Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
2.
Appl Microbiol Biotechnol ; 99(7): 2999-3013, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25646963

ABSTRACT

H9N2 subtype avian influenza viruses are widespread in domestic poultry, and vaccination remains the most effective way to protect the chicken population from avian influenza pandemics. Currently, egg-based H9N2 influenza vaccine production has several disadvantages and mammalian MDCK cells are being investigated as candidates for influenza vaccine production. However, little research has been conducted on low pathogenic avian influenza viruses (LPAIV) such as H9N2 replicating in mammalian cells using microcarrier beads in a bioreactor. In this study, we present a systematic analysis of a safe H9N2 influenza vaccine derived from MDCK cells for protecting chickens against influenza virus infection. In 2008, we isolated two novel H9N2 influenza viruses from chickens raised in southern China, and these H9N2 viruses were adapted to MDCK cells. The H9N2 virus was produced in MDCK cells in a scalable bioreactor, purified, inactivated, and investigated for use as a vaccine. The MDCK-derived H9N2 vaccine was able to induce high titers of neutralizing antibodies in chickens of different ages. Histopathological examination, direct immunofluorescence, HI assay, CD4(+)/CD8(+) ratio test, and cytokine evaluation indicated that the MDCK-derived H9N2 vaccine evoked a rapid and effective immune response to protect chickens from influenza infection. High titers of H9N2-specific antibodies were maintained in chickens for 5 months, and the MDCK-derived H9N2 vaccine had no effects on chicken growth. The use of MDCK cells in bioreactors for LPAIV vaccine production is an attractive option to prevent outbreaks of LPAIV in poultry.


Subject(s)
Biotechnology/methods , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza Vaccines/immunology , Influenza in Birds/prevention & control , Animals , Antibodies, Neutralizing , Bioreactors , Chickens/immunology , Chickens/virology , Culture Media, Serum-Free , Dogs , Fluorescent Antibody Technique, Direct , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/isolation & purification , Influenza in Birds/immunology , Madin Darby Canine Kidney Cells/virology , Phylogeny , Vaccination
3.
Mol Cancer ; 9: 112, 2010 May 19.
Article in English | MEDLINE | ID: mdl-20482842

ABSTRACT

BACKGROUND: Chronic myelogenous leukemia (CML) is characterized by the chimeric tyrosine kinase Bcr-Abl. Bcr-Abl-T315I is the notorious point mutation that causes resistance to imatinib and the second generation tyrosine kinase inhibitors, leading to poor prognosis. CML blasts have constitutive p65 (RelA NF-kappaB) transcriptional activity, and NF-kappaB may be a potential target for molecular therapies in CML that may also be effective against CML cells with Bcr-Abl-T315I. RESULTS: In this report, we discovered that pristimerin, a quinonemethide triterpenoid isolated from Celastraceae and Hippocrateaceae, inhibited growth and induced apoptosis in CML cells, including the cells harboring Bcr-Abl-T315I mutation. Additionally, pristimerin inhibited the growth of imatinib-resistant Bcr-Abl-T315I xenografts in nude mice. Pristimerin blocked the TNFalpha-induced IkappaBalpha phosphorylation, translocation of p65, and expression of NF-kappaB-regulated genes. Pristimerin inhibited two steps in NF-kappaB signaling: TAK1TauIKK and IKKTauIkappaBalpha. Pristimerin potently inhibited two pairs of CML cell lines (KBM5 versus KBM5-T315I, 32D-Bcr-Abl versus 32D-Bcr-Abl-T315I) and primary cells from a CML patient with acquired resistance to imatinib. The mRNA and protein levels of Bcr-Abl in imatinib-sensitive (KBM5) or imatinib-resistant (KBM5-T315I) CML cells were reduced after pristimerin treatment. Further, inactivation of Bcr-Abl by imatinib pretreatment did not abrogate the TNFalpha-induced NF-kappaB activation while silencing p65 by siRNA did not affect the levels of Bcr-Abl, both results together indicating that NF-kappaB inactivation and Bcr-Abl inhibition may be parallel independent pathways. CONCLUSION: To our knowledge, this is the first report to show that pristimerin is effective in vitro and in vivo against CML cells, including those with the T315I mutation. The mechanisms may involve inhibition of NF-kappaB and Bcr-Abl. We concluded that pristimerin could be a lead compound for further drug development to overcome imatinib resistance in CML patients.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Signal Transduction/drug effects , Triterpenes/pharmacology , Adolescent , Adult , Animals , Benzamides , Cell Proliferation/drug effects , Cell Separation , Child , Drug Resistance, Neoplasm/genetics , Electrophoretic Mobility Shift Assay , Enzyme Activation/drug effects , Flow Cytometry , Fluorescent Antibody Technique , Fusion Proteins, bcr-abl , Gene Expression/drug effects , Humans , Imatinib Mesylate , Immunohistochemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Male , Mice , Mice, Nude , Middle Aged , NF-kappa B/drug effects , NF-kappa B/metabolism , Pentacyclic Triterpenes , Piperazines/pharmacology , Point Mutation , Pyrimidines/pharmacology , RNA, Small Interfering , Transcription, Genetic/drug effects , Transfection , Xenograft Model Antitumor Assays , Young Adult
4.
Cancer Res ; 70(6): 2516-27, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20215516

