Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Acta Pharmacol Sin ; 42(4): 604-612, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32694757

ABSTRACT

The kinase FLT3 internal tandem duplication (FLT3-ITD) is related to poor clinical outcomes of acute myeloid leukemia (AML). FLT3 inhibitors have provided novel strategies for the treatment of FLT3-ITD-positive AML. But they are limited by rapid development of acquired resistance and refractory in monotherapy. Recent evidence shows that inducing the degradation of FLT3-mutated protein is an attractive strategy for the treatment of FLT3-ITD-positive AML, especially those with FLT3 inhibitor resistance. In this study we identified Wu-5 as a novel USP10 inhibitor inducing the degradation of FLT3-mutated protein. We showed that Wu-5 selectively inhibited the viability of FLT3 inhibitor-sensitive (MV4-11, Molm13) and -resistant (MV4-11R) FLT3-ITD-positive AML cells with IC50 of 3.794, 5.056, and 8.386 µM, respectively. Wu-5 (1-10 µM) dose-dependently induced apoptosis of MV4-11, Molm13, and MV4-11R cells through the proteasome-mediated degradation of FLT3-ITD. We further demonstrated that Wu-5 directly interacted with and inactivated USP10, the deubiquitinase for FLT3-ITD in vitro (IC50 value = 8.3 µM) and in FLT3-ITD-positive AML cells. Overexpression of USP10 abrogated Wu-5-induced FLT3-ITD degradation and cell death. Also, the combined treatment of Wu-5 and crenolanib produced synergistic cell death in FLT3-ITD-positive cells via the reduction of both FLT3 and AMPKα proteins. In support of this, AMPKα inhibitor compound C synergistically enhanced the anti-leukemia effect of crenolanib, while AMPKα activator metformin inhibited the anti-leukemia effect of crenolanib. In summary, we demonstrate that Wu-5, a novel USP10 inhibitor, can overcome FLT3 inhibitor resistance and synergistically enhance the anti-AML effect of crenolanib through targeting FLT3 and AMPKα pathway.


Subject(s)
Apoptosis/drug effects , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Signal Transduction/drug effects , Thiophenes/pharmacology , Ubiquitin Thiolesterase/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Cell Line, Tumor , Drug Synergism , Humans , Leukemia, Myeloid, Acute/drug therapy , Piperidines/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/metabolism
2.
Acta Pharmacol Sin ; 39(3): 492-498, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29168472

ABSTRACT

Deubiquitinating protease USP7 is a promising therapeutic target for cancer treatment, and interest in developing USP7 inhibitors has greatly increased. In the present study, we reported a series of natural pentacyclic triterpenes with USP7 inhibitory activity in vitro. Among them, both the ursane triterpenes and oleanane triterpenes were more active than the lupine triterpenes, whereas ursolic acid was the most potent with IC50 of 7.0±1.5 µmol/L. Molecular docking studies showed that ursolic acid might occupy the ubiquitin binding pocket of USP7, with the 17-carboxyl group and 3-hydroxyl group playing a vital role in the USP7-ursolic acid interaction. Using the cellular thermal shift assay, we demonstrated that ursolic acid interacted with USP7 in RPMI8226 human myeloma cells. Ursolic acid dose-dependently inhibited the proliferation of the myeloma cells with IC50 of 6.56 µmol/L, accompanied by reductions in USP7 substrates such as MDM2, UHRF1 and DNMT1. Overexpression of USP7 partially, but significantly attenuated ursolic acid-induced cell death as well as downregulation of MDM2, UHRF1 and DNMT1. In conclusion, we demonstrate for the first time that pentacyclic triterpenes represent a novel scaffold for developing USP7 inhibitors and that USP7 inhibition contributes to the anti-cancer effect of ursolic acid.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Pentacyclic Triterpenes/pharmacology , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , Structure-Activity Relationship , Triterpenes/antagonists & inhibitors , Triterpenes/pharmacology , Ubiquitin-Protein Ligases , Ubiquitin-Specific Peptidase 7/biosynthesis , Ursolic Acid
3.
J Proteome Res ; 11(3): 1773-81, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22268729

