Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Basic Clin Pharmacol Toxicol ; 117(2): 105-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25615234

ABSTRACT

Gelatinases play important roles in tumour invasion and metastasis and are thus considered promising targets for cancer therapy. In this study, a new single-chain variable fragment (scFv)-based fusion protein Fv-LDP, composed of the anti-gelatinases scFv and lidamycin apoprotein (LDP), was prepared, and its combination with angiogenesis inhibitor Endostar was then investigated. The fusion protein Fv-LDP specifically bound to various tumour cells, and its binding capability to human pulmonary giant cell carcinoma (PG) cells was higher than that of LDP. Fv-LDP inhibited the expression and secretion of gelatinases and could be internalized into tumour cells via endocytosis. Fv-LDP also suppressed the growth of human hepatoma cells and murine hepatoma 22 transplanted in Kunming mice in various degrees. In addition, Endostar could enhance the synergistic or additive inhibition of Fv-LDP on the growth, migration or invasion of human hepatoma cells shown by a colony formation assay and a transwell-based migration or invasion assay, respectively. In vivo, Fv-LDP/Endostar combination showed a significantly synergistic effect on the growth of a human hepatoma xenograft, with an inhibition rate of 80.8% compared with the Fv-LDP (44.1%) or Endostar (8.9%)-treated group. The above-mentioned results indicate that the fusion protein Fv-LDP is effective against transplantable hepatoma in mice and human hepatoma xenografts in athymic mice. Moreover, Endostar can potentiate the inhibition effect of Fv-LDP on the growth of human hepatoma cells and xenografts. These data will provide a new combined strategy for improving the therapeutic efficacy of treatments for hepatoma or other gelatinase-overexpressing tumours.


Subject(s)
Aminoglycosides/pharmacology , Carcinoma, Hepatocellular/drug therapy , Endostatins/pharmacology , Enediynes/pharmacology , Liver Neoplasms/drug therapy , Single-Chain Antibodies/pharmacology , Aminoglycosides/administration & dosage , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacology , Animals , Apoproteins/administration & dosage , Apoproteins/pharmacology , Carcinoma, Giant Cell/drug therapy , Carcinoma, Giant Cell/enzymology , Carcinoma, Giant Cell/pathology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Drug Synergism , Endostatins/administration & dosage , Enediynes/administration & dosage , Female , Gelatinases/metabolism , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Recombinant Proteins , Single-Chain Antibodies/administration & dosage , Xenograft Model Antitumor Assays
2.
J Pharm Sci ; 103(4): 1204-13, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24504597

ABSTRACT

We have chemically synthesized NGR-LDP-PYM, a novel CD13/aminopeptidase (APN)-targeting and hydrolase-resistant conjugate by cross-linking of the antitumor antibiotic pingyangmycin (bleomycin A5 , PYM) to an engineered NGR motif-integrated apoprotein (NGR-LDP) with a noncleavable linker. This protein-drug conjugate not only basically retains the original properties of PYM but also can specifically deliver PYM to the CD13/APN-expressing tumor cells. Furthermore, the resulting conjugate exhibits more resistance to hydrolysis of recombinant human bleomycin hydrolase than parental PYM. These results may be useful for improving the therapeutic efficacy of PYM and have implications in the treatment of PYM-refractory and CD13/APN-overexpressing tumors.


Subject(s)
Aminopeptidases/metabolism , Antibiotics, Antineoplastic/administration & dosage , Apoproteins/chemistry , Bleomycin/analogs & derivatives , CD13 Antigens/metabolism , Drug Delivery Systems , Amino Acid Sequence , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Antibodies, Monoclonal , Apoproteins/metabolism , Bleomycin/administration & dosage , Bleomycin/chemistry , Bleomycin/metabolism , Cell Line, Tumor , Humans , Hydrolases/metabolism , Hydrolysis , Molecular Sequence Data , Neoplasms/drug therapy , Neoplasms/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism
3.
Yao Xue Xue Bao ; 48(7): 1113-8, 2013 Jul.
Article in Chinese | MEDLINE | ID: mdl-24133977