ABSTRACT

NF-kappaB may be a potential therapeutic target for acute myelogenous leukemia (AML) because NF-kappaB activation is found in primitive human AML blast cells. In this report, we initially discovered that the potent antineoplastic effect of niclosamide, a Food and Drug Administration-approved antihelminthic agent, was through inhibition of the NF-kappaB pathway in AML cells. Niclosamide inhibited the transcription and DNA binding of NF-kappaB. It blocked tumor necrosis factor-induced IkappaBalpha phosphorylation, translocation of p65, and expression of NF-kappaB-regulated genes. Niclosamide inhibited the steps TAK1-->IkappaB kinase (IKK) and IKK-->IkappaBalpha. Niclosamide also increased the levels of reactive oxygen species (ROS) in AML cells. Quenching ROS by the glutathione precursor N-acetylcysteine attenuated niclosamide-induced apoptosis. Our results together suggest that niclosamide inhibited the NF-kappaB pathway and increased ROS levels to induce apoptosis in AML cells. On translational study of the efficacy of niclosamide against AML, niclosamide killed progenitor/stem cells from AML patients but spared those from normal bone marrow. Niclosamide was synergistic with the frontline chemotherapeutic agents cytarabine, etoposide, and daunorubicin. It potently inhibited the growth of AML cells in vitro and in nude mice. Our results support further investigation of niclosamide in clinical trials of AML patients.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , NF-kappa B/metabolism , Niclosamide/pharmacology , Reactive Oxygen Species/metabolism , Adult , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Drug Synergism , Genes, Reporter , HL-60 Cells , Humans , I-kappa B Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Inbred BALB C , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Neoplastic Stem Cells , Niclosamide/administration & dosage , Protein Serine-Threonine Kinases/metabolism , Transcription Factor RelA/metabolism , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology , U937 Cells
5.
Cancer Biol Ther ; 9(8): 640-50, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20160501

ABSTRACT

Accumulating evidence indicates that survivin plays a pivotal role in not only cell survival but also cell cycle progression. Op18/stathmin is an oncoprotein that regulates microtubule stabilization. Both survivin and Op18 have been proposed as therapeutic targets for cancer. However, few small molecule inhibitors of survivin and Op18 have been reported. In this study, we have identified a novel small molecule compound (GDP366) which potently and selectively inhibited the expression of both survivin and Op18. It decreased both the mRNA and protein levels of survivin and Op18. This inhibitory effect was not dependent on the status of p53 and p21 although GDP366 potently increased p53 and p21 levels. GDP366 significantly inhibited the growth of tumor cells in vitro and in vivo (nude mouse model) without rapid induction of apoptosis. GDP366 induced polyploidy in multiple types of cancer cell lines. GDP366 increased chromosomal instability, and induced cellular senescence by inhibiting telomerase activity. We conclude that GDP366 is a novel dual inhibitor of survivin and Op18. Our results warrant further translational evaluation of this compound.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Colorectal Neoplasms/drug therapy , Microtubule-Associated Proteins/antagonists & inhibitors , Phenylurea Compounds/pharmacology , Stathmin/antagonists & inhibitors , Animals , Cellular Senescence/drug effects , Colorectal Neoplasms/pathology , HCT116 Cells , HeLa Cells , Humans , Inhibitor of Apoptosis Proteins , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Survivin , Xenograft Model Antitumor Assays
6.
Bioorg Med Chem ; 18(5): 1806-15, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20149665