ABSTRACT

FBOX6 ubiquitin ligase complex is involved in the endoplasmic reticulum-associated degradation pathway by mediating the ubiquitination of glycoproteins. FBXO6 interacts with the chitobiose in unfolded N-glycoprotein, pointing glycoproteins toward E2 for ubiquitination. Although the glycoprotein-recognizing mechanism of FBXO6 is well documented, its bona fide interacting glycoproteins are largely unknown. Here we utilized a protein purification approach combined with LC-MS to systematically identify the FBXO6-interacting glycoproteins. Following identification of 39 proteins that specifically interact with FBXO6 in all three different cell lines, 293T, HeLa and Jurkat cells, we compared the protein complex organization between wild-type FBXO6 and its mutant, which fails to recognize glycoproteins. Combining these databases, 29 highly confident glycoproteins that interact with FBXO6 in an N-glycan dependent manner are identified. Our data provide valuable information for the discovery of the interacting glycoproteins of FBXO6 and also demonstrate the potential of these approaches as general platforms for the global discovery of interacting glycoproteins of other FBAs (F-box associated regions) containing F-box proteins.


Subject(s)
Glycoproteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Cullin Proteins/metabolism , Glycoproteins/isolation & purification , HEK293 Cells , HeLa Cells , Humans , Jurkat Cells , Membrane Glycoproteins/metabolism , Oxidoreductases/metabolism , Protein Binding , Protein Interaction Mapping , Protein Interaction Maps , Proteomics
4.
Blood ; 116(24): 5289-97, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20739655

ABSTRACT

All-trans retinoic acid (ATRA), a natural ligand for the retinoic acid receptors (RARs), induces clinical remission in most acute promyelocytic leukemia (APL) patients through the induction of differentiation and/or eradication of leukemia-initiating cells. Here, we identify a novel natural ent-kaurene diterpenoid derived from Isodon pharicus leaves, called pharicin B, that can rapidly stabilize RAR-α protein in various acute myeloid leukemic (AML) cell lines and primary leukemic cells from AML patients, even in the presence of ATRA, which is known to induce the loss of RAR-α protein. Pharicin B also enhances ATRA-dependent the transcriptional activity of RAR-α protein in the promyelocytic leukemia-RARα-positive APL cell line NB4 cells. We also showed that pharicin B presents a synergistic or additive differentiation-enhancing effect when used in combination with ATRA in several AML cell lines and, especially, some primary leukemic cells from APL patients. In addition, pharicin B can overcome retinoid resistance in 2 of 3 NB4-derived ATRA-resistant subclones. These findings provide a good example for chemical biology-based investigations of pathophysiological and therapeutic significances of RAR-α and PML-RAR-α proteins. The effectiveness of the ATRA/pharicin B combination warrants further investigation on their use as a therapeutic strategy for AML patients.


Subject(s)
Cell Differentiation/drug effects , Diterpenes, Kaurane/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Receptors, Retinoic Acid/chemistry , Tretinoin/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Line, Tumor , Diterpenes, Kaurane/therapeutic use , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Humans , Isodon/chemistry , Leukemia, Myeloid, Acute/pathology , Protein Stability/drug effects , Retinoic Acid Receptor alpha , Transcriptional Activation/drug effects , Tretinoin/therapeutic use , Tumor Cells, Cultured
5.
Biochem Biophys Res Commun ; 406(3): 430-4, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21329675

ABSTRACT

Ikaros is an important transcription factor involved in the development and differentiation of hematopoietic cells. In this work, we found that chemotherapeutic drugs or ultraviolet radiation (UV) treatment could reduce the expression of full-length Ikaros (IK1) protein in less than 3h in leukemic NB4, Kasumi-1 and Jurkat cells, prior to the activation of caspase-3. Etoposide treatment could not alter the mRNA level of IK1 but it could shorten the half-life of IK1. Co-treatment with the proteasome inhibitor MG132 or epoxomicin but not calpain inhibitor calpeptin inhibited etoposide-induced Ikaros downregulation. Overexpression of IK1 could accelerate etoposide-induced apoptosis in NB4 cells, as evidenced by the increase of Annexin V positive cells and the more early activation of caspase 3. To our knowledge, this is the first report to show that upon chemotherapy drugs or UV treatment, IK1 could be degraded via the proteasome system in the early phase of apoptosis induction. These data might shed new insight on the role of IK1 in apoptosis and the post-translational regulation of IK1.


