neoplasms/metabolism , Cardiac Glycosides/pharmacology , Cell Line, Tumor , Endoplasmic Reticulum Chaperone BiP/metabolism , Humans , Unfolded Protein Response/drug effects , Pancreatic Neoplasms
9.
Commun Biol ; 4(1): 260, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33637884

ABSTRACT

Neuroinflammation is a key component of virtually all neurodegenerative diseases, preceding neuronal loss and associating directly with cognitive impairment. Neuroinflammatory signals can originate and be amplified at barrier tissues such as brain vasculature, surrounding meninges and the choroid plexus. We designed a high content screening system to target inflammation in human brain-derived cells of the blood-brain barrier (pericytes and endothelial cells) to identify inflammatory modifiers. Screening an FDA-approved drug library we identify digoxin and lanatoside C, members of the cardiac glycoside family, as inflammatory-modulating drugs that work in blood-brain barrier cells. An ex vivo assay of leptomeningeal and choroid plexus explants confirm that these drugs maintain their function in 3D cultures of brain border tissues. These results suggest that cardiac glycosides may be useful in targeting inflammation at border regions of the brain and offer new options for drug discovery approaches for neuroinflammatory driven degeneration.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Blood-Brain Barrier/drug effects , Choroid Plexus/drug effects , Digoxin/pharmacology , Endothelial Cells/drug effects , Inflammation/drug therapy , Lanatosides/pharmacology , Meninges/drug effects , Pericytes/drug effects , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Cells, Cultured , Choroid Plexus/metabolism , Choroid Plexus/pathology , Drug Evaluation, Preclinical , Endothelial Cells/metabolism , Endothelial Cells/pathology , High-Throughput Screening Assays , Humans , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Meninges/metabolism , Meninges/pathology , Pericytes/metabolism , Pericytes/pathology , Tissue Culture Techniques
10.
Biomolecules ; 9(12)2019 11 27.
Article in English | MEDLINE | ID: mdl-31783627

ABSTRACT

Cardiac glycosides (CGs) are a diverse family of naturally derived compounds having a steroid and glycone moiety in their structures. CG molecules inhibit the α-subunit of ubiquitous transmembrane protein Na+/K+-ATPase and are clinically approved for the treatment of cardiovascular diseases. Recently, the CGs were found to exhibit selective cytotoxic effects against cancer cells, raising interest in their use as anti-cancer molecules. In this current study, we explored the underlying mechanism responsible for the anti-cancer activity of Lanatoside C against breast (MCF-7), lung (A549), and liver (HepG2) cancer cell lines. Using Real-time PCR, western blot, and immunofluorescence studies, we observed that (i) Lanatoside C inhibited cell proliferation and induced apoptosis in cell-specific and dose-dependent manner only in cancer cell lines; (ii) Lanatoside C exerts its anti-cancer activity by arresting the G2/M phase of cell cycle by blocking MAPK/Wnt/PAM signaling pathways; (iii) it induces apoptosis by inducing DNA damage and inhibiting PI3K/AKT/mTOR signaling pathways; and finally, (iv) molecular docking analysis shows significant evidence on the binding sites of Lanatoside C with various key signaling proteins ranging from cell survival to cell death. Our studies provide a novel molecular insight of anti-cancer activities of Lanatoside C in human cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Lanatosides/pharmacology , Neoplasms/metabolism , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , Wnt Proteins/antagonists & inhibitors , A549 Cells , Cell Survival/drug effects , DNA Damage , G2 Phase Cell Cycle Checkpoints/drug effects , Hep G2 Cells , Humans , M Phase Cell Cycle Checkpoints/drug effects , MCF-7 Cells , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinases/metabolism , Signal Transduction/drug effects , Wnt Proteins/metabolism
11.
Biochem Pharmacol ; 150: 280-292, 2018 04.
Article in English | MEDLINE | ID: mdl-29475060

