Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Publication year range
1.
Exp Hematol Oncol ; 11(1): 36, 2022 Jun 07.
Article in English | MEDLINE | ID: mdl-35672796

ABSTRACT

BACKGROUND: Tyrosine kinase inhibitors have achieved quite spectacular advances in the treatment of chronic myeloid leukemia (CML), but disease progression and drug resistance that related to the T315I mutation, remain major obstacles. Dendritic cell-derived exosomes (Dex) induce NK cell immunity, but have yet to achieve satisfactory clinical efficacy. An approach to potentiate antitumor immunity by inducing both NK- and T-cell activation is urgently needed. Retinoic acid early inducible-1γ (RAE-1γ), a major ligand of natural killer group 2 member D (NKG2D), plays an important role in NK-cell and T-lymphocyte responses. We generated RAE-1γ enriched CML-specific Dex (CML-RAE-1γ-Dex) from dendritic cells (DCs) pulsed with lysates of RAE-1γ-expressing CML cells or T315I-mutant CML cells, aiming to simultaneously activate NK cells and T lymphocytes. METHODS: We generated novel CML-RAE-1γ-Dex vaccines, which expressed RAE-1γ, and were loaded with CML tumor cell lysates. NK cells or T lymphocytes were coincubated with CML-RAE-1γ-Dex vaccines. Flow cytometry was performed to evaluate the activation and proliferation of these immune cells. Cytokine production and cytotoxicity toward CML cells with or without the T315I mutation were detected by ELISPOT, ELISA and LDH assays. CML models induced by BCR-ABL or BCR-ABLT315I were used to determine the immunological function of Dex in vivo. RESULTS: Herein, CML-RAE-1γ-Dex were prepared. CML-RAE-1γ-Dex effectively enhanced the proliferation and effector functions of NK cells, CD4+ T cells and CD8+ T cells, which in turn produced strong anti-CML efficacy in vitro. Moreover, CML-RAE-1γ-Dex-based immunotherapy inhibited leukemogenesis and generated durable immunological memory in CML mouse models. Similar immune responses were also observed with imatinib-resistant CML cells carrying the T315I mutation. CONCLUSIONS: This approach based on CML-RAE-1γ-Dex vaccines may be a promising strategy for CML treatment, especially for cases with the T315I mutation.

2.
Cancer Lett ; 482: 44-55, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32278814

ABSTRACT

Although targeted therapy using tyrosine kinase inhibitors (TKIs) has made remarkable progress in treating chronic myeloid leukemia (CML), this disease remains largely incurable, warranting further investigation of new therapeutic strategies. BCR-ABL is a highly specific tumor antigen in CML and provides an attractive opportunity for vaccination therapy. Exogenous antigens must be presented on MHC class I molecules-via a process termed cross-presentation-to activate specific cytotoxic T lymphocyte response. The relative efficiency of cross-presentation is determined in part by the ability of dendritic cells (DCs) to internalize and present antigens. Here, we present a novel tool that uses cytoplasmic transduction peptide (CTP) to facilitate the internalization of antigens by DCs in an endocytosis-independent manner, which greatly enhances the efficiency of antigen presentation, thereby inducing stronger cytotoxic activity to ensure the elimination of CML cells. The data suggest that CTP-fused CML-specific peptides can be applied in vaccination therapies for CML patients.


Subject(s)
Fusion Proteins, bcr-abl/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Peptides/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , Animals , Cell Line, Tumor , Cross-Priming/drug effects , Endocytosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Male , Mice , Peptides/chemical synthesis , Peptides/immunology , Treatment Outcome , Xenograft Model Antitumor Assays
3.
Onco Targets Ther ; 12: 10455-10467, 2019.
Article in English | MEDLINE | ID: mdl-31819526