ABSTRACT

LPS stimulation of macrophages production of IFN-beta plays a key role in innate immunity defending the microbial invasion. In this study, the effect of S632A3 promoting LPS-induced IFN-beta production and the underlying mechanism were investigated, mRNA level was measured by real-time PCR, cytokine production was determined by ELISA, GSK-3beta activity was investigated by kinase assay, protein phosphorylation and expression were evaluated by Western blotting. The results revealed that S632A3 significantly augmented IFN-beta production by LPS-stimulated macrophages. S632A3 inhibition of the activation of GSK-3beta, reduced the threonine 239 phosphorylation of transcription factor c-Jun but increased the total level of c-Jun in LPS-stimulated macrophages. Moreover, small interfering RNA-mediated knockdown of c-Jun level abrogated the ability of S632A3 to augment IFN-beta. The study thus demonstrates S632A3 being a new anti-inflammation lead compound and provides a molecular mechanism by which S632A3 promoted LPS-induced IFN-beta production in macrophages through inhibiting the activation of GSK-3beta.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Interferon-beta/biosynthesis , Macrophages/metabolism , Piperidones/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Cell Line , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3 beta , Interferon-beta/genetics , Lipopolysaccharides/pharmacology , Macrophages/cytology , Mice , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Transfection
4.
Food Chem Toxicol ; 62: 499-505, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24055767

ABSTRACT

Salidroside (SAL) is one of main active components of Rhodiola rosea L. and possesses diverse pharmacological effects. However, the direct role of SAL in bone metabolism remains elusive. In this study, effects of SAL on osteoblast differentiation of murine pluripotent mesenchymal cell line C3H10T1/2 and osteoblastic cell line MC3T3-E1 were examined. We first identified SAL as a potential BMP2 activator in a cell-based screening assay. SAL (0.5-10 µM) could slightly promote the proliferation and greatly increase the alkaline phosphatase (ALP) activity in both cells. Furthermore, SAL increased the mRNA expressions of osteoblast marker genes in either C3H10T1/2 or MC3T3-E1 cells after treatment for different time. Moreover, the mineralization of C3H10T1/2 cells assayed by Alizarin red S staining was dose-dependently increased by SAL. Mechanistically, SAL increased the mRNA level of genes involved in the regulation of BMP signaling pathway, including BMP2, BMP6 and BMP7 and enhanced the phosphorylation of Smad1/5/8 and ERK1/2. The osteogenic effect of SAL was abolished by BMP antagonist noggin or by BMP receptor kinase inhibitor dorsomorphin. Further in vivo study demonstrated that SAL reversed bone loss in ovariectomized rats. Collectively, our findings indicate that SAL regulates bone metabolism through BMP signaling pathway.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation/drug effects , Glucosides/pharmacology , Osteoblasts/drug effects , Phenols/pharmacology , Animals , Biomarkers , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Proteins/genetics , Cell Line , Gene Expression Regulation/drug effects , Mice , Osteoblasts/metabolism , Ovariectomy , Phosphorylation/drug effects , Pluripotent Stem Cells/drug effects , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
5.
Chin J Integr Med ; 19(4): 243-52, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23546627

ABSTRACT

To provide a better service for senior health care, we summarized screening studies of traditional Chinese anti-aging materia medica (TCAM). We collected and analyzed literature of TCAM screening studies using the lifespan test and animal models of aging from 1984 to 2012. We found 26 screening methods for TCAM, and 153 single herbs or active ingredients of TCAM that have been screened out during the past 28 years. The cell lifespan test, the fruit fly lifespan test, and D-galactose aging model were the most widely used and intensively studied screening methods. However, the method for establishing the D-galactose aging model needs to be standardized, and the D-galactose aging model cannot completely be a substitute for the normal aging mouse model. Great success has been achieved in screening studies in TCAM. To further improve screening studies in TCAM, we suggest that the D-galactose aging model be incorporated into the lifespan test in the New Drugs of Traditional Chinese Medicine Research Guide.


Subject(s)
Aging/drug effects , Drug Evaluation, Preclinical , Drugs, Chinese Herbal/pharmacology , Materia Medica/pharmacology , Medicine, Chinese Traditional , Animals , Humans , Models, Animal
6.
Phytother Res ; 27(3): 417-21, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22628222

ABSTRACT

Berberine (BBR) has been suggested to be a hepatoprotective agent for oxidative-stress-related liver diseases because of its antioxidant activity. However, the antioxidant mechanisms of BBR are still not fully understood. In the present study, the protective effect of BBR was evaluated, and the underlying molecular mechanisms were investigated in hepatic cell line L02. Results from cell viability and apoptosis assay showed that in cells exposed to hydrogen peroxide (H2 O2 ), the pretreatment of 12 µM BBR could increase cell viability by 19.10 ± 7.40% and reduce apoptotic cells by 7.91 ± 0.78%. A significant change in the expression levels of sirtuin 1 (SIRT1) and apoptosis-related proteins was also observed in the BBR-pretreated hepatocytes under exposure to H2 O2 . Furthermore, BBR exhibited a time-dependent effect on upregulation of SIRT1 in L02 cells. This study demonstrated that the protective effect of BBR against H2 O2 -induced apoptosis was associated with regulation of SIRT1 in hepatic cell line L02, which provided a possible explanation for its antioxidant activity, and implied an application of BBR for the therapeutic relevance in oxidative-stress-related liver diseases.