ABSTRACT

Gastric cancer is the third common cause of cancer mortality in the world with poor prognosis and high recurrence due to lack of effective medicines. Our studies revealed that lanatoside C, a FDA-approved cardiac glycoside, had an anti-proliferation effect on different human cancer cell lines (MKN-45; SGC-7901; HN4; MCF-7; HepG2) and gastric cell lines MKN-45 and SGC-7901 were the most sensitive cell lines to lanatoside C. MKN-45 cells treated with lanatoside C showed cell cycle arrest at G2/M phase and inhibition of cell migration. Meanwhile, upregulation of cleaved caspase-9 and cleaved PARP and downregulation of Bcl-xl were accompanied with the loss of mitochondrial membrane potential (MMP) and induction of intracellular reactive oxygen species (ROS). Lanatoside C inhibited Wnt/ß-catenin signaling with downregulation of c-Myc, while overexpression of c-Myc reversed the anti-tumor effect of lanatoside C, confirming that c-Myc is a key drug target of lanatoside C. Furthermore, we discovered that lanatoside C prompted c-Myc degradation in proteasome-ubiquitin pathway with attenuating the binding of USP28 to c-Myc. These findings indicate that lanatoside C targeted c-Myc ubiquitination to inhibit MKN-45 proliferation and support the potential value of lanatoside C as a chemotherapeutic candidate.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , DNA-Binding Proteins/metabolism , Lanatosides/pharmacology , Stomach Neoplasms/metabolism , Transcription Factors/metabolism , Wnt Signaling Pathway/physiology , Apoptosis/drug effects , Cell Proliferation/drug effects , DNA-Binding Proteins/antagonists & inhibitors , Dose-Response Relationship, Drug , HEK293 Cells , Hep G2 Cells , Humans , Lanatosides/therapeutic use , MCF-7 Cells , Stomach Neoplasms/drug therapy , Transcription Factors/antagonists & inhibitors , Wnt Signaling Pathway/drug effects
12.
Sci Rep ; 7: 46134, 2017 04 07.
Article in English | MEDLINE | ID: mdl-28387249

ABSTRACT

Recent studies have revealed that cardiac glycosides, such as digitalis and digoxin, have anticancer activity and may serve as lead compounds for the development of cancer treatments. The poor prognosis of hepatocellular carcinoma (HCC) patients reflects the development of resistance to current chemotherapeutic agents, highlighting the need for discovering new small-molecule therapeutics. Here, we found that lanatoside C, an anti-arrhythmic agent extracted from Digitalis lanata, inhibited the growth of HCC cells and dramatically decreased tumor volume as well as delayed tumor growth without obvious body weight loss. Moreover, lanatoside C triggered mitochondrial membrane potential (MMP) loss, activation of caspases and translocation of apoptosis-inducing factor (AIF) into the nucleus, which suggests that lanatoside C induced apoptosis through both caspase-dependent and -independent pathways. Furthermore, we discovered that lanatoside C activated protein kinase delta (PKCδ) via Thr505 phosphorylation and subsequent membrane translocation. Inhibition of PKCδ reversed lanatoside C-induced MMP loss and apoptosis, confirming that lanatoside C caused apoptosis through PKCδ activation. We also found that the AKT/mTOR pathway was negatively regulated by lanatoside C through PKCδ activation. In conclusion, we provide the first demonstration that the anticancer effects of lanatoside C are mainly attributable to PKCδ activation.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cardiac Glycosides/pharmacology , Lanatosides/pharmacology , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Protein Kinase C-delta/metabolism , Animals , Cardiac Glycosides/chemistry , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Humans , Lanatosides/chemistry , Membrane Potential, Mitochondrial/drug effects , Mice, SCID , Mitochondrial Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
13.
Chin Med Sci J ; 21(1): 16-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16615278

ABSTRACT

OBJECTIVE: To investigate the effectiveness and safety of various agents on paroxysmal atrial fibrillation in the elderly over 75 years old. METHODS: Totally 264 in-patients (75-91 years old, 185 males and 79 females) with atrial fibrillation history of less than 7 days were enrolled in this study. A total of 611 atrial fibrillation episodes were recorded, but 130 episodes (22.3%) of atrial fibrillation were auto-converted to sinus rhythm. The rest 481 episodes of atrial fibrillation were divided into six groups based on the drug used. RESULTS: The cardioversion ratio of atrial fibrillation were 9.5%, 46.9%, 71.7%, 55.9%, 32.7%, and 73.6% in control, cedilanid, amiodarone, propafenone, verapamil, and quinidine groups, respectively. Ventricular rate control were 5.4%, 83.6%, 84.9%, 77.9%, 78.8%, and 11.3% in those groups, respectively. The total effective rates of amiodarone and cedilanid groups were the highest. When the ventricular rate was controlled to below 90 bpm, the patients would almost complain of no discomfort. No severe side-effect was observed in each group. CONCLUSION: Amiodarone and cedilanid may be the proper drugs for the treatment of paroxysmal atrial fibrillation in the elderly. The above antiarrhythmics in each therapeutic group were relatively safe and effective.