Subject(s)
Apoptosis , Ikaros Transcription Factor/metabolism , Proteasome Endopeptidase Complex/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Caspase 3/metabolism , Cell Line, Tumor , Down-Regulation , Etoposide/pharmacology , Humans , Jurkat Cells , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/radiation effects , Ubiquitin/metabolism , Ultraviolet Rays
6.
Nat Commun ; 12(1): 51, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33397955

ABSTRACT

Identifying novel drug targets to overcome resistance to tyrosine kinase inhibitors (TKIs) and eradicating leukemia stem/progenitor cells are required for the treatment of chronic myelogenous leukemia (CML). Here, we show that ubiquitin-specific peptidase 47 (USP47) is a potential target to overcome TKI resistance. Functional analysis shows that USP47 knockdown represses proliferation of CML cells sensitive or resistant to imatinib in vitro and in vivo. The knockout of Usp47 significantly inhibits BCR-ABL and BCR-ABLT315I-induced CML in mice with the reduction of Lin-Sca1+c-Kit+ CML stem/progenitor cells. Mechanistic studies show that stabilizing Y-box binding protein 1 contributes to USP47-mediated DNA damage repair in CML cells. Inhibiting USP47 by P22077 exerts cytotoxicity to CML cells with or without TKI resistance in vitro and in vivo. Moreover, P22077 eliminates leukemia stem/progenitor cells in CML mice. Together, targeting USP47 is a promising strategy to overcome TKI resistance and eradicate leukemia stem/progenitor cells in CML.


Subject(s)
Drug Resistance, Neoplasm , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/pharmacology , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Specific Proteases/metabolism , Animals , Cell Proliferation/drug effects , DNA Damage , DNA Repair/drug effects , Drug Resistance, Neoplasm/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fusion Proteins, bcr-abl , Gene Expression Regulation, Leukemic/drug effects , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice, Knockout , Proteasome Endopeptidase Complex/metabolism , Protein Binding/drug effects , Protein Stability/drug effects , Proteolysis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects , Thiophenes/pharmacology , Xenograft Model Antitumor Assays , Y-Box-Binding Protein 1/metabolism , ras Proteins/metabolism
7.
Cell Cycle ; 17(24): 2779-2789, 2018.
Article in English | MEDLINE | ID: mdl-30526252

ABSTRACT

The spindle assembly checkpoint prevents chromosome mis-segregation during mitosis by delaying sister chromatid separation. Several F-box protein members play critical roles in maintaining genome stability and regulating cell cycle progress via ubiquitin-mediated protein degradation. Here, we showed that Fbxo6 critically regulated spindle checkpoint and chromosome segregation. Fbxo6 was phosphorylated during mitosis. Overexpression of Fbxo6 lead to faster exit from nocodazole-induced mitosis arrest through premature sister chromatid separation. Moreover, we found substantially more binuclear and multilobed nuclei cells accompanied with impaired cell viability in Fbxo6-overexpressed HeLa cells. Mechanistically, Fbxo6 interacted with spindle checkpoint proteins including Mad2 and BubR1 leading to the premature exit from mitosis. Overall, we revealed a novel role of Fbxo6 in regulating spindle checkpoint, which may shed light on the regulation of genome instability of cancer cells.


Subject(s)
Mad2 Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Chromatids/metabolism , Genomic Instability , HeLa Cells , Humans , M Phase Cell Cycle Checkpoints/drug effects , Mitosis/drug effects , Nocodazole/pharmacology , Phosphorylation/drug effects , SKP Cullin F-Box Protein Ligases/genetics , Spindle Apparatus/metabolism
8.
PLoS One ; 10(7): e0132337, 2015.
Article in English | MEDLINE | ID: mdl-26134508

ABSTRACT

Synthetic triterpenoid methyl-2-cyano-3, 12-dioxooleana-1, 9(11)-dien-28-oate (CDDO-Me) has been shown as a promising agent against ovarian cancer. However, the underlying mechanism is not well understood. Here, we demonstrate that CDDO-Me directly interacts with Hsp90 in cells by cellular thermal shift assay. CDDO-Me treatment leads to upregulation of Hsp70 and degradation of Hsp90 clients (ErbB2 and Akt), indicating the inhibition of Hsp90 by CDDO-Me in cells. Knockdown of Hsp90 significantly inhibits cell proliferation and enhances the anti-proliferation effect of CDDO-Me in H08910 ovarian cancer cells. Dithiothreitol inhibits the interaction of CDDO-Me with Hsp90 in cells and abrogates CDDO-Me induced upregulation of Hsp70, degradation of Akt and cell proliferation inhibition. This suggests the anti-ovarian cancer effect of CDDO-Me is possibly mediated by the formation of Michael adducts between CDDO-Me and reactive nucleophiles on Hsp90. This study identifies Hsp90 as a novel target protein of CDDO-Me, and provides a novel insight into the mechanism of action of CDDO-Me in ovarian cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Oleanolic Acid/analogs & derivatives , Ovarian Neoplasms/pathology , Antineoplastic Agents/chemistry , Cell Division/drug effects , Cell Line, Tumor , Dithiothreitol/pharmacology , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, erbB-2 , HSP70 Heat-Shock Proteins/biosynthesis , HSP70 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/genetics , Humans , MAP Kinase Signaling System/drug effects , Molecular Structure , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Oleanolic Acid/antagonists & inhibitors , Oleanolic Acid/chemistry , Oleanolic Acid/pharmacology , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , RNA, Small Interfering/genetics , Receptor, ErbB-2/biosynthesis , Structure-Activity Relationship , Transfection , Up-Regulation/drug effects
9.
PLoS One ; 9(8): e104985, 2014.
Article in English | MEDLINE | ID: mdl-25115845

