Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Org Biomol Chem ; 16(9): 1489-1495, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29411820

ABSTRACT

Maternal embryonic leucine zipper kinase (MELK), a serine/threonine protein kinase, has oncogenic properties and plays a key functional role in various cancer cells. Although MELK may not be a cancer addiction target, the development of specific MELK inhibitors would provide useful chemical tools for synthetic lethal investigation. Herein, we identified several hit compounds using a customized structure-based virtual screening, among which compounds 4 and 16 showed the most potent inhibition to MELK with IC50 values of 3.52 µM and 178.3 nM, respectively. In vitro cell-based assays revealed that 16 has no effect on the growth of various types of cancer cells, but has the potential to inhibit cancer cell migration and invasion. Western blotting analyses revealed that 16 suppresses the phosphorylation of focal adhesion kinase (FAK), a downstream molecule of MELK, which is a key kinase in regulating cancer cell migration and invasion.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Discovery , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Cell Line, Tumor , Cell Movement/drug effects , Focal Adhesion Kinase 1/metabolism , Humans , Neoplasm Invasiveness , Signal Transduction/drug effects
2.
Int J Mol Sci ; 17(2): 228, 2016 Feb 08.
Article in English | MEDLINE | ID: mdl-26867193

ABSTRACT

Plant lectins have been investigated to elucidate their complicated mechanisms due to their remarkable anticancer activities. Although plant lectins seems promising as a potential anticancer agent for further preclinical and clinical uses, further research is still urgently needed and should include more focus on molecular mechanisms. Herein, a Naïve Bayesian model was developed to predict the protein-protein interaction (PPI), and thus construct the global human PPI network. Moreover, multiple sources of biological data, such as smallest shared biological process (SSBP), domain-domain interaction (DDI), gene co-expression profiles and cross-species interolog mapping were integrated to build the core apoptotic PPI network. In addition, we further modified it into a plant lectin-induced apoptotic cell death context. Then, we identified 22 apoptotic hub proteins in mesothelioma cells according to their different microarray expressions. Subsequently, we used combinational methods to predict microRNAs (miRNAs) which could negatively regulate the abovementioned hub proteins. Together, we demonstrated the ability of our Naïve Bayesian model-based network for identifying novel plant lectin-treated cancer cell apoptotic pathways. These findings may provide new clues concerning plant lectins as potential apoptotic inducers for cancer drug discovery.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Neoplasms/metabolism , Plant Lectins/pharmacology , Signal Transduction/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cluster Analysis , Computational Biology , Databases, Protein , Drug Discovery , Gene Expression Profiling , Gene Ontology , Humans , MicroRNAs/genetics , Models, Biological , Neoplasms/genetics , Protein Interaction Mapping , Protein Interaction Maps , RNA Interference , RNA, Messenger/genetics
3.
Z Naturforsch C J Biosci ; 70(1-2): 7-13, 2015.
Article in English | MEDLINE | ID: mdl-25854839

ABSTRACT

Lectins, a group of highly diverse proteins of non-immune origin and are ubiquitously distributed in plants, animals and fungi, have multiple significant biological functions, such as anti-fungal, anti-viral and, most notably, anti-tumor activities. A lectin was purified from the rhizomes of Aspidistra elatior Blume, named A. elatior lectin (AEL). In vitro experiments showed that the minimum inhibitory concentrations of AEL against the vesicular stomatitis virus, Coxsackie virus B4, and respiratory syncytial virus were all the same at about 4 µg/mL. However, AEL was ineffective against the Sindbis virus and reovirus-1. AEL also showed significant in vitro antiproliferative activity towards Bre-04, Lu-04, HepG2, and Pro-01 tumor cell lines by increasing the proportion of their sub-G1 phase. However, AEL failed to restrict the proliferation of the HeLa cell line. Western blotting indicated that AEL induced the upregulation of cell cycle-related proteins p53 and p21. The molecular basis and species-specific effectiveness of the anti-proliferative and anti-viral potential of AEL are discussed.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antiviral Agents/pharmacology , Liliaceae , Plant Extracts/pharmacology , Plant Lectins/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Antiviral Agents/isolation & purification , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , HeLa Cells , Hep G2 Cells , Humans , Liliaceae/chemistry , Microbial Sensitivity Tests , Neoplasms/metabolism , Neoplasms/pathology , Phytotherapy , Plant Extracts/isolation & purification , Plant Lectins/isolation & purification , Plants, Medicinal , Rhizome , Time Factors , Tumor Suppressor Protein p53/metabolism , Viruses/drug effects , Viruses/growth & development
4.
Apoptosis ; 19(1): 1-18, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24081390