Subject(s)
Antioxidants/pharmacology , Apoptosis/drug effects , Berberine/pharmacology , Hepatocytes/drug effects , Oxidative Stress/drug effects , Sirtuin 1/metabolism , Cell Line , Hepatocytes/metabolism , Humans , Hydrogen Peroxide/pharmacology , Up-Regulation/drug effects
7.
Yao Xue Xue Bao ; 48(10): 1563-9, 2013 Oct.
Article in Chinese | MEDLINE | ID: mdl-24417083

ABSTRACT

This study is to optimize the preparation process of fusion protein Fv-LDP which was expressed in the form of inclusion body and consisted of lidamycin apoprotein LDP and single-chain Fv antibody (scFv) directed against type IV collagenase. The preparation and the dissolution of inclusion body, the immobilized metal affinity chromatography of the target protein and the renaturization by stepwise dialysis were optimized by single-factor analysis or orthogonal design. In addition, the refolded fusion protein Fv-LDP was refined by Sephadex G-75 chromatography followed by fluorescence-activated cell sorter (FACS)-based saturation binding assay to measure its antigen-binding activity. After optimization of the process, the purity of fusion protein Fv-LDP existed in the inclusion body was 63.9% and the corresponding solubility was 95.7%; Under denaturing conditions, the purity of fusion protein Fv-LDP was more than 95% after the purification process. The percentage of monomeric fusion protein Fv-LDP was 60% after the refolding process, while it was further refined to 85% which was 5.6-fold higher than that of the initial refolding condition. The refined fusion protein Fv-LDP could bind to human lung adenocarcinoma PAa cells and human hepatoma BEL-7402 cells with the dissociation constants (Kd) of 0.176 micromol x L(-1) and 0.904 micromol x L(-1), respectively. The preparation process of fusion protein Fv-LDP has been successfully optimized, which provides the experimental basis for the production and future development of fusion protein Fv-LDP, and might serve as a relatively practical system for the preparation of other scFv-based proteins expressed in the form of inclusion body.


Subject(s)
Aminoglycosides , Apoproteins , Enediynes , Recombinant Fusion Proteins , Single-Chain Antibodies , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Aminoglycosides/chemistry , Aminoglycosides/metabolism , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/metabolism , Apoproteins/chemistry , Apoproteins/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Collagenases/immunology , Enediynes/chemistry , Enediynes/metabolism , Escherichia coli/chemistry , Escherichia coli/metabolism , Humans , Inclusion Bodies/chemistry , Inclusion Bodies/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/metabolism
8.
Exp Cell Res ; 318(20): 2592-603, 2012 Dec 10.
Article in English | MEDLINE | ID: mdl-22975730

ABSTRACT

Inflammatory mediators including inducible nitric oxide (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNF-α) and Interleukin-6 (IL-6) contribute to the course of a variety of inflammatory diseases. S632A3 is a new member of the glutarimide antibiotics isolated from a cultured broth of Streptomyces hygroscopicus S632 with a potent NF-κB inhibitory activity. In the present study, we investigated the anti-inflammatory effects and the underlying molecular mechanism of S632A3 on lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. S632A3 concentration-dependently inhibited LPS-induced NO and prostaglandin E(2) (PGE(2)) production through the suppression of iNOS and COX-2 at gene transcription levels. In addition, S632A3 suppressed NF-κB-dependent inflammatory responses by inhibiting the activation of glycogen synthase kinase 3ß (GSK-3ß), while the activation of IκB kinase (IKK) complex was unaffected. S632A3 suppressed NF-κB activity by differentially affecting the CREB (cAMP response element-binding protein) and NF-κB p65 interacting with the coactivator CBP (CREB binding protein). S632A3 also inhibited GSK-3ß-elicited iNOS and COX-2 expression. Moreover, S632A3 was shown to inhibit the activation of ASK1 (Apoptosis-signal regulating kinase 1) and p38 mitogen-activated protein kinase, therefore attenuated the LPS-induced NF-κB activity in macrophages. Furthermore, S632A3 significantly reduced the pro-inflammatory cytokines TNF-α and IL-6 production while increased the anti-inflammatory cytokine IL-10 production in LPS-stimulated RAW264.7 cells. Our study thus provides a molecular mechanism by which S632A3 inhibited LPS-induced pro-inflammatory response in macrophages through interfering with the activation of GSK-3ß and ASK1-p38 signaling.