ABSTRACT

Despite considerable efficacy of arsenic trioxide (As2O3) in acute promyelocytic leukemia (APL) treatment, other non-APL leukemias, such as chronic myeloid leukemia (CML), are less sensitive to As2O3 treatment. However, the underlying mechanism is not well understood. Here we show that relative As2O3-resistant K562 cells have significantly lower ROS levels than As2O3-sensitive NB4 cells. We compared the expression of several antioxidant enzymes in these two cell lines and found that peroxiredoxin 1/2/6 and catalase are expressed at high levels in K562 cells. We further investigated the possible role of peroxirdoxin 1/2/6 and catalase in determining the cellular sensitivity to As2O3. Interestingly, knockdown of peroxiredoxin 1/2/6 did not increase the susceptibility of K562 cells to As2O3. On the contrary, knockdown of catalase markedly enhanced As2O3-induced apoptosis. In addition, we provide evidence that overexpression of BCR/ABL cannot increase the expression of PRDX 1/2/6 and catalase. The current study reveals that the functional role of antioxidant enzymes is cellular context and treatment agents dependent; targeting catalase may represent a novel strategy to improve the efficacy of As2O3 in CML treatment.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Catalase/antagonists & inhibitors , Oxides/pharmacology , Peroxiredoxins/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Apoptosis/genetics , Apoptosis/physiology , Arsenic Trioxide , Catalase/genetics , Catalase/metabolism , Cell Line, Tumor , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Knockdown Techniques , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Leukemia, Promyelocytic, Acute/pathology , Peroxiredoxins/genetics , Peroxiredoxins/metabolism , Reactive Oxygen Species/metabolism
10.
Chinese Journal of Hematology ; (12): 294-300, 2019.
Article in Zh | WPRIM | ID: wpr-1011978

ABSTRACT

Objective: To compare the clinical efficacy of umbilical cord blood transplantation (UCBT) and hematopoietic stem cell transplantation from HLA-matched sibling donors (MSD-HSCT) in the treatment of myelodysplastic syndrome-EB (MDS-EB) or acute myeloid leukemia with myelodysplasia-related changes (AML-MRC) . Methods: A cohort of 64 patients (including 38 cases of MDS-EB and 26 cases of AML-MRC) who received UCBT/MSD-HSCT from February 2011 to December 2017 were retrospectively analyzed. Results: ①Compared with MSD-HSCT group, UCBT group had a higher proportion of AML-MRC patients [52.8% (19/36) vs 25.0% (7/28) , P=0.025], and a lower median age [13 (1.5-52) years vs 32 (10-57) years, P=0.001]. ②The engraftment of neutrophils both in UCBT and MSD-HSCT groups on +42 d was 100%, and the median engraftment time was 17.5 (11-31) d and 11.5 (10-20) d, respectively. The engraftment of platelet at +100 d in UCBT group was 91.4%, the median engraftment time was 40 (15-96) d; The engraftment of platelet at +100 d in MSD-HSCT group was 100%, and the median engraftment time was 15 (11-43) d. ③There were no statistically significant differences in terms of the cumulative incidence of Ⅱ-Ⅳ and Ⅲ/Ⅳ aGVHD of 100 d and transplant related mortality (TRM) of 180 d, relapse rate, overall survival (OS) , disease-free survival (DFS) between UCBT and MSD-HSCT groups (P>0.05) . ④The 3-year cumulative incidence of chronic GVHD (cGVHD) and severe chronic GVHD in UCBT group were lower than of MSD-HSCT group [28.3% (95%CI 13.4%-45.3%) vs 67.9% (95%CI 46.1%-82.4%) , P=0.002; 10.3% (95%CI 2.5%-24.8%) vs 50.0% (95%CI 30.0%-67.1%) , respectively, P<0.001]. The cumulative 3-year incidence of GVHD-free and relapse-free survival (GRFS) of UCBT group was significantly higher than of MSD-HSCT group [55.0% (95%CI 36.0%-70.6%) vs 28.6% (95%CI 13.5%-45.6%) , P=0.038]. Conclusion: UCBT could obtain better quality of life after transplantation than MSD-HSCT in treatment of MDS-EB/AML-MRC.