Subject(s)
Amiodarone/therapeutic use , Anti-Arrhythmia Agents/therapeutic use , Atrial Fibrillation/drug therapy , Lanatosides/therapeutic use , Aged , Aged, 80 and over , Amiodarone/adverse effects , Anti-Arrhythmia Agents/adverse effects , Bradycardia/chemically induced , Cardiac Glycosides/adverse effects , Cardiac Glycosides/therapeutic use , Female , Heart Rate/drug effects , Humans , Lanatosides/adverse effects , Male , Nausea/chemically induced , Propafenone/adverse effects , Propafenone/therapeutic use
14.
Sci Rep ; 6: 20154, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26821916

ABSTRACT

Lanatoside C's impact on atherosclerosis is poorly understood. The present study was conducted to determine whether lanatoside C affects the development of atherosclerosis in apolipoprotein E-deficient (ApoE(-/-)) mice. ApoE(-/-) mice were administered either phosphate-buffered saline (PBS) containing 0.1% DMSO (the vehicle control group) or lanatoside C at low (1 mg/kg per day) or high (2 mg/kg per day) doses, and fed a Western diet for 12 weeks. Lanatoside C dose-dependently aggravated the development of atherosclerosis in the ApoE(-/-) mice compared with the vehicle control group. In an effort to determine the mechanism by which lanatoside C increased atherosclerosis, we found that lanatoside C significantly promoted the uptake of oxidised low-density lipoprotein (oxLDL) and increased foam-cell formation by upregulation of scavenger receptor class A (SR-A) and the class B scavenger receptor (CD36) in macrophages. Meanwhile, the effects of lanatoside C were abolished using small interfering RNA (siRNA) inhibition of peroxisome proliferator-activated receptors ß/δ (PPARß/δ). Overall, our data demonstrate that lanatoside C aggravates the development of atherosclerosis by inducing PPARß/δ expression, which mediates upregulation of SR-A and CD36, and promotes oxLDL uptake and foam-cell formation.


Subject(s)
Atherosclerosis/metabolism , Foam Cells/metabolism , Lanatosides/toxicity , Macrophages, Peritoneal/metabolism , Animals , Apolipoproteins E/deficiency , Atherosclerosis/chemically induced , Atherosclerosis/genetics , Atherosclerosis/pathology , Diet, Western/adverse effects , Foam Cells/pathology , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Knockout
15.
Phytomedicine ; 23(1): 42-51, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26902406

ABSTRACT

BACKGROUND: Hepatocellular carcinoma is the second deadliest cancer with limited treatment options. Loss of PTEN causes the P13K/Akt pathway to be hyperactive which contributes to cell survival and resistance to therapeutics in various cancers, including the liver cancer. Hence molecules targeting this pathway present good therapeutic strategies for liver cancer. HYPOTHESIS: It was previously reported that Cardiac glycosides possessed antitumor activity by inducing apoptosis of multiple cancer cells through oxidative stress. However, whether Cardiac glycoside Lanatoside C can induce oxidative stress in liver cancer cells and induce cell death both in vitro and in vivo remains unknown. METHODS: Cell viability was measured by SRB assay. Cell death analysis was investigated by propidium iodide staining with flow cytometry and PARP cleavage. DCFH-DA staining and cytometry were used for intracellular ROS measurement. Protein levels were analyzed by western blot analysis. Antitumor activity was investigated on mice xenografts in vivo. RESULTS: In this study, we found that Cardiac glycosides, particularly Lanatoside C from Digitalis ferruginea could significantly inhibit PTEN protein adequate Huh7 and PTEN deficient Mahlavu human liver cancer cell proliferation by the induction of apoptosis and G2/M arrest in the cells. Lanatoside C was further shown to induce oxidative stress and alter ERK and Akt pathways. Consequently, JNK1 activation resulted in extrinsic apoptotic pathway stimulation in both cells while JNK2 activation involved in the inhibition of cell survival only in PTEN deficient cells. Furthermore, nude mice xenografts followed by MRI showed that Lanatoside C caused a significant decrease in the tumor size. In this study apoptosis induction by Lanatoside C was characterized through ROS altered ERK and Akt pathways in both PTEN adequate epithelial and deficient mesenchymal liver cancer cells. CONCLUSION: The results indicated that Lanatoside C could be contemplated in liver cancer therapeutics, particularly in PTEN deficient tumors. This is due to Lanatoside C's stress inducing action on ERK and Akt pathways through differential activation of JNK1 and JNK2 by GSK3ß.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Lanatosides/pharmacology , Liver Neoplasms/pathology , PTEN Phosphohydrolase/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Digitalis/chemistry , Humans , Mice , Mice, Nude , Oxidative Stress , Signal Transduction , Xenograft Model Antitumor Assays
16.
Oncotarget ; 7(5): 6074-87, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26756216