Subject(s)
Anti-Bacterial Agents/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Inflammation Mediators/antagonists & inhibitors , Lipopolysaccharides/antagonists & inhibitors , Macrophages/drug effects , Piperidones/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line , Glycogen Synthase Kinase 3 beta , Inflammation Mediators/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/pathology , Mice
9.
Int J Biochem Cell Biol ; 44(11): 1813-24, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22750415

ABSTRACT

Na(+)/K(+)-ATPase targeted cancer therapy has attracted increasing interests of oncologists in lung cancer field. Although multiple anti-cancer mechanisms of cardiac glycosides as Na(+)/K(+)-ATPase inhibitors are revealed, the role of autophagy and related molecular signaling pathway for the class of compounds in human non-small cell lung cancer (NSCLC) cells has not been systematically examined. We herein investigated the anti-cancer effects of two representative cardiac glycosides, digoxin and ouabain, in A549 and H460 cell lines. Both agents caused significant growth inhibition at nanomolar level. The cardiac glycosides were found to induce moderate G(2)/M arrest but not apoptosis at IC(50) level in the NSCLC cell lines. Moreover, autophagy was markedly induced by both agents, as evidenced by the time- and dose-dependent increase of LC3-II, up-regulation of Atg5 and Beclin1, as well as by the observations through acridine orange staining, transmission electron microscopy and quantification of GFP-LC3 fluorescence. Importantly, AMP-activated protein kinase (AMPK) pathway was activated, resulting in mammalian target of rapamycin (mTOR) deactivation during autophagy induction. Moreover, extracellular-signal-regulated kinase 1/2 (ERK1/2) activation was simultaneously found to be involved in the autophagy regulation. Co-treatment with respective inhibitors or siRNAs could either block the autophagic phenotypes and signals, or significantly increase the cellular viability, indicating the drugs-induced autophagy plays tumor-suppressing role. This work provides first evidence showing that the cardiac glycosides induce autophagy in human NSCLC cells through regulation of both mTOR and ERK1/2 signaling pathways. The autophagy may at least partially account for the growth inhibitory effects of the compounds in human NSCLC cells.


Subject(s)
Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/pathology , Cardiac Glycosides/pharmacology , Lung Neoplasms/pathology , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Androstadienes/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Protein 5 , Beclin-1 , Biomarkers/metabolism , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/ultrastructure , Cell Line, Tumor , Digoxin/pharmacology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Humans , Inhibitory Concentration 50 , Lung Neoplasms/enzymology , Lung Neoplasms/ultrastructure , MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Mitosis/drug effects , Ouabain/pharmacology , TOR Serine-Threonine Kinases/metabolism , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism , Vacuoles/drug effects , Vacuoles/metabolism , Vacuoles/ultrastructure , Wortmannin
10.
Int J Biochem Cell Biol ; 43(3): 409-15, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21095239

ABSTRACT

The glycoprotein erythropoietin (Epo) is a hematopoietic cytokine necessary for the survival of erythrocytes from immature erythroid cells. The mitogen-activated c-Jun N-terminal kinase 1 (JNK1) plays an important role in the proliferation and survival of erythroid cells in response to Epo. However, the precise mechanism of JNK1 activation promoting erythroid cell survival is incompletely understood. Here, we reported that JNK1 is required for Epo-mediated cell survival through phosphorylation and inactivation of the pro-apoptotic, Bcl-2 homology domain 3 (BH3)-only Bcl-associated death protein (Bad). Upon Epo withdrawal, HCD57 cells, a murine Epo-dependent cell line, displayed increased apoptotic cell death that was associated with decreased JNK1 activity. Epo withdrawal-induced apoptosis was promoted by inhibition of JNK1 activity but suppressed by expression of a constitutively active JNK1. Furthermore, Epo-activated JNK1 phosphorylated Bad at threonine 201, thereby inhibiting the association of Bad with the anti-apoptotic molecule B-cell lymphoma-extra large (Bcl-X(L)). Replacement of threonine 201 by alanine in Bad promoted Epo withdrawal-induced apoptosis. Thus, our results provide a molecular mechanism by which JNK1 contributes to the survival of erythroid cells.