Subject(s)
Adolescent , Adult , Humans , Middle Aged , Young Adult , Cord Blood Stem Cell Transplantation , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/therapy , Quality of Life , Retrospective Studies , Siblings
11.
Journal of Experimental Hematology ; (6): 1246-1252, 2019.
Article in Zh | WPRIM | ID: wpr-775733

ABSTRACT

OBJECTIVE@#To analyze the clinical outcomes of engraftment, graft-versus-host disease (GVHD) and survival in the patients with AML1-ETO positive acute myeloid leukemia (AML) treated with unrelated umbilical cord blood transplantation (UCBT).@*METHODS@#Forty-Five patients with high-risk refractory AML1-ETO positive AML were treated with a single UCBT in a single center from July 2010 to April 2018. All the patients underwent a myeloablative preconditioning regimen,and cyclosporine A (CSA) combined with mycophenolate mofetil (MMF) was used to prevent GVHD.@*RESULTS@#The median value of total nucleated cells (TNC) in cord blood was 5.21 (1.96-12.68)×10/kg recipient body weight, and that of CD34+ cells was 5.61 (0.56-15.4)×10/kg recipient weight. The implantation rate of neutrophil at 42 d and that of platelet at 120 d were 95.6% and 86.7%, respectively. The median time of absolute neutrophil count (ANC)>0.5×10/L and platelet 20×10/L were 16 (12-18) d and 37 (17-140) d after transplantation, respectively. The cumulative incidence of Ⅰ -Ⅳ grade acute GVHD (aGVHD) at 100 d after transplantation was 48.9% (95% CI 33.5%-62.6%), Ⅱ-Ⅳ grade aGVHD occurred in 12 cases (33.3%) (95% CI 20%-47.2%) , and Ⅲ-Ⅳ grade a GVHD in 8 cases (20%) (95% CI 9.8% -32.8%). In 5 cases of 40 patients survived over 100 days, the chronic GVHD (cGVHD) occurred after transplantation, among which 4 were localized, and 1 was extensive. 3 patients relapsed, and the 2-year cumulative relapse rate was 9.5% (95% CI 2.4%-22.8%). The median follow-up time was 23.5 (0.9-89.67) months, 10 patients died, 2-year disease-free survival rate (DFS) was 72.7%, and overall survival rate (OS) was 75.5%. Multivariate analysis showed that Ⅲ-Ⅳ. acute GVHD (aGVHD) affected overall survival.@*CONCLUSION@#UCBT is an effective rescue treatment for patients with high-risk refractory AML1-ETO positive AML.


Subject(s)
Humans , Cord Blood Stem Cell Transplantation , Core Binding Factor Alpha 2 Subunit , Graft vs Host Disease , Leukemia, Myeloid, Acute , Mycophenolic Acid , Oncogene Proteins, Fusion , Peripheral Blood Stem Cell Transplantation , RUNX1 Translocation Partner 1 Protein , Transplantation Conditioning
12.
Chinese Journal of Hematology ; (12): 105-109, 2018.
Article in Zh | WPRIM | ID: wpr-1011705