ABSTRACT

Green tea catechins have been extensively studied for their cancer preventive effects. Accumulating evidence has shown that green tea catechins, like (-)-epigallocatechin-3-gallate, have strong anti-oxidant activity and affect several signal transduction pathways relevant to cancer development. Here, we review the biological properties of green tea catechins and the molecular mechanisms of their anticancer effects, including the suppression of cancer cell proliferation, induction of apoptosis, and inhibition of tumor metastasis and angiogenesis. We summarize the efficacy of a single catechin and the synergetic effects of multiple catechins. We also discuss the enhanced anticancer effects of green tea catechins when they are combined with anticancer drugs. The information present in this review might promote the development of strategy for the co-administration of green tea catechins with other anticancer drugs to increase the potency of currently available anticancer medicine. This new strategy should in turn lower the cytotoxicity and cost of anticancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Camellia sinensis/chemistry , Catechin/administration & dosage , Neoplasms/drug therapy , Plant Extracts/administration & dosage , Animals , Apoptosis/drug effects , Humans , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/physiopathology
5.
Acta Pharmacol Sin ; 35(2): 248-56, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24362332

ABSTRACT

AIM: Proteins with legume lectin domains are known to possess a wide range of biological functions. Here, the antitumor effects of two representative legume lectins, concanavalin A (ConA) and Sophora flavescens lectin (SFL), on human breast carcinoma cells were investigated in vitro and in vivo. METHODS: Human breast carcinoma MCF-7 cells and human normal mammary epithelial MCF-10A cells were examined. Cell viability was detected using WST-1 and CCK-8 assays. Cell apoptosis was analyzed with Hoechst 33258 staining. Cell cycle was investigated using flow cytometry. The expression of relevant proteins was measured using Western blotting. Breast carcinoma MCF-7 bearing nude mice were used to study the antitumor effects in vivo. The mice were injected with ConA (40 mg/kg, ip) and SFL (55 mg/kg, ip) daily for 14 d. RESULTS: ConA and SFL inhibited the growth of MCF-7 cells in dose- and time-dependent manners (IC50 values were 15 and 20 µg/mL, respectively). Both ConA and SFL induced apoptotic morphology in MCF-7 cells without affecting MCF-10A cells. ConA and SFL dose-dependently increased the sub-G1 proportion in MCF-7 cells, while SFL also triggered the G2/M phase cell cycle arrest. Both ConA and SFL dose-dependently increased the activities of caspase-3 and caspase-9 and release of cytochrome C from mitochondria into cytoplasm, up-regulated Bax and Bid, and down-regulated Bcl-2 and Bcl-XL in MCF-7 cells. ConA reduced NF-κB, ERK, and JNK levels, and increased p53 and p21 levels, while SFL caused similar changes in NF-κB, ERK, p53, and p21 levels, but did not affect JNK expression. Administration of ConA and SFL significantly decreased the subcutaneous tumor mass volume and weight in MCF-7 bearing nude mice. CONCLUSION: ConA and SFL exert anti-tumor actions against human breast carcinoma MCF-7 cells both in vitro and in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Concanavalin A/pharmacology , Plant Lectins/pharmacology , Sophora/metabolism , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Female , Humans , MCF-7 Cells
6.
J Formos Med Assoc ; 113(3): 143-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24342026

ABSTRACT

The newly emerged Middle East respiratory syndrome coronavirus (MERS-CoV) is becoming another "SARS-like" threat to the world. It has an extremely high death rate (∼50%) as there is no vaccine or efficient therapeutics. The identification of the structures of both the MERS-CoV receptor binding domain (RBD) and its complex with dipeptidyl peptidase 4 (DPP4), raises the hope of alleviating this currently severe situation. In this review, we examined the molecular basis of the RBD-receptor interaction to outline why/how could we use MERS-CoV RBD to develop vaccines and antiviral drugs.