ABSTRACT

Cardiac glycosides are clinically used for cardiac arrhythmias. In this study, we investigated the mechanism responsible for anti-cancer and radiosensitizing effects of lanatoside C in colorectal cancer cells. Lanatoside C-treated cells showed classic patterns of autophagy, which may have been caused by lanatoside C-induced mitochondrial aggregation or degeneration. This mitochondrial dysfunction was due to disruption of K+ homeostasis, possibly through inhibition of Na+/K+-ATPase activity. In addition, lanatoside C sensitized HCT116 cells (but not HT-29 cells) to radiation in vitro. γ-H2AX, a representative marker of DNA damage, were sustained longer after combination of irradiation with lanatoside C, suggesting lanatoside C impaired DNA damage repair processes. Recruitment of 53BP1 to damaged DNA, a critical initiation step for DNA damage repair signaling, was significantly suppressed in lanatoside C-treated HCT116 cells. This may have been due to defects in the RNF8- and RNF168-dependent degradation of KDM4A/JMJD2A that increases 53BP1 recruitment to DNA damage sites. Although lanatoside C alone reduced tumor growth in the mouse xenograft tumor model, combination of lanatoside C and radiation inhibited tumor growth more than single treatments. Thus, lanatoside C could be a potential molecule for anti-cancer drugs and radiosensitizing agents.


Subject(s)
Colorectal Neoplasms/drug therapy , DNA Repair/drug effects , Lanatosides/pharmacology , Mitochondria/drug effects , Animals , Autophagy/drug effects , Cell Growth Processes/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA Damage , HCT116 Cells , HT29 Cells , Humans , Mice , Mitochondria/genetics , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Random Allocation , Signal Transduction
17.
Mol Oncol ; 10(4): 625-34, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26708508

ABSTRACT

Glioblastoma (GBM) is the most common malignant brain tumor in adults. We designed an adeno-associated virus (AAV) vector for intracranial delivery of secreted, soluble tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL) to GBM tumors in mice and combined it with the TRAIL-sensitizing cardiac glycoside, lanatoside C (lan C). We applied this combined therapy to two different GBM models using human U87 glioma cells and primary patient-derived GBM neural spheres in culture and in orthotopic GBM xenograft models in mice. In U87 cells, conditioned medium from AAV2-sTRAIL expressing cells combined with lan C induced 80% cell death. Similarly, lan C sensitized primary GBM spheres to sTRAIL causing over 90% cell death. In mice bearing intracranial U87 tumors treated with AAVrh.8-sTRAIL, administration of lan C caused a decrease in tumor-associated Fluc signal, while tumor size increased within days of stopping the treatment. Another round of lan C treatment re-sensitized GBM tumor to sTRAIL-induced cell death. AAVrh.8-sTRAIL treatment alone and combined with lanatoside C resulted in a significant decrease in tumor growth and longer survival of mice bearing orthotopic invasive GBM brain tumors. In summary, AAV-sTRAIL combined with lanatoside C induced cell death in U87 glioma cells and patient-derived GBM neural spheres in culture and in vivo leading to an increased in overall mice survival.


Subject(s)
Brain Neoplasms , Dependovirus , Genetic Vectors , Glioblastoma , Lanatosides/pharmacology , Neoplasms, Experimental , TNF-Related Apoptosis-Inducing Ligand , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Glioblastoma/therapy , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , TNF-Related Apoptosis-Inducing Ligand/biosynthesis , TNF-Related Apoptosis-Inducing Ligand/genetics , Xenograft Model Antitumor Assays
18.
Curr Opin Chem Biol ; 8(5): 498-507, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15450492

ABSTRACT

Recent reports have provided the first insights into the mechanisms of the extensive post-translational modifications involved in the biosynthesis of the lantibiotics, a class of peptide antimicrobial agents. These modifications involve dehydration of several serine and threonine residues followed by intramolecular conjugate additions of cysteines, resulting in extensively cross-linked polycyclic structures. Both in vivo and in vitro studies indicate low substrate specificity of the modification machinery, which has been explored for re-engineering of the structures of a number of members. In addition to these developments in understanding their biosynthesis, studies on the mode of action of several lantibiotics have shown a unique mechanism of binding to lipid II, an intermediate in cell wall biosynthesis.