ABSTRACT

Objective: To explore the clinical efficacy and safety of unrelated umbilical cord blood transplantation (UCBT) in the treatment of refractory and relapsed acute leukemia (AL) patients. Methods: The clinical data of 22 refractory and relapsed AL patients who were treated with UCBT as salvage therapy from November 2009 to May 2017 were retrospectively analyzed. All patients received a myeloablative conditioning regimen for prevention of graft-versus-host disease (GVHD) with cyclosporine A (CSA)/short course of mycophenolate mofetil (MMF). Results: ①Of 22 patients, 9 cases were male and 13 female. The median age was 23 (15-44) years and median weight of 52.5 (43-82) kg. All patients were transplanted with a median umbilical cord blood nucleated cells of 3.07 (1.71-5.30)×107/kg (by weight), the median CD34+ cells was 1.60 (0.63-3.04)×105/kg (by weight). ②The myeloid cumulative implantation rate was 95.5% (95%CI 45.2-99.7%) after transplantation of 42 d, with the median implantation time of 19 (13-27) d. The platelet cumulative implantation rate after transplantation of 120 d was 81.8% (95%CI 54.2-93.6%), the median implantation time of 42 (20-164) d. ③The incidence of Ⅱ-Ⅳ, Ⅲ-Ⅳ aGVHD and the 2 year cumulative incidence of cGVHD were 36.4%, 13.6% and 40.3% respectively. ④ The transplant related mortality (TRM) after transplantation of 180d was 22.7%, 2 year cumulative rate of relapse was 18.7% (95%CI 3.6-42.5%), 2 year disease-free survival rate (DFS) and overall survival rate (OS) were 53.7% and 58.1%, respectively. Conclusion: The preliminary results show that the use of UCBT is safe and effective for refractory and relapsed AL patients who fail to respond to conventional chemotherapy.


Subject(s)
Adolescent , Adult , Female , Humans , Male , Young Adult , Acute Disease , Cord Blood Stem Cell Transplantation , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Leukemia/therapy , Peripheral Blood Stem Cell Transplantation , Retrospective Studies , Transplantation Conditioning
13.
Oncol Rep ; 28(4): 1399-405, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22859015

ABSTRACT

The transcription factor Ikaros was originally found to function as a key regulator of lymphocyte differentiation. In this study, we provide the first evidence that Ikaros is expressed at higher levels in ovarian cancer tissues compared with normal ovarian tissues and is significantly associated with high FIGO stage and low differentiation state in ovarian serous adenocarcinoma. To this end, we transfected IK1 (full length of Ikaros) into the SKOV3 ovarian cancer cell line and examined cell biological behaviors including proliferation, migration and invasion. We found that overexpression of IK1 inhibited cell proliferation by inducing G1 arrest, accompanied by the upregulation of P27 and P21 and downregulation of cyclin D1 and D2. On the other hand, IK1 increased the migration and invasion of ovarian cancer cells, as assessed by scratch-wound assay, transwell migration assay, and invasion assay. Overexpression of IK1 significantly increased Slug but not Snail1 expression at both mRNA and protein levels. It also downregulated and upregulated E-cadherin and MMP-2, two target genes of Slug involved in migration, respectively. Furthermore, knocking down Slug abrogated IK1-mediated increase in migration and invasion. These data suggest that Slug plays an important role in IK1-induced migration and invasion. In conclusion, we show for the first time that IK1 plays a dual role in the proliferation, migration and invasion of ovarian cancer cells, providing new insights into their metastasis.


Subject(s)
Cystadenocarcinoma, Serous/pathology , Ikaros Transcription Factor/metabolism , Ovarian Neoplasms/pathology , Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cystadenocarcinoma, Serous/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Ikaros Transcription Factor/genetics , Neoplasm Metastasis/genetics , Ovarian Neoplasms/metabolism , Reference Values , Snail Family Transcription Factors , Transcription Factors/genetics , Up-Regulation
14.
FEBS Lett ; 585(2): 375-80, 2011 Jan 21.
Article in English | MEDLINE | ID: mdl-21176776

ABSTRACT

RIG-G is a retinoic acid- or interferon-induced gene with potential anti-proliferation function. However, the mechanism underlying ATRA-induced RIG-G induction is not completely understood. Here, we demonstrate that ATRA up-regulates the expression of PU.1, which in turn directly binds to the promoter and increases the expression of RIG-G gene. Luciferase reporter assay and electrophoretic mobility shift assay reveal that PU.1 preferentially binds to one of the two putative binding sites on the RIG-G promoter. Moreover, silencing of PU.1 by shRNA markedly inhibited ATRA- but not IFNα-induced expression of RIG-G. These data provide new insight into the mechanism of ATRA-induced RIG-G expression.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Leukemia/metabolism , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Tretinoin/pharmacology , Cell Line, Tumor , Humans , Interferon-alpha/pharmacology , Leukemia/pathology , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/pharmacology , Transcriptional Activation/drug effects
15.
Cell Cycle ; 9(14): 2897-907, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20603598