Subject(s)
Erythropoietin/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , bcl-Associated Death Protein/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Enzyme Activation/drug effects , Mice , Phosphorylation/drug effects , Phosphothreonine/metabolism
11.
Mech Ageing Dev ; 131(11-12): 723-31, 2010.
Article in English | MEDLINE | ID: mdl-21035481

ABSTRACT

Although salidroside and salidroside-like compounds are considered as most critical constitutes needed and responsible for multiple therapeutic benefits of Rhodiola rosea L., including anti-aging, direct demonstration regarding the role of salidroside in anti-aging process is still deficient. In this study, we selected the H(2)O(2)-induced premature senescence model in human fetal lung diploid fibroblasts to investigate the protection of salidroside against aging in vitro and associated molecular mechanisms. We found that salidroside considerably reversed senescence-like phenotypes in the oxidant challenged model, including alterations of morphology, cell cycle, SA-ß-gal staining, DNA damage, as well as related molecules expression such as p53, p21 and p16. The protection occurred in a dose-dependent manner, with 5µM offering best efficacy. The proposed antioxidant property of the compound was confirmed in this cellular system, and thus at least partially accounted for the protection of the compound against premature senescence. Similar protection of salidroside against replicative senescence was observed as well. Interestingly, the regulation of senescence-related molecules by salidroside involved ROS-irrelevant mechanisms in both models. This finding presents salidroside as an attractive agent with potential to retard aging and attenuate age-related diseases in humans.


Subject(s)
Aging, Premature/prevention & control , Antioxidants/pharmacology , Cellular Senescence/drug effects , Cytoprotection , Glucosides/pharmacology , Hydrogen Peroxide/antagonists & inhibitors , Phenols/pharmacology , Aging, Premature/pathology , Antioxidants/chemistry , Biomarkers/metabolism , Cell Cycle/drug effects , Cell Line , DNA Damage/drug effects , Fibroblasts/drug effects , Glucosides/chemistry , Humans , Hydrogen Peroxide/toxicity , Oxidative Stress/drug effects , Phenols/chemistry , Proteins/metabolism , Rhodiola/chemistry , Superoxide Dismutase/metabolism
12.
Mol Immunol ; 48(1-3): 287-93, 2010.
Article in English | MEDLINE | ID: mdl-20728221

ABSTRACT

T cell dysfunction is the primary immunologic abnormality associated with aging. Many age-related defects stem from a decline in CD4(+) T cell function. Resistance of aged CD4(+) T cells to apoptosis is associated with autoimmune and infectious diseases. Previous studies suggest that IκB kinase (IKK) may be a key player in cell survival via its inhibition of c-Jun N-terminal protein kinase (JNK) activation. However, the role of IKK-mediated JNK inactivation in the age-related apoptosis of T cells is unclear. Here, we report that splenic CD4(+) T cells in aged mice are resistant to activation-induced cell death (AICD) induced by anti-CD3 plus IL-2 stimulation. Furthermore, aged CD4(+) T cells display increased IKKß activity that is associated with attenuated JNK activation. The IKKß-mediated JNK inactivation in aged CD4(+) T cells reduces the degradation of c-FLIP(L) and the interaction of Bad with Bcl-X(L), but it increases the affinity of Bad for 14-3-3. Pretreatment of aged CD4(+) T cells with a specific IKK inhibitor, PS1145, increases the JNK activity blocked by IKKß and consequently sensitizes the aged CD4(+) T cells to AICD. Our study thus demonstrates that IKK antagonizes the AICD of CD4(+) T cells in aged mice via inhibition of JNK activation.


Subject(s)
Aging/physiology , Apoptosis/physiology , CD4-Positive T-Lymphocytes/metabolism , I-kappa B Kinase/metabolism , MAP Kinase Kinase 4/metabolism , Signal Transduction/physiology , Animals , DNA Fragmentation , Enzyme Activation/physiology , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred C57BL
13.
Biomed Environ Sci ; 23(2): 161-6, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20514993

ABSTRACT

OBJECTIVE: To investigate the protective effects of putative AGEs (advanced glycation endproducts) inhibitor salidroside against aging in an accelerated mouse aging model induced by D-galactose. METHODS: A group of 5-month-old C57BL/6J mice were treated daily with D-galactose, D-galactose combined with salidroside, salidroside alone, and control buffer for 8 weeks. At the end of the treatment, serum AGEs levels, neurological activities, expression of glial fibrillary acidic protein (GFAP) and neurotrophin-3 (NT-3) in the cerebral cortex, as well as lymphocyte proliferation and IL-2 production were determined. RESULTS: D-galactose induced mouse aging model was developed as described before. As expected, salidroside blocked D-galactose induced increase of serum AGEs levels. It also reversed D-galactose induced aging effects in neural and immune system, as evidenced by improving motor activity, increasing memory latency time, and enhancing lymphocyte mitogenesis and interleukin-2 (IL-2) production. Furthermore, elevated expression of GFAP and NT-3 in the aged model mice was also reduced upon salidroside treatment. CONCLUSION: Salidroside inhibits AGEs formation in vivo, which at least partially contributes to its anti-aging effect in D-galactose induced aging model.