ABSTRACT

Imatinib (STI571) is the frontline targeted-therapeutic agent for patients with chronic myelogenous leukemia (CML). However, resistance to imatinib due to point mutations in Bcr-Abl kinase domain is an emerging problem. We recently reported that triptolide (compound 1) could effectively kill CML cells including those harboring T315I mutant Bcr-Abl. In the present study, we designed a series of C-14 triptolide derivatives with C-14-hydroxyl substituted by different amine esters (3-18): 3-6 and 13 (by aliphatic chain amine esters); 7-9, 11, 12 and 15-18 (by alicyclic amine esters with different size), and 10 and 14 (by aralkylamine esters).The compounds were examined for their antineoplastic activity against CML cells (including KBM5-T315I cells) in terms of proliferation inhibition, apoptosis and signal transduction. Nude mouse xenograft model was also used to evaluate the in vivo activity. Compounds 2-9, 11-14, 17 and 18 exhibited a potent inhibitory activity against KBM5 and KBM5-T315I cells. This series of derivatives down-regulated Bcr-Abl mRNA. Compounds 4, 5, 8 and 9 were further examined for their impact on signaling and apoptosis with immunoblotting. Compound 5 was chosen for evaluation in a nude mouse xenograft model. The stereo-hindrance of C-14 group appeared to be responsible for the antitumor effect. The computational small molecule-protein docking analysis illustrated the possible interaction between compound 9 and RNA polymerase II. Our results suggest that this series of derivatives may be promising agents to overcome imatinib-resistance caused by the Bcr-Abl-T315I mutation.


Subject(s)
Antineoplastic Agents/chemical synthesis , Diterpenes/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Phenanthrenes/chemistry , Amino Acid Substitution , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis , Benzamides , Binding Sites , Cell Line , Computer Simulation , Diterpenes/chemical synthesis , Diterpenes/therapeutic use , Down-Regulation , Drug Design , Drug Resistance, Neoplasm , Epoxy Compounds/chemical synthesis , Epoxy Compounds/chemistry , Epoxy Compounds/therapeutic use , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Mice , Mice, Nude , Phenanthrenes/chemical synthesis , Phenanthrenes/therapeutic use , Piperazines/therapeutic use , Pyrimidines/therapeutic use , RNA, Messenger/metabolism , Signal Transduction , Water/chemistry , Xenograft Model Antitumor Assays
7.
Mol Cancer Ther ; 9(1): 211-23, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20053766

ABSTRACT

Gain-of-function mutations of the receptor tyrosine kinase KIT play a critical role in the pathogenesis of systemic mastocytosis (SM) and gastrointestinal stromal tumors. The various juxtamembrane type of KIT mutations, including V560G, are found in 60% to 70% of patients with gastrointestinal stromal tumors; loop mutant D816V, which exists in approximately 80% of SM patients, is completely resistant to imatinib. In the present study, we hypothesized that homoharringtonine (HHT), a protein synthesis inhibitor, would decrease the level of KIT protein by inhibiting translation, resulting in a decreased level of phospho-KIT and abrogating its constitutive downstream signaling. Imatinib-sensitive HMC-1.1 cells harboring the mutation V560G in the juxtamembrane domain of KIT, imatinib-resistant HMC-1.2 cells harboring both V560G and D816V mutations, and murine P815 cells were treated with HHT and analyzed in terms of growth, apoptosis, and signal transduction. The in vivo antitumor activity was evaluated by using the murine mast cell leukemia model. Our results indicated that HHT effectively inhibited the growth and induced apoptosis in cells bearing both V560G and D816V or D814Y KIT. Additionally, HHT inhibited the KIT-dependent phosphorylation of downstream signaling molecules Akt, signal transducer and activator of transcription 3 and 5, and extracellular signal-regulated kinase 1/2. Furthermore, HHT significantly prolonged the survival duration of mice with aggressive SM or mast cell leukemia by inhibiting the expansion and infiltration of imatinib-resistant mast tumor cells harboring imatinib-resistant D814Y KIT. Collectively, we show that HHT circumvents D816V KIT-elicited imatinib resistance. Our findings warrant a clinical trial of HHT in patients with SM harboring D816V or D814Y KIT.


Subject(s)
Amino Acid Substitution/genetics , Antineoplastic Agents/pharmacology , Harringtonines/pharmacology , Mast Cells/drug effects , Mast Cells/metabolism , Mutation/genetics , Proto-Oncogene Proteins c-kit/genetics , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Gene Silencing/drug effects , Homoharringtonine , Humans , Mast Cells/cytology , Mice , Myeloid Cell Leukemia Sequence 1 Protein , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Survival Analysis
8.
Cancer Lett ; 291(2): 246-55, 2010 May 28.
Article in English | MEDLINE | ID: mdl-19942343

ABSTRACT

The discovery of oncogene addiction in myeloproliferative disorders (MPDs) driven by the gain-of-function mutant Jak2V617F has attracted intense interest in targeted therapy for MPDs. In this report, we demonstrate that triptolide potently downregulated the transcription of Jak2 by inhibiting the activity of RNA polymerase. Triptolide inhibited the in vitro and in vivo growth of tumor cells harboring Jak2V617F. Triptolide induced abundant apoptosis with a prominent decline of Bcl-2, Bcl-X(L), survivin and Mcl-1. As well, triptolide induced caspase-3-dependent Mcl-1 cleavage, which may potentiate apoptosis. These findings suggest that triptolide is a promising agent to kill Jak2V617F-harboring cells.