ABSTRACT

In this study, we report the functional characterization of a new ent-kaurene diterpenoid termed pharicin A, which was originally isolated from Isodon, a perennial shrub frequently used in Chinese folk medicine for tumor treatment. Pharicin A induces mitotic arrest in leukemia and solid tumor-derived cells identified by their morphology, DNA content and mitotic marker analyses. Pharicin A-induced mitotic arrest is associated with unaligned chromosomes, aberrant BubR1 localization and deregulated spindle checkpoint activation. Pharicin A directly binds to BubR1 in vitro, which is correlated with premature sister chromatid separation in vivo. Pharicin A also induces mitotic arrest in paclitaxel-resistant Jurkat and U2OS cells. Combined, our study strongly suggests that pharicin A represents a novel class of small molecule compounds capable of perturbing mitotic progression and initiating mitotic catastrophe, which merits further preclinical and clinical investigations for cancer drug development.


Subject(s)
Antineoplastic Agents/pharmacology , Diterpenes, Kaurane/isolation & purification , Diterpenes, Kaurane/pharmacology , Mitosis/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , CDC2 Protein Kinase/metabolism , Chromatids/drug effects , Diterpenes, Kaurane/chemistry , Humans , Isodon/chemistry , Jurkat Cells , Medicine, Chinese Traditional , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/metabolism
16.
Chem Biol Interact ; 183(1): 222-30, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-19781537

ABSTRACT

The treatment outcome of acute lymphoblastic leukemia (ALL) has improved steadily over the last 50 years. However, the cure rates are unlikely to be raised further with current therapies. Since increasing the dosage of chemotherapeutic agents could also elevate toxicity, a solution to how one could achieve maximum therapeutic effect with the minimum dosage possible is imminent. One possibility is the employment of combination drug therapies. Arsenic trioxide (ATO) is a widely used drug for acute promyelocytic leukemia (APL). Its combination with other drugs presented therapeutic activities in malignant cancers other than APL. Considering the fact that ATO induces mitotic arrest prior to apoptosis induction, we attempted to investigate the potential anti-cancer effects of ATO in combination with the microtubule-stabilizing agent, paclitaxel (PTX), using malignant lymphocytes as in vitro models. Three malignant lymphocytic cell lines and primary cells were treated with ATO and/or PTX. Using the Chou-Talalay analysis for evaluation of combined effect of ATO and PTX, we found a synergistic effect of the two drugs in the inhibition of cell growth. We also found that the combination of ATO and PTX at low concentrations synergistically induced mitotic arrest followed by apoptosis in malignant lymphocytes, which increased phosphorylated cyclin-dependent kinase 1 (Cdk1) on Thr(161) and promoted the dysregulated activation of Cdk1. The ATO/PTX combination also significantly enhanced the activation of spindle checkpoint by inducing the formation of the inhibitory checkpoint complex BubR1/Cdc20. Our study provided the first in vitro demonstration that low concentrations of ATO and PTX synergistically induce mitotic arrest in malignant lymphocytes.


Subject(s)
Antineoplastic Agents/pharmacology , Arsenicals/pharmacology , Mitosis/drug effects , Oxides/pharmacology , Paclitaxel/pharmacology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Tubulin Modulators/pharmacology , Apoptosis , Arsenic Trioxide , CDC2 Protein Kinase/metabolism , Cells, Cultured , Drug Synergism , Humans , Jurkat Cells
17.
Cell Cycle ; 8(16): 2643-9, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19625775

ABSTRACT

KIF18A, a molecular motor, is an essential component in the regulation of orderly chromosome congression by attenuation of the kinetochore oscillation amplitude at the midzone during mitosis in vertebrate cells. Here we report that KIF18A depletion resulted in mitotic arrest which was accompanied by the presence of unaligned chromosomes in HeLa cells. This resembles the phenotype induced by an impaired function of CENP-E, also a mitotic kinesin essential for the formation of the mitotic spindles. Our further analysis showed that KIF18A depletion caused specific downregulation of CENP-E. Downregulation of CENP-E as the result of KIF18A silencing was not due to reduced transcription but primarily due to the enhanced protein degradation. Co-immunoprecipitation revealed that KIF18A physically interacted with CENP-E and BubR1 during mitosis. Ectopic expression of the wild-type tail domain of CENP-E, but not a corresponding mutant, significantly suppressed chromosome congression defects in mitotic cells. Together, our studies strongly suggest that chromosome congression defects as the result of KIF18A depletion is at least in part mediated through destabilizing kinetochore CENP-E.