Subject(s)
Aging, Premature/prevention & control , Drugs, Chinese Herbal/therapeutic use , Glucosides/therapeutic use , Phenols/therapeutic use , Aging, Premature/blood , Aging, Premature/chemically induced , Animals , Cerebral Cortex/metabolism , Drugs, Chinese Herbal/pharmacology , Galactose , Glial Fibrillary Acidic Protein , Glucosides/pharmacology , Glycation End Products, Advanced/blood , Interleukin-2/metabolism , Memory/drug effects , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Nerve Growth Factors/metabolism , Nerve Tissue Proteins/metabolism , Phenols/pharmacology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/drug effects
14.
Oncol Rep ; 23(2): 477-83, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20043110

ABSTRACT

Chemomicin (CHM), an angucyclinone antibiotic extracted from the fermentation broth of Nocardia Mediterranei subsp. Kanglensis 1747-64, shows immunosuppressive activity. However, whether it can inhibit growth of tumor cells remains elusive. In the present study, we show that CHM potently inhibited the proliferations of eight various types of human tumor cell lines and non-cross resistant to multidrug-resistant cells. In contrast to action of doxorubicin, the generation of reactive oxygen species was observed as early as 30 min after addition of CHM and its process did not involve iron. The apoptotic cells with chromatin condensation and Annexin V staining markedly increased after the human hepatoma HepG2 was exposed to 1, or 2 microg/ml CHM for 24 h. In the CHM-induced apoptosis, robust increment of p53 expression, activation of caspase-3, -7, -8, -9, cleavage of PARP and the phosphorylation of p38 and JNK, were detected by Western blot analysis. Further investigation revealed the disruption of mitochondrial membrane potential in the cells with CHM incubation for 4 h. Taken together, the results demonstrated that potent proliferation inhibitory effect of CHM on tumor cells is due to activation of the apoptotic pathway.


Subject(s)
Anthraquinones/pharmacology , Apoptosis/drug effects , Neoplasms/pathology , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , G2 Phase/drug effects , HCT116 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Models, Biological , Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Cells, Cultured
15.
Biomed Environ Sci ; 22(3): 244-52, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19725468

ABSTRACT

OBJECTIVE: Lidamycin, an enediyne antibiotic, leads to apoptosis and mitotic cell death of human tumor cells at high and low concentrations. The reason why tumor cells have distinct responses to lidamycin remains elusive. This study was to elucidate if cellular prosurvival molecules are involved in these responses. METHODS: Cleavage of chromatin and DNA was observed by chromatin condensation and agarose gel electrophoresis. Accumulation of rhodamine 123 in lidamycin-treated cells was assayed by flow cytometry. Cell multinucleation was detected by staining with Hoechst 33342. Western blot and senescence-associated beta-galactosidase (SA-beta-gal) staining were used to analyze protein expression and senescence-like phenotype, respectively. RESULTS: SIRT1 deacetylase remained unchanged in 0.5 nmol/L lidamycin whereas cleavage occurred when apoptosis was induced by lidamycin. Increased FOXO3a, SOD-1 and SOD-2 expression and transient phosphorylation of ERK were detected after exposure of human hepatoma BEL-7402 cells to 0.5 nmol/L lidamycin. High expressions of SIRT1 and Akt were found in colon carcinoma HCT116 p53 knock-out cells exposed to lidamycin. Degradation of PARP and p53 by lidamycin as a substitute for SIRT1 and Akt was confirmed with caspase inhibitor Q-VD-OPh and proteasome inhibitor MG132. Resistance to lidamycin-induced DNA cleavage was observed in breast cancer doxorubicin-resistant MCF-7 cells. This was not induced by P-glycoprotein as no accumulation of rhodamine 123 was detected in the resistant cells following exposure to lidamycin. In contrast to sensitive MCF-7 cells, a lower multinucleation rate for the resistant cells was measured following exposure to equal concentrations of lidamycin. CONCLUSIONS: Cellular prosurvival molecules, such as SIRT1, Akt, SOD-1, SOD-2 and other unknown factors can influence the action of lidamycin on human tumor cells.