Subject(s)
Anti-Bacterial Agents/metabolism , Peptides/metabolism , Protein Processing, Post-Translational , Amino Acid Sequence , Bacteriocins , Cell Wall/metabolism , Cysteine/chemistry , Cysteine/metabolism , Lanatosides/chemistry , Lanatosides/metabolism , Lipid Metabolism , Molecular Sequence Data , Serine/chemistry , Serine/metabolism , Substrate Specificity , Threonine/chemistry , Threonine/metabolism
19.
Klin Med (Mosk) ; 83(7): 59-63, 2005.
Article in Russian | MEDLINE | ID: mdl-16117429

ABSTRACT

The authors of the article studied potential of various drug combinations (atenolol + enalapril + indapamide, and celanid + enalapril + indapamide) in complex treatment of patients with heart failure (HF) and supraventricular arrhythmias. The subjects were 106 patients, of whom 74 had coronary heart disease, of whom 51 had old myocardial infarction, and 18 had undergone coronary artery bypass grafting. Dilatation cardiomyopathy was found in 8, aortomitral valvular disease--in 18, and combined mitral valvular disease--in 6 patients. 36 subjects had permanent atrial fibrillation, 48--a paroxysmal form of ciliary arrhythmia, 22--paroxysms of atrial tachycardia. NYHA functional class (FC) II HF was diagnosed in 64 patients, NYHA FC III HF--in 42 patients. According to what treatment regimen was applied, the patients were divided into two groups, comparable in the main clinical and functional characteristics. Two-month therapy resulted in improvement of hemodynamics, increase of activity tolerance and improvement of life quality in both groups. The combination celanide + enalapril + indapamide in individual doses was effective in FC II-III HF with supraventricular arrhythmias. The cost of treatment with celanid is lower.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Anti-Arrhythmia Agents/therapeutic use , Cardiac Glycosides/therapeutic use , Diuretics/therapeutic use , Heart Failure/drug therapy , Tachycardia, Supraventricular/drug therapy , Aged , Atenolol/therapeutic use , Blood Pressure/drug effects , Drug Therapy, Combination , Echocardiography , Electrocardiography , Enalapril/therapeutic use , Follow-Up Studies , Heart Failure/complications , Heart Failure/physiopathology , Heart Rate/drug effects , Humans , Indapamide/therapeutic use , Lanatosides/therapeutic use , Male , Middle Aged , Tachycardia, Supraventricular/complications , Tachycardia, Supraventricular/physiopathology , Treatment Outcome
20.
Cardiovasc Res ; 17(10): 633-41, 1983 Oct.
Article in English | MEDLINE | ID: mdl-6414704

ABSTRACT

Several studies in animals and in man have suggested that the inhibitory influence of baroreceptors on heart rate and peripheral circulation is enhanced by digitalis. Because the atrio-ventricular node represents a key site for the clinical action of digitalis we studied how baroreceptor control of atrio-ventricular conduction is modified by digitalis at therapeutical doses. In eight subjects heart rate was kept constant by atrial pacing to assess neural influences on atrio-ventricular conduction rate without the modifications caused by simultaneous changes in cardiac cycle length. Arterial baroreceptors were stimulated by increasing or reducing blood pressure (intra-arterial recording), via an iv bolus of phenylephrine or nitroglycerine. The baroreflex sensitivity was assessed in ms . mmHg-1 as the slope of the linear regressions relating the rise or fall in systolic blood pressure to the lengthening or shortening in St- (atrial stimulus artifact) Q interval (ECG recording). The study was performed before and 45 min after iv administration of digitalis (0.8 mg of Lanatoside C). Baroreflex sensitivity during baroreceptor stimulation was 2.9 +/- 1.1 ms . mmHg-1 (mean +/- SE) before digitalis, whereas after digitalis a significantly and markedly greater value of 5.6 +/- 1.5 ms . mmHg-1 was found. Baroreflex sensitivity during baroreceptor deactivation was 0.9 +/- 0.1 ms . mmHg-1 before digitalis, and was not significantly affected by the drug. Thus in man the baroreceptor control of atrio-ventricular conduction is strikingly potentiated by digitalis although this potentiation is only evident in the upper portion of the stimulus-response curve of the reflex.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Atrioventricular Node/physiology , Heart Conduction System/physiology , Lanatosides/pharmacology , Pressoreceptors/drug effects , Adult , Blood Pressure/drug effects , Female , Humans , Hypertension/physiopathology , Male , Nitroglycerin/pharmacology , Phenylephrine/pharmacology , Pressoreceptors/physiology , Reflex/drug effects , Sinoatrial Node/physiology
SELECTION OF CITATIONS
SEARCH DETAIL