Subject(s)
Antiviral Agents/chemistry , Coronavirus Infections/virology , Coronavirus/immunology , Dipeptidyl Peptidase 4/immunology , Drug Design , Receptors, Virus/immunology , Viral Vaccines/chemistry , Antiviral Agents/therapeutic use , Coronavirus/chemistry , Coronavirus/metabolism , Coronavirus Infections/drug therapy , Coronavirus Infections/immunology , Coronavirus Infections/prevention & control , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/metabolism , Humans , Receptors, Virus/chemistry , Receptors, Virus/metabolism
7.
Int J Mol Sci ; 15(5): 9016-35, 2014 May 20.
Article in English | MEDLINE | ID: mdl-24853132

ABSTRACT

Mangiferin, a xanthonoid found in plants including mangoes and iris unguicularis, was suggested in previous studies to have anti-hyperglycemic function, though the underlying mechanisms are largely unknown. This study was designed to determine the therapeutic effect of mangiferin by the regeneration of ß-cells in mice following 70% partial pancreatectomy (PPx), and to explore the mechanisms of mangiferin-induced ß-cell proliferation. For this purpose, adult C57BL/6J mice after 7-14 days post-PPx, or a sham operation were subjected to mangiferin (30 and 90 mg/kg body weight) or control solvent injection. Mangiferin-treated mice exhibited an improved glycemia and glucose tolerance, increased serum insulin levels, enhanced ß-cell hyperplasia, elevated ß-cell proliferation and reduced ß-cell apoptosis. Further dissection at the molecular level showed several key regulators of cell cycle, such as cyclin D1, D2 and cyclin-dependent kinase 4 (Cdk4) were significantly up-regulated in mangiferin-treated mice. In addition, critical genes related to ß-cell regeneration, such as pancreatic and duodenal homeobox 1 (PDX-1), neurogenin 3 (Ngn3), glucose transporter 2 (GLUT-2), Forkhead box protein O1 (Foxo-1), and glucokinase (GCK), were found to be promoted by mangiferin at both the mRNA and protein expression level. Thus, mangiferin administration markedly facilitates ß-cell proliferation and islet regeneration, likely by regulating essential genes in the cell cycle and the process of islet regeneration. These effects therefore suggest that mangiferin bears a therapeutic potential in preventing and/or treating the diabetes.


Subject(s)
Insulin-Secreting Cells/cytology , Regeneration/drug effects , Up-Regulation/drug effects , Xanthones/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin D2/genetics , Cyclin D2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Glucose/metabolism , Insulin/blood , Insulin-Secreting Cells/metabolism , Islets of Langerhans/physiology , Islets of Langerhans/surgery , Male , Mice , Mice, Inbred C57BL
8.
Glycoconj J ; 30(3): 269-79, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22893111

ABSTRACT

Galanthus nivalis agglutinin (GNA)-related lectin family, a superfamily of strictly mannose-binding specific lectins widespread among monocotyledonous plants, is well-known to possess a broad range of biological functions such as anti-tumor, anti-viral and anti-fungal activities. Herein, we mainly focused on exploring the precise molecular mechanisms by which GNA-related lectins induce cancer cell apoptotic and autophagic death targeting mitochondria-mediated ROS-p38-p53 apoptotic or autophagic pathway, Ras-Raf and PI3K-Akt anti-apoptotic or anti-autophagic pathways. In addition, we further discussed the molecular mechanisms of GNA-related lectins exerting anti-viral activities by blocking the entry of the virus into its target cells, preventing transmission of the virus as well as forcing virus to delete glycan in its envelope protein and triggering neutralizing antibody. In conclusion, these findings may provide a new perspective of GNA-related lectins as potential drugs for cancer and virus therapeutics in the future.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antiviral Agents/pharmacology , Galanthus/chemistry , Plant Lectins/pharmacology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antiviral Agents/chemistry , Apoptosis/drug effects , Autophagy/drug effects , Cell Transformation, Neoplastic/drug effects , Humans , Plant Lectins/chemistry , Virus Physiological Phenomena/drug effects
9.
Acta Pharmacol Sin ; 34(5): 612-24, 2013 May.
Article in English | MEDLINE | ID: mdl-23564085