ABSTRACT

BACKGROUND: Karyopherin-ß1 (KPNB1) belongs to the karyopherin superfamily, which functions as shuttling proteins from the cytoplasm to nuclear. A high level of KPNB1 has been reported in various cancers which promotes cell proliferation and inhibits apoptosis. However, the role of KPNB1 in chronic myeloid leukemia (CML) remains uncertain. METHODS: Expression level of KPNB1 in CML patient samples and cell lines was analyzed by Western blotting. The proliferation assays and colony formation assay were used to study the CML cell proliferation when KPNB1 knockdown in vitro. Next, Western blotting was used to evaluate the effects of KPNB1 on E2F1 and other cell cycle regulators. Then, the location of E2F1 was detected by immunofluorescence. Finally, flow cytometry was used to detect the effect of KPNB1 inhibitor importazole (IPZ) on CML cells. RESULTS: In this study, we firstly showed that KPNB1 is over-expressed in CML cells. Targeting KPNB1 with small interfering RNA (siRNA) and IPZ reduced proliferation and induced apoptosis of CML cells. The underlying mechanisms were also investigated that E2F1 nuclear transport was blocked after inhibiting KPNB1 with siRNA, suggesting KPNB1 over-expression mediates the excessive nuclear transport of E2F1 in CML cells. Moreover, the expression of the E2F1 targeted molecule such as c-Myc and KPNA2 was markedly reduced. The IPZ arrested CML cells at G2/M phase and induced cell apoptosis. CONCLUSION: In summary, our results clearly showed that KPNB1 is over-expressed in CML cells and mediates the translocation of E2F1 into the nucleus of CML cells, thereby inhibition of KPNB1 reduced proliferation and induced apoptosis of CML cells which provides new insights for targeted CML therapies.

4.
Oncol Rep ; 42(5): 1755-1766, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31432184

ABSTRACT

Diffuse large B­cell lymphoma (DLBCL), the most common type of non­Hodgkin's lymphoma, is classified into germinal center and activated B cell (ABC) subtypes. The myeloid differentiation primary response gene 88 (MYD88) L265P mutation is the most prevalent oncogenic mutation among patients with ABC DLBCL, the subtype that has the more inferior outcome. MYD88 oligomerization driven by the L265P mutant augments myddosome assembly and triggers the activation of nuclear factor kappa­light­chain­enhancer of activated B cells (NF­κB) signaling, highlighting MYD88 oligomerization as a potential therapeutic target for this malignancy. The synthetic peptidomimetic compound ST2825, which has previously been used as an anti­inflammatory agent, has been reported to inhibit MYD88 dimerization. In the present study, the anticancer effects of ST2825 were investigated using L265P­expressing ABC DLBCL cell lines. Using confocal microscopy and high­molecular­weight fraction experiments, it was revealed that L265P­associated myddosome assembly was disrupted by ST2825. The results also revealed that disrupting myddosome assembly promoted the death of ABC DLBCL cells harboring the L265P mutation, as well as downregulating survival signals, including the inhibition of NF­κB and the suppression of IL­10 and interferon­ß production. Further co­immunoprecipitation studies demonstrated that MYD88 bound to BTK in L265P­DLBCL cells, and that this binding was abrogated following ST2825 treatment. Furthermore, the combination of myddosome­assembly disruption and BTK or BCL­2 signaling inhibition led to synergistic ABC DLBCL cell death, and more robust inhibition of NF­κB activity or increased apoptosis, respectively. The results of the present study provide evidence that the synthetic peptidomimetic compound ST2825, which targets myddosome assembly, may serve as a pharmacological inhibitor. ST2825 has the potential for clinical use in patients with L265P DLBCL, and other B­cell neoplasms driven by activated MYD88 signaling.