Subject(s)
Aminoglycosides/pharmacology , Antibiotics, Antineoplastic/pharmacology , Enediynes/pharmacology , Cell Death/drug effects , Cell Line, Tumor , DNA Cleavage , Doxorubicin/pharmacology , Forkhead Box Protein O3 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Sirtuin 1 , Sirtuins/genetics , Sirtuins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Biomed Environ Sci ; 21(3): 212-7, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18714818

ABSTRACT

OBJECTIVE: To study whether Lycium barbarum glycopeptide 3 (LBGP3) affects T cell apoptosis in aged mice. METHODS: LBGP3 was purified with DEAE cellulose and Sephadex columns. Apoptotic "sub-G1 peak" was detected by flow cytometry and DNA ladder was resolved by agarose gel electrophoresis. Levels of IFN-gamma and IL-10 were measured with specific kits and mRNA expression was detected by RT-PCR. Apoptosis-related proteins of FLIP, FasL, and Bcl-2 were determined by Western blotting. RESULTS: LBGP3 was purified from Fructus Lycii water extracts and identified as a 41 kD glycopeptide. Treatment with 200 microg/mL LBGP3 increased the apoptotic rate of T cells from aged mice and showed a similar DNA ladder pattern to that in young T cells. The reversal of apoptotic resistance was involved in down-regulating the expression of Bcl-2 and FLIP, and up-regulating the expression of FasL. CONCLUSION: Lycium barbarum glycopeptide 3 reverses apoptotic resistance of aged T cells by modulating the expression of apoptosis-related molecules.


Subject(s)
Aging/immunology , Apoptosis/drug effects , Glycopeptides/pharmacology , Lycium/chemistry , T-Lymphocytes/drug effects , Animals , Fas Ligand Protein/immunology , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-bcl-2/immunology , RNA, Messenger/immunology , T-Lymphocytes/immunology
17.
Yao Xue Xue Bao ; 43(10): 1003-10, 2008 Oct.
Article in Chinese | MEDLINE | ID: mdl-19127863

ABSTRACT

The mechanism of apoptosis induced by SIRT1 deacetylase inhibitors in both human breast cancer MCF-7 and MCF-7 doxorubicin-resistant cells was studied. MTT assay was used to detect growth-inhibitory effect on the cells. Protein expression was detected by Western blotting. Chromatin condensation was detected by a fluorescent microscope after Hoechst 33342 staining. Cell cycle distribution was analyzed with flow cytometry. Apoptotic cells were detected with Annexin V staining. Nicotinamide (NAM) and Sirtinol, two SIRT1 deacetylase inhibitors, exhibited the similar growth-inhibitory effects on MCF-7/DOX cells and MCF-7 cells, but no potentiation of DOX activities. The arrest at G2/M phase was detected by flow cytometry in both MCF-7 and MCF-7/DOX cells after NAM treatment. Activation of caspase pathway in MCF-7 cells, such as the cleavages of PARP, caspase-6, -7, -9, were observed after exposure to NAM 50 mmol x L(-1), accompanied by the occurrence of chromatin condensation and Annexin V positive cells. However, the cleavages of PARP, caspase-6 and -7 in MCF-7/DOX cells delayed after exposure to NAM for 24 h and obviously increased at 48 h with appearance of chromatin condensation and Annexin V positive cells. SIRT1 deacetylase inhibitors show no cross resistance to MCF-7 drug-resistant cells, and the similar growth-inhibitory actions of them to MCF-7 sensitive and drug-resistant cells by which it is mediated by activation of apoptotic caspase pathway.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Sirtuin 1/antagonists & inhibitors , Benzamides/pharmacology , Breast Neoplasms/metabolism , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Doxorubicin/pharmacology , Enzyme Inhibitors/pharmacology , Female , Humans , Naphthols/pharmacology , Niacinamide/pharmacology
18.
Biomed Environ Sci ; 20(3): 189-97, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17672208

ABSTRACT

OBJECTIVE: To investigate the effect of lidamycin (LDM) on telomerase activity in human hepatoma BEL-7402 cells under the condition of LDM inducing mitotic cell death and senescence. METHODS: Chromatin condensation was detected by co-staining with Hoechst 33342 and PI. Cell multinucleation was observed by Giemsa staining and genomic DNA was separated by agarose gel electrophoresis. Fluorescent intensity of Rho123 was determined for mitochondrial membrane potential. MTT assay and SA-beta-gal staining were employed to analyze the senescence-like phenotype. The expression of proteins was analyzed by Western blot. Telomerase activity was assayed by telomerase PCR-ELISA. RESULTS: Mitotic cell death occurred in LDM-treated cells characterized by unique and atypical chromatin condensation, multinucleation and increased mitochondrial membrane potential. However, no apoptotic bodies or DNA ladders were found. In addition, apoptosis-related proteins remained nearly unaltered. Senescence-like phenotype was identified by increased and elongated size of cells, growth retardation, enhanced SA-beta-gal activity and the changes of senescence-related protein expression. Telomerase activity markedly decreased (P<0.01) in LDM-treated hepatoma BEL-7402 cells. CONCLUSION: Mitotic cell death and senescence could be triggered simultaneously or sequentially after exposure of hepatoma BEL-7402 cells to LDM. The decrease in telomerase activity may play a key role in the defective mitosis and aging morphology. Further investigation of detailed mechanism is needed.