ABSTRACT

Autophagy, an evolutionarily conserved catabolic process involving the engulfment and degradation of non-essential or abnormal cellular organelles and proteins, is crucial for homeostatic maintenance in living cells. This highly regulated, multi-step process has been implicated in diverse diseases including cancer. Autophagy can function as either a promoter or a suppressor of cancer, which makes it a promising and challenging therapeutic target. Herein, we overview the regulatory mechanisms and dual roles of autophagy in cancer. We also describe some of the representative agents that exert their anticancer effects by regulating autophagy. Additionally, some emerging strategies aimed at modulating autophagy are discussed as having the potential for future anticancer drug discovery. In summary, these findings will provide valuable information to better utilize autophagy in the future development of anticancer therapeutics that meet clinical requirements.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Drug Discovery/methods , Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Humans , MicroRNAs/genetics , Neoplasms/genetics , Neoplasms/metabolism
10.
Cell Mol Life Sci ; 69(7): 1125-36, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22080117

ABSTRACT

Microautophagy, the non-selective lysosomal degradative process, involves direct engulfment of cytoplasmic cargo at a boundary membrane by autophagic tubes, which mediate both invagination and vesicle scission into the lumen. With its constitutive characteristics, microautophagy of soluble substrates can be induced by nitrogen starvation or rapamycin via regulatory signaling complex pathways. The maintenance of organellar size, membrane homeostasis, and cell survival under nitrogen restriction are the main functions of microautophagy. In addition, microautophagy is coordinated with and complements macroautophagy, chaperone-mediated autophagy, and other self-eating pathways. Three forms of selective microautophagy, including micropexophagy, piecemeal microautophagy of the nucleus, and micromitophagy, share common ground with microautophagy to some degree. As the accumulation of experimental data, the precise mechanisms that govern microautophagy are becoming more appreciated. Here, we review the microautophagic molecular machinery, its physiological functions, and relevance to human diseases, especially in diseases involving multivesicular bodies and multivesicular lysosomes.


Subject(s)
Autophagy , Animals , Cytoplasmic Vesicles/metabolism , Homeostasis , Humans , Signal Transduction
11.
Chin J Cancer ; 32(3): 113-20, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22835386

ABSTRACT

Autophagy, an evolutionarily conserved lysosomal degradation process, has drawn an increasing amount of attention in recent years for its role in a variety of human diseases, such as cancer. Notably, autophagy plays an important role in regulating several survival and death signaling pathways that determine cell fate in cancer. To date, substantial evidence has demonstrated that some key autophagic mediators, such as autophagy-related genes (ATGs), PI3K, mTOR, p53, and Beclin-1, may play crucial roles in modulating autophagic activity in cancer initiation and progression. Because autophagy-modulating agents such as rapamycin and chloroquine have already been used clinically to treat cancer, it is conceivable that targeting autophagic pathways may provide a new opportunity for discovery and development of more novel cancer therapeutics. With a deeper understanding of the regulatory mechanisms governing autophagy, we will have a better opportunity to facilitate the exploitation of autophagy as a target for therapeutic intervention in cancer. This review discusses the current status of targeting autophagic pathways as a potential cancer therapy.


Subject(s)
Autophagy , Drug Discovery , Neoplasms/pathology , Antibiotics, Antineoplastic/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Autophagy/genetics , Beclin-1 , Chloroquine/therapeutic use , Humans , Membrane Proteins/metabolism , Molecular Targeted Therapy , Neoplasms/metabolism , Neoplasms/therapy , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism
12.
Front Pharmacol ; 13: 936818, 2022.
Article in English | MEDLINE | ID: mdl-35924055