Subject(s)
Heterocyclic Compounds, 2-Ring/pharmacology , Lymphoma, Large B-Cell, Diffuse/genetics , Myeloid Differentiation Factor 88/chemistry , Myeloid Differentiation Factor 88/metabolism , Spiro Compounds/pharmacology , Agammaglobulinaemia Tyrosine Kinase/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/metabolism , Mutation , Myeloid Differentiation Factor 88/genetics , NF-kappa B/metabolism , Protein Binding/drug effects , Protein Multimerization/drug effects , Signal Transduction/drug effects
5.
J Exp Clin Cancer Res ; 38(1): 224, 2019 May 28.
Article in English | MEDLINE | ID: mdl-31138265

ABSTRACT

BACKGROUND: The bcr-abl fusion gene encodes BCR-ABL oncoprotein and plays a crucial role in the leukemogenesis of chronic myeloid leukemia (CML). Current therapeutic methods have limited treatment effect on CML patients with drug resistance or disease relapse. Therefore, novel therapeutic strategy for CML is essential to be explored and the CRISPR RNA-guided FokI nucleases (RFNs) meet the merits of variable target sites and specificity of cleavage enabled its suitability for gene editing of CML. The RFNs provide us a new therapeutic direction to obliterate this disease. METHODS: Guide RNA (gRNA) expression plasmids were constructed by molecular cloning technique. The modification rate of RFNs on bcr-abl was detected via NotI restriction enzyme digestion and T7 endonuclease 1 (T7E1) assay. The expression of BCR-ABL and its downstream signaling molecules were determined by western blotting. The effects of RFNs on cell proliferation and apoptosis of CML cell lines and CML stem/progenitor cells were evaluated by CCK-8 assay and flow cytometry. In addition, murine xenograft model was adopted to evaluate the capacity of RFNs in attenuating the tumorigenic ability of bcr-abl. RESULTS: The RFNs efficiently disrupted bcr-abl and prematurely terminated its translation. The destruction of bcr-abl gene suppressed cell proliferation and induced cell apoptosis in CML lines and in CML stem/progenitor cells. Moreover, the RFNs significantly impaired the leukemogenic capacity of CML cells in xenograft model. CONCLUSION: These results illustrate that the RFNs can target to disrupt bcr-abl gene and may provide a new therapeutic option for CML patients affiliated by drug resistance or disease relapse.


Subject(s)
Deoxyribonucleases/metabolism , Fusion Proteins, bcr-abl/antagonists & inhibitors , Gene Editing/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Animals , Apoptosis , CRISPR-Cas Systems , Cell Line, Tumor , Cell Proliferation , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Signal Transduction , Xenograft Model Antitumor Assays
6.
J Exp Clin Cancer Res ; 37(1): 62, 2018 Mar 20.
Article in English | MEDLINE | ID: mdl-29554925

ABSTRACT

BACKGROUND: The bcr-abl fusion gene is the pathological origin of chronic myeloid leukemia (CML) and plays a critical role in the resistance of imatinib. Thus, bcr-abl disruption-based novel therapeutic strategy may warrant exploration. In our study, we were surprised to find that the characteristics of bcr-abl sequences met the design requirements of zinc finger nucleases (ZFNs). METHODS: We constructed the ZFNs targeting bcr-abl with high specificity through simple modular assembly approach. Western blotting was conducted to detect the expression of BCR-ABL and phosphorylation of its downstream STAT5, ERK and CRKL in CML cells. CCK8 assay, colony-forming assay and flow cytometry (FCM) were used to evaluate the effect of the ZFNs on the viablity and apoptosis of CML cells and CML CD34+ cells. Moreover, mice model was used to determine the ability of ZFNs in disrupting the leukemogenesis of bcr-abl in vivo. RESULTS: The ZFNs skillfully mediated 8-base NotI enzyme cutting site addition in bcr-abl gene of imatinib sensitive and resistant CML cells by homology-directed repair (HDR), which led to a stop codon and terminated the translation of BCR-ABL protein. As expected, the disruption of bcr-abl gene induced cell apoptosis and inhibited cell proliferation. Notably, we obtained similar result in CD34+ cells from CML patients. Moreover, the ZFNs significantly reduced the oncogenicity of CML cells in mice. CONCLUSION: These results reveal that the bcr-abl gene disruption based on ZFNs may provide a treatment choice for imatinib resistant or intolerant CML patients.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Drug Resistance, Neoplasm/genetics , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Imatinib Mesylate/pharmacology , Zinc Finger Nucleases/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Fusion Proteins, bcr-abl/chemistry , Gene Editing , Genetic Vectors , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Neoplastic Stem Cells , Recombinational DNA Repair , Sequence Analysis, DNA , Xenograft Model Antitumor Assays
7.
J Exp Clin Cancer Res ; 35(1): 134, 2016 09 06.
Article in English | MEDLINE | ID: mdl-27599610