Subject(s)
Caspase 3/metabolism , Diterpenes/pharmacology , Janus Kinase 2/genetics , Myeloproliferative Disorders/genetics , Phenanthrenes/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Transcription, Genetic/drug effects , Amino Acid Substitution , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Caspase Inhibitors , Cell Cycle/drug effects , Cell Line, Tumor , Cycloheximide/pharmacology , DNA Primers , Epoxy Compounds/pharmacology , Flow Cytometry , Humans , Leukemia, Erythroblastic, Acute/genetics , Myeloid Cell Leukemia Sequence 1 Protein , RNA, Messenger/drug effects , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects
9.
Cancer Lett ; 290(2): 182-91, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-19819619

ABSTRACT

T315I Bcr-Abl in chronic myelogenous leukemia (CML) is the most notorious point mutations to elicit acquired resistance to imatinib. In the present study, we investigated the effect of celastrol on CML cells bearing wild-type Bcr-Abl or T315I-mutant. The results revealed that celastrol potently downregulated the protein levels of Bcr-Abl, and inhibited the growth in CML cells in vitro and in nude mouse xenografts regardless of Bcr-Abl mutation status. Celastrol induced mitochondrial-dependent apoptosis. In conclusion, celastrol exhibits potent activity against CML cells bearing wild-type Bcr-Abl or -the T315I-mutant.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Fusion Proteins, bcr-abl/drug effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Triterpenes/pharmacology , Animals , Benzamides , Blotting, Western , Cell Line, Tumor , Cell Separation , Drug Resistance, Neoplasm/drug effects , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Imatinib Mesylate , Immunohistochemistry , Male , Mice , Mice, Nude , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Pentacyclic Triterpenes , Piperazines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
10.
Cancer Sci ; 100(11): 2210-7, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19671059

ABSTRACT

The pathogenesis of hypereosinophilic syndrome (HES) in some patients is highly dependent on FIP1-Like-1 (FIP1L1)-platelet-derived growth factor receptor alpha (PDGFRalpha), which can generate sustained activation signaling to maintain a cell malignant phenotype. HES usually shows good response to the tyrosine kinase inhibitor imatinib, but mutations in FIP1L1-PDGFRalpha (e.g. T674I) can confer acquired resistance to imatinib. An alternative therapeutic strategy other than with tyrosine kinase inhibitors is needed to overcome acquired drug resistance. We hypothesized that switching off the crucial chimeric oncoprotein FIP1L1-PDGFRalpha on which HES cells depend, should have deleterious effects on the cancer cells. We used low concentrations of triptolide, a transcription inhibitor, to shut down the expression of FIP1L1-PDGFRalpha. EOL-1 cells and BaF3 cells expressing wild-type or T674I FIP1L1-PDGFRalpha were treated with triptolide, and signaling pathways, cell cycling, and apoptosis were analyzed by RT-PCR, immunoblotting, and flow cytometry, respectively. The results revealed that at nanomolar concentrations triptolide decreased the levels of mRNA and protein of FIP1L1-PDGFRalpha and the growth of the neoplastic cells, regardless of the mutational status of PDGFRalpha. Triptolide also downregulated the signaling molecules Stat3, Akt, and Erk1/2, which are downstream from PDGFRalpha, and induced G1 cell-cycle arrest. Triptolide time- and dose-dependently induced apoptosis by decreasing the anti-apoptotic proteins Mcl-1 and Bcl-X(L),triggering the intrinsic apoptotic pathway. In conclusion, triptolide has potent activity against malignant cells in HES bearing FIP1L1-PDGFRalpha, regardless of its mutational status that confer acquired resistance to imatinib. Our results suggest that triptolide may be a promising agent in the treatment of HES.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Diterpenes/pharmacology , Hypereosinophilic Syndrome/drug therapy , Oncogene Proteins, Fusion/antagonists & inhibitors , Phenanthrenes/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/antagonists & inhibitors , mRNA Cleavage and Polyadenylation Factors/antagonists & inhibitors , Cell Cycle/drug effects , Cell Line , Cell Survival/drug effects , Epoxy Compounds/pharmacology , Humans , Hypereosinophilic Syndrome/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphorylation , RNA, Messenger/analysis , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , mRNA Cleavage and Polyadenylation Factors/genetics , mRNA Cleavage and Polyadenylation Factors/metabolism
11.
Cancer Sci ; 100(7): 1335-43, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19383029