ABSTRACT

Parkinson's disease (PD) is one of the neurodegenerative diseases that is characterized by obvious motor and some nonmotor symptoms. Various therapeutics failed in the effective treatment of PD because of impaired neurological function in the brain and various complications. Periplaneta Americana oligosaccharides (OPA), the main active ingredients extracted from the medicine residues of Periplaneta Americana (P. Americana), have been reported to exert anti-inflammatory effects. The purpose of this study was to evaluate the possible mechanisms of OPA against 1-methyl-4-phenylpyridinium (MPP+)-induced apotosis in SH-SY5Y cells and its potential neuroprotective effects in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD subacute model mice. The data demonstrated that OPA significantly reversed the MPP+-induced decrease in SH-SY5Y cell viability, reduced the proportion of apoptotic cells, and protected SH-SY5Y cells from apoptosis in a dose-dependent manner by regulating the expression of apoptosis-related genes. Furthermore, OPA also alleviated the motor dysfunction of PD model mice, prevented the loss of tyrosine hydroxylase positive cells, suppressed the apoptosis of substantia nigra cells, and improved the dysbiosis of gut microbiota in vivo, suggesting that OPA demonstrated a significantly neuroprotective effect on PD model mice. These results indicated that OPA might be the possibility of PD therapeutics with economic utility and high safety.

13.
Biochem Biophys Res Commun ; 406(4): 497-500, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21329660

ABSTRACT

Polygonatum cyrtonema lectin (PCL), a mannose/sialic acid-binding plant lectin, has recently drawn a rising attention for cancer biologists because PCL bears remarkable anti-tumor activities and thus inducing programmed cell death (PCD) including apoptosis and autophagy in cancer cells. In this review, we focus on exploring the precise molecular mechanisms by which PCL induces cancer cell apoptotic death such as the caspase-dependent pathway, mitochondria-mediated ROS-p38-p53 pathway, Ras-Raf and PI3K-Akt pathways. In addition, we further elucidate that PCL induces cancer cell autophagic death via activating mitochondrial ROS-p38-p53 pathway, as well as via blocking Ras-Raf and PI3K-Akt pathways, suggesting an intricate relationship between autophagic and apoptotic death in PCL-induced cancer cells. In conclusion, these findings may provide a new perspective of Polygonatum cyrtonema lectin (PCL) as a potential anti-tumor drug targeting PCD pathways for future cancer therapeutics.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Plant Lectins/pharmacology , Polygonatum/chemistry , Amino Acid Sequence , Caspases/metabolism , Cell Line, Tumor , Humans , Molecular Sequence Data , Phosphatidylinositol 3-Kinases/metabolism , Plant Lectins/chemistry , Protein Conformation , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , raf Kinases/metabolism , ras Proteins/metabolism
14.
Biochem Biophys Res Commun ; 414(2): 282-6, 2011 Oct 22.
Article in English | MEDLINE | ID: mdl-21951850

ABSTRACT

Concanavalin A (ConA), a Ca(2+)/Mn(2+)-dependent and mannose/glucose-binding legume lectin, has drawn a rising attention for its remarkable anti-proliferative and anti-tumor activities to a variety of cancer cells. ConA induces programmed cell death via mitochondria-mediated, P73-Foxo1a-Bim apoptosis and BNIP3-mediated mitochondrial autophagy. Through IKK-NF-κB-COX-2, SHP-2-MEK-1-ERK, and SHP-2-Ras-ERK anti-angiogenic pathways, ConA would inhibit cancer cell survival. In addition, ConA stimulates cell immunity and generates an immune memory, resisting to the same genotypic tumor. These biological findings shed light on new perspectives of ConA as a potential anti-neoplastic agent targeting apoptosis, autophagy and anti-angiogenesis in pre-clinical or clinical trials for cancer therapeutics.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Concanavalin A/pharmacology , Neoplasms/drug therapy , Amino Acid Sequence , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Cell Survival/drug effects , Concanavalin A/chemistry , Concanavalin A/therapeutic use , Humans , MAP Kinase Signaling System , Molecular Sequence Data , Neoplasms/blood supply , Neoplasms/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , ras Proteins/metabolism
15.
Curr Comput Aided Drug Des ; 17(4): 523-537, 2021.
Article in English | MEDLINE | ID: mdl-32598264

ABSTRACT

INTRODUCTION: In many diseased states, especially fibrosis and cancer, TGF-ß family members are overexpressed and the outcome of signaling is diverted toward disease progression. As the result of activin receptor-like kinase 1 (ALK1) plays a key role in TGF-ß signaling, discovering inhibitors of ALK1 to block TGF-ß signaling for a therapeutic benefit has become an effective strategy. METHODS: In this work, ZINC15894217 and ZINC12404282 were identified as potential ALK1 inhibitors using molecular docking, molecular dynamics simulation and MM/PBSA calculations studies. The analysis of energy decomposition found that Val208, Val216, Lys229, Gly283, Arg334 and Leu337 acted as crucial residues for ligand binding and system stabilizing. RESULTS: In addition, these compounds displayed excellent pharmacological and structural properties, which can be further evaluated through in vitro and in vivo experiments for the inhibition of ALK1 to be developed as drugs against fibrosis and tumor. CONCLUSION: Overall, our study illustrated a time- and cost-effective computer aided drug design procedure to identify potential ALK1 inhibitors. It would provide useful information for further development of ALK1 inhibitors to improve disease related to TGF-ß signal pathway.