Subject(s)
Aminoglycosides/pharmacology , Antibiotics, Antineoplastic/pharmacology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Enediynes/pharmacology , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Telomerase/metabolism , Apoptosis/drug effects , Azure Stains , Benzimidazoles , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cellular Senescence/drug effects , Chromatin/metabolism , DNA, Neoplasm/analysis , Dose-Response Relationship, Drug , Genome, Human/genetics , Humans , Membrane Potential, Mitochondrial/drug effects , Mitosis/drug effects , Phenotype , Propidium , Time Factors , beta-Galactosidase/metabolism
19.
Yao Xue Xue Bao ; 42(2): 132-8, 2007 Feb.
Article in Chinese | MEDLINE | ID: mdl-17518039

ABSTRACT

Although enediyne antibiotic lidamycin ( LDM) is a potent inducer of apoptosis, the underlying mechanisms of its apoptotic functions remain to be explored. Here, we aim to elucidate its possible mechanisms in mitochondria initiated apoptotic pathway involved in human BEL-7402 and MCF-7 cells. Cytochrome c released from mitchondria to cytosol fraction was detected by Western blotting. p53 and Bax, Bcl-2 expressions were detected by Western blotting and RT-PCR. MTT assay was used to detect cytotoxicity of LDM with or without caspase inhibitor z-VAD-fmk. After the BEL-7402 cells were exposed to 0. 1 micromol x L(-1) LDM within 6 h, the increase of cytochrome c in the cytosol and decrease in the mitochondria were observed when compared with untreated cells. The expression of Bax, an important proapoptotic member of the Bcl-2 family, increased gradually in the BEL-7402 cells after exposure to LDM of 0. 1 micromol x L (-1) for 2, 6, and 9 h, separately, while Bcl-2 increased at 2 and 6 h, and decreased at 9 h after LDM treatment. Enhanced protein expressions were parallel with respective increased mRNA level for Bax only, but not p53. Caspase inhibitor may inhibit partially the killing effects induced by LDM. Therefore we conclude that the rapid activation of mitochondrial pathway induced by LDM in tumor cells might contribute to its highly potent cytotoxicities.


Subject(s)
Aminoglycosides/pharmacology , Apoptosis/drug effects , Enediynes/pharmacology , Mitochondria/drug effects , Signal Transduction/drug effects , Amino Acid Chloromethyl Ketones/pharmacology , Antibiotics, Antineoplastic/pharmacology , Blotting, Western , Caspase Inhibitors , Caspases/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Cytosol/drug effects , Cytosol/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
20.
Cancer Biol Ther ; 6(6): 965-73, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17534142

ABSTRACT

Enediyne compound is one class of antibiotics with very potent anti-cancer activity. However, the role of p53 in enediyne antibiotic-induced cell killing remains elusive. Here we reported the involvement of p53 signaling pathway in apoptosis induction by lidamycin (LDM), a member of the enediyne antibiotic family. We found that LDM at low drug concentration of 10 nmol/L induces apoptotic cell death much more effectively in human colorectal cancer cells with wild type p53 than those with mutant or deleted p53. p53 is functionally activated as an early event in response to low dose LDM that precedes the significant apoptosis induction. The primarily activation of mitochondria as well as the activation of p53 transcriptional targets such as Puma, Bad and Bax in HCT116 p53 wild type cells further demonstrates the key role of p53 in mediating the compound-induced apoptosis. This is further supported by the observation that the absence of Bax or Puma decreases apoptosis dramatically while Bcl-2 overexpression confers partially resistance after drug treatment. Activation of p53 signaling pathway leads to activation of caspases and caspases inhibitor VAD-fmk completely blocks low dose LDM induced apoptosis through the inhibition of mitochondria pathway. In contrast, LDM at higher concentration causes rapid apoptosis through more direct DNA damaging mechanism that is independent of activation of p53 and caspases and cannot be blocked by caspase inhibitor. Taken together, LDM induces apoptosis in a p53-dependent manner when given at low doses, but in a p53-independent manner when given at high doses. This dosage-dependent regimen can be applied to cancer clinic based upon the p53 status of cancer patients.


Subject(s)
Aminoglycosides/pharmacology , Antibiotics, Antineoplastic/pharmacology , Apoptosis , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Enediynes/pharmacology , Gene Expression Regulation, Neoplastic , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Chromatin/metabolism , Dose-Response Relationship, Drug , Humans , In Situ Nick-End Labeling , Membrane Potentials , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...