ABSTRACT

BACKGROUND: Yes-associated protein (YAP), an essential component of Hippo pathway, was identified as an oncoprotein which participated in the progression of various malignancies. However, its role in chronic myeloid leukemia (CML) remains to be further clarified. METHODS: The expression of YAP in CML cells was determined by western blotting. Next, the effects of YAP knockdown and YAP inhibitor on CML cells were evaluated by MTT assay, flow cytometry (FCM) and Wright's staining. Moreover, K562 induced mice model was employed to further investigate the role of YAP in vivo. RESULTS: YAP was overexpressed in CML cells. Knockdown of YAP by si-RNA or inhibition the function of YAP using verteporfin (VP) not only inhibited the proliferation, induced the apoptosis of CML cells but also reduced the expression of YAP target genes c-myc and survivin. Additionally, VP enhanced the efficacy of imatinib (IM) in vitro and suppressed leukemogenesis in vivo. CONCLUSION: Our results indicate that YAP may play an important role in the proliferation and leukemogenesis of CML cells. Genetic or pharmacological inhibition of YAP provides a novel treatment strategy for CML.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Imatinib Mesylate/administration & dosage , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Phosphoproteins/antagonists & inhibitors , Porphyrins/administration & dosage , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , HL-60 Cells , Humans , Imatinib Mesylate/pharmacology , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Phosphoproteins/genetics , Phosphoproteins/metabolism , Porphyrins/pharmacology , Transcription Factors , Up-Regulation/drug effects , Verteporfin , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
8.
Nan Fang Yi Ke Da Xue Xue Bao ; 37(2): 226-231, 2016 Feb 20.
Article in Chinese | MEDLINE | ID: mdl-28219868

ABSTRACT

OBJECTIVE: To investigate the effects of Sinopodophyllum hexundrum on apoptosis in K562 cells. METHODS: K562 cells were treated with Sinopodophyllum hexundrum at different concentrations and for different lengths of time to determine the optimal conditions of SinoPodophyllum hexandrum treatment for K562 cells using CCK8 assay. The cell apoptotic rate was detected by flow cytometry, and the cell morphology and nuclear morphology of K562 cells were observed with Wright staining and DPAI staining, respectively. The protein expressions of BCR/ABL, p-BCR/ABL, STAT5, p-STAT5 and the apoptosis-related proteins PARP, caspase-3 and cleaved-caspase-3 were determined with Western blotting. RESULTS: The cell proliferation was inhibited in a concentration-and time-dependent manner by 1, 2, and 3 µg/mL Sinopodophyllum hexundrum. The treatment was optimal with a Sinopodophyllum hexundrum concentration of 2 µg/mL a treatment time of 48 h, and the cell apoptotic rate increased in a time-dependent manner and significantly increased at 48 h (P<0.001). The expression of apoptosis-related proteins PARP, caspase-3 and cleaved-caspase-3 were also activated in a time-dependent manner. The cells showed typical apoptotic changes after treatment with 2 µg/mL Sinopodophyllum hexundrum for 48 h with significantly reduced expressions of BCR/ABL, p-BCR/ABL, STAT5, AND p-STAT5. CONCLUSION: Sinopodophyllum hexundrum promotes K562 cell apoptosis possibly by inhibiting BCR/ABL-STAT5 survival signal pathways and activating the mitochondrion-associated apoptotic pathways.


Subject(s)
Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Berberidaceae , Caspase 3/metabolism , Cell Proliferation , Fusion Proteins, bcr-abl/metabolism , Humans , K562 Cells , Mitochondria/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...