Subject(s)
Neoplasms , Transforming Growth Factor beta , Humans , Molecular Docking Simulation , Neoplasms/drug therapy , Signal Transduction
16.
Biochim Biophys Acta ; 1790(8): 840-4, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19414060

ABSTRACT

BACKGROUND: The Galanthus nivalis agglutinin (GNA)-related lectins have been reported to bear antiproliferative and apoptosis-inducing activities in cancer cells; however, the precise mechanisms by which GNA-related lectins induce cell death are still only rudimentarily understood. METHODS: In the present study, Polygonatum odoratum lectin (designated POL), a mannose-binding specific GNA-related lectin, possessed a remarkable antiproliferative activity toward murine fibrosarcoma L929 cells. And, this lectin induced L929 cell apoptosis in a caspase-dependent manner. In addition, POL treatment increased the levels of FasL and Fas-Associated protein with Death Domain (FADD) proteins and resulted in caspase-8 activation. Also, POL treatment caused mitochondrial transmembrane potential collapse and cytochrome c release, leading to activations of caspase-9 and caspase-3. Moreover, POL treatment enhanced tumor necrosis factor alpha (TNFalpha)-induced L929 cell apoptosis. RESULTS: Our data demonstrate for the first time that this lectin induces apoptosis through both death-receptor and mitochondrial pathways, as well as amplifies TNFalpha-induced L929 cell apoptosis. GENERAL SIGNIFICANCE: These inspiring findings would provide new molecular basis for further understanding cell death mechanisms of the Galanthus nivalis agglutinin (GNA)-related lectins in future cancer investigations.


Subject(s)
Apoptosis/drug effects , Fibrosarcoma/pathology , Plant Lectins/pharmacology , Polygonatum/metabolism , Animals , Caspases/metabolism , Cell Line, Tumor , Drug Screening Assays, Antitumor , Fibrosarcoma/enzymology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Phytotherapy , Receptors, Death Domain/metabolism , Tumor Necrosis Factor-alpha/pharmacology
17.
Acta Pharmacol Sin ; 31(9): 1154-64, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20694022

ABSTRACT

Autophagy is an evolutionarily conserved lysosomal self-digestion process involved in degradation of long-lived proteins and damaged organelles. In recent years, increasing evidence indicates that autophagy is associated with a number of pathological processes, including cancer. In this review, we focus on the recent studies of the evolutionarily conserved autophagy-related genes (ATGs) that are implicated in autophagosome formation and the pathways involved. We discuss several key autophagic mediators (eg, Beclin-1, UVRAG, Bcl-2, Class III and I PI3K, mTOR, and p53) that play pivotal roles in autophagic signaling networks in cancer. We discuss the Janus roles of autophagy in cancer and highlighted their relationship to tumor suppression and tumor progression. We also present some examples of targeting ATGs and several protein kinases as anticancer strategy, and discuss some autophagy-modulating agents as antitumor agents. A better understanding of the relationship between autophagy and cancer would ultimately allow us to harness autophagic pathways as new targets for drug discovery in cancer therapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy , Neoplasms/drug therapy , Signal Transduction/drug effects , Animals , Humans , Neoplasms/metabolism
18.
Nanoscale ; 12(3): 2002-2010, 2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31912068