ABSTRACT

Gain-of-function mutations of the receptor tyrosine kinase KIT can cause systemic mastocytosis (SM) and gastrointestinal stromal tumors. Most of the constitutively active KIT can be inhibited by imatinib; D816V KIT cannot. In this study, we investigated the activity of triptolide, a diterpenoid isolated from the Chinese herb Tripterygium wilfordii Hook. f., in cells expressing mutant KIT, including D816V KIT. Imatinib-sensitive HMC-1.1 cells harboring the mutation V560G in the juxtamembrane domain of KIT, imatinib-resistant HMC-1.2 cells harboring both V560G and D816V mutations, and murine P815 cells, were treated with triptolide, and analyzed in terms of growth, apoptosis, and signal transduction. The in vivo antitumor activity was evaluated by using the nude mouse xenograft model. Our results demonstrated that triptolide potently inhibits the growth of both human and murine mast cells harboring not only imatinib-sensitive KIT mutation but also imatinib-resistant D816V KIT. Triptolide markedly inhibited KIT mRNA levels and strikingly reduced the levels of phosphorylated and total Stat3, Akt, and Erk1/2, downstream targets of KIT. Triptolide triggered apoptosis by inducing depolarization of mitochondrial potential and release of cytochrome c, downregulation of Mcl-1 and XIAP. Furthermore, triptolide significantly abrogated the growth of imatinib-resistant HMC-1.2 cell xenografts in nude mice and decreased KIT expression in xenografts. Our data demonstrate that triptolide inhibits imatinib-resistant mast cells harboring D816V KIT. Further investigation of triptolide for treatment of human neoplasms driven by gain-of-function KIT mutations is warranted.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Diterpenes/pharmacology , Mast Cells/drug effects , Mutation , Phenanthrenes/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Animals , Down-Regulation , Epoxy Compounds/pharmacology , Humans , Male , Mast Cells/metabolism , Mice , Mice, Nude , Proto-Oncogene Proteins c-kit/metabolism , RNA, Messenger/metabolism
12.
Clin Cancer Res ; 15(5): 1686-97, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19240172

ABSTRACT

PURPOSE: Resistance to STI571 is an emerging problem for patients with chronic myelogenous leukemia (CML). Mutation in the kinase domain of Bcr-Abl is the predominant mechanism of the acquired resistance to STI571. In the present study, we investigated the effect of triptolide on cell survival or apoptosis in CML cells bearing Bcr-Abl-T315I or wild-type Bcr-Abl. EXPERIMENTAL DESIGN: CML cell lines (KBM5 versus KBM5-T315I, BaF3-Bcr-Abl versus BaF3-Bcr-Abl-T315I) and primary cells from CML patients with clinical resistance to STI571 were treated with triptolide, and analyzed in terms of growth, apoptosis, and signal transduction. Nude mouse xenograft model was also used to evaluate the antitumor activity. RESULTS: Triptolide potently down-regulated the mRNA and protein levels of Bcr-Abl independently of the caspase or proteosome activation in CML cells. It induced mitochondrial-dependent apoptosis in Bcr-Abl-T315I CML cells and primary cells from CML patients with clinical resistance to STI571. Additionally, triptolide inhibited the growth of STI571-sensitive KBM5 and STI571-resistant KBM5-T315I CML cells in nude mouse xenografts. Triptolide also down-regulated the expression of survivin, Mcl-1, and Akt in CML cells, which suggests that it may have multiple targets. CONCLUSIONS: These findings suggest that triptolide is a promising agent to overcome STI571-resistant CML cells, and warrant a clinical trial of triptolide derivatives for CML with Bcr-Abl-T315I mutation.


Subject(s)
Apoptosis/drug effects , Diterpenes/therapeutic use , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mutation/genetics , Phenanthrenes/therapeutic use , Transcription, Genetic/drug effects , Adolescent , Adult , Aged , Animals , Antineoplastic Agents, Alkylating/therapeutic use , Benzamides , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Epoxy Compounds/therapeutic use , Female , Flow Cytometry , Fusion Proteins, bcr-abl/antagonists & inhibitors , Fusion Proteins, bcr-abl/metabolism , Humans , Imatinib Mesylate , Immunoenzyme Techniques , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Myeloid Cell Leukemia Sequence 1 Protein , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrimidines/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tripterygium/chemistry , Tumor Cells, Cultured , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...