Subject(s)
Chromosomal Proteins, Non-Histone/physiology , Kinesins/physiology , Mitosis/physiology , Blotting, Western , Cell Line , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Flow Cytometry , Humans , Immunoprecipitation , Kinesins/genetics , Kinesins/metabolism , Microscopy, Fluorescence , Mitosis/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/physiology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
18.
Journal of Experimental Hematology ; (6): 1302-1305, 2010.
Article in Zh | WPRIM | ID: wpr-332372

ABSTRACT

This study was purposed to evaluate the sensitivity and specificity of G/(1, 3-β-D glucan)/GM (galactomannan) combined detection for early diagnosis of invasive fungal disease (IFD), determine the GM positive cut-off value, and investigate the change of diagnostic level before and after G/GM test and the relation of GM value with therapeutic effect. The ELISA with double antibody sandwich was used to detect the serum levels of G and G/M. The results showed that according to determined GM positive cut-off value, the sensitivity, specificity, positive and negative predictive value of G/GM combined detection were 100%, 65.7%, 67.6% and 100%, respectively. The case number of the clinical diagnosis of IFD increased from 1 to 24 cases, the diagnosis of 12 non-infective patients was changed as suspected IFD with the help of G/GM combined detection; the GM cut-off value decreased in patients whose GM cut-off value was higher before therapy and antifungal therapy was effective, while the GM cut-off value increased in patients no-responded to therapy. It is concluded that G/GM combined detection can increased the sensitivity of the diagnosis and reduce false negative rate in the early diagnosis of IFD. The dynamically monitoring G/GM cut-off value can be used as evaluation indicator of therapeutic efficacy.


Subject(s)
Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Young Adult , Antigens, Fungal , Early Diagnosis , Hematologic Neoplasms , Microbiology , Hematopoietic Stem Cell Transplantation , Mycoses , Diagnosis , Allergy and Immunology , Sensitivity and Specificity
19.
Article in Zh | WPRIM | ID: wpr-243338

ABSTRACT

This study was aimed to investigate the function defect of partial homing receptor on cord blood hematopoietic stem cells (CBHSC) and explore efficacy and feasibility of intervention in vitro. The expression and activity of active groups in P, E-selectin ligands on CD34+ cells from cord blood, bone marrow and peripheral blood were detected by flow cytometry; meanwhile the expression of active groups in selectin ligands on CD34+ cells treated by fucosyl transferase in vitro was determined by flow cytometry. The results indicated that the expression levels of CD26 on the surface of stem/progenitor cells (CD34+) from cord blood, bone marrow and peripheral blood were (7.62+/-0.63)%, (6.35+/-0.89)% and (6.18+/-0.91)% (p>0.05) respectively. And the activities of CD26 of the three sources of stem cells were 67.15 U/1000 cells (1 U=1 pmol/min), 26.85 U/1000 cells and 20.95 U/1000 cells respectively, in which the activity of CD26 on surface of CD34+ from cord blood was significantly higher than that from other both sources (p<0.01). The expression levels of P-selectin ligand on the stem/progenitor cells three kinds were (83.46+/-6.33)%, (15.65+/-0.89)% and (80.17+/-6.85)%, and the expression levels of E-selectin ligand on stem/progenitor cells of three kinds were (25.31+/-1.03)%, (26.34+/-0.89)% and (29.79+/-1.78)% respectively. The expression of E-selectin ligand on the surface of cord blood stem/progenitor cell CD34+ increased from (25.31+/-1.03)% to (63.23+/-1.08)% after glycosylation engineering. It is concluded that there is no significant difference of the expression of CD26 between the three sources of stem/progenitor cells, but the activity of CD26 in cord blood was obviously higher than that in bone marrow and peripheral blood. The expression of P-selectin ligand on bone marrow stem/progenitor cell was lower than that on stem cells of cord blood and peripheral blood. Glycosylation engineering can promote and elevate the expression of E-selectin ligand on the surface of CD34+ cells from cord blood.


Subject(s)
Humans , Antigens, CD34 , Metabolism , Bone Marrow Cells , Cell Biology , Metabolism , Cells, Cultured , Dipeptidyl Peptidase 4 , Metabolism , Fetal Blood , Cell Biology , Hematopoietic Stem Cells , Cell Biology , Metabolism , Receptors, Fibroblast Growth Factor , Metabolism , Sialoglycoproteins , Metabolism , Stem Cells , Cell Biology , Metabolism
SELECTION OF CITATIONS
SEARCH DETAIL