ABSTRACT

A number of multimodal agents have been developed for tumour imaging and diagnosis, but most of them cannot be used to study the detailed physiological or pathological changes in living cells at the same time. Herein, a series of pH-responsive magnetic resonance and fluorescence imaging (MRI/FI) dual-modal "nanovehicles" are developed and tested. These new dual-modal materials allow for intercellular pH sensing, and those with units that are dually sensitive towards both acidic and basic environments have the ability for intracellular pH mapping and can be used to quantify pH at the cellular level. In addition, detailed pH changes in organelles (including lysosomes and mitochondria) can be investigated at the same time. On the other hand, with the tumour-targeting peptide (cRGD)-modified dual-modal nanovehicles, in vivo tumour MR and fluorescence imaging, which is suitable for cancer diagnosis, can be achieved. Moreover, it has been proved that these materials can pass through the blood brain barrier (BBB). By combining the above mentioned promising properties, these novel multifunctional "nanovehicles" may provide a new method for studying the role of pH during cancer diagnosis and treatment.


Subject(s)
Drug Delivery Systems , Gold , Magnetic Resonance Imaging , Metal Nanoparticles , Neoplasms, Experimental , Optical Imaging , Animals , Female , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , HeLa Cells , Humans , Hydrogen-Ion Concentration , MCF-7 Cells , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oligopeptides/chemistry , Oligopeptides/pharmacology , Xenograft Model Antitumor Assays
19.
Arch Biochem Biophys ; 482(1-2): 1-6, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19111670

ABSTRACT

The objective of this study was to investigate the antiproliferative activity and apoptosis-inducing mechanism of Concanavalin A (ConA) on human melanoma A375 cells. We firstly simulated the three-dimensional structure of ConA. Subsequently, we found that ConA possessed remarkable antiproliferative effect on A375 cells. Further experimental data indicated that there was a link between its hemagglutinating activity, mannose-binding activity and antiproliferative activity. In addition, we showed that ConA induced A375 cell apoptosis in a caspase-dependent manner. Then, we demonstrated that the treatment of ConA caused mitochondrial transmembrane potential (MMP) collapse, cytochrome c release and caspase activation. In conclusion, we report for the first time that there may be a close correlation between carbohydrate-binding activity of ConA and its antiproliferative activity. Also, we demonstrate firstly that ConA induces A375 cell death in a caspase-dependent manner as well as through a mitochondrial apoptotic pathway.


Subject(s)
Apoptosis/drug effects , Cell Division/drug effects , Concanavalin A/pharmacology , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Computer Simulation , Concanavalin A/chemistry , Hemagglutination/drug effects , Humans , Mannose/pharmacology , Melanoma , Membrane Potentials/drug effects , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/physiology , Models, Molecular , Molecular Conformation
20.
Thorac Cancer ; 10(4): 715-727, 2019 04.
Article in English | MEDLINE | ID: mdl-30806032

ABSTRACT

BACKGROUND: Although cigarette smoking is considered one of the key risk factors for lung cancer, 15% of male patients and 53% of female patients with lung cancer are non-smokers. Metabolic changes are critical features of cancer. Therapeutic target identification from a metabolic perspective in non-small cell lung cancer (NSCLC) tissue of female non-smokers has long been ignored. RESULTS: Based on microarray data retrieved from Affymetrix expression arrays E-GEOD-19804, we found that the downregulated genes in non-smoking female NSCLC patients tended to participate in protein/amino acid and lipid metabolism, while upregulated genes were more involved in protein/amino acid and carbohydrate metabolism. Combining nutrient metabolic co-expression, protein-protein interaction network construction and overall survival assessment, we identified NR4A1 and TIE1 as potential therapeutic targets for NSCLC in female non-smokers. To accelerate the drug development for non-smoking female NSCLC patients, we identified nilotinib as a potential agonist targeting NR4A1 encoded protein by molecular docking and molecular dynamic stimulation. We also show that nilotinib inhibited proliferation and induced senescence of cells in non-smoking female NSCLC patients in vitro. CONCLUSIONS: These results not only uncover nutrient metabolic characteristics in non-smoking female NSCLC patients, but also provide a new paradigm for identifying new targets and drugs for novel therapy for such patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Pyrimidines/pharmacology , Receptor, TIE-1/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Non-Smokers/statistics & numerical data , Nuclear Receptor Subfamily 4, Group A, Member 1/antagonists & inhibitors , Nuclear Receptor Subfamily 4, Group A, Member 1/chemistry , Protein Interaction Maps , Pyrimidines/therapeutic use , Receptor, TIE-1/metabolism , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL