Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Br J Haematol ; 2024 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-39169447

RESUMO

Recent evidence suggests that immune thrombocytopenia (ITP), a common bleeding disorder, is linked to an imbalance in macrophage polarization and impaired bone marrow mesenchymal stem cells (BMSCs). However, the relationship between macrophage polarization imbalance and functional defects in BMSCs, as well as the involvement of associated molecules in BMSCs' defects, is not well understood. This study aimed to investigate the regulatory effects of high mobility group protein 1 (HMGB1) on the physiological functions of BMSCs, specifically in relation to macrophage polarization imbalance. Patients with ITP showed dysregulation in monocyte/macrophage polarization and impaired BMSCs function. HMGB1 was found to have a negative impact on the ability of BMSCs to regulate the imbalance in macrophage polarization, especially when inflammatory factors are present. The MyD88-dependent pathway downstream of BMSCs was found to be significantly enhanced with HMGB1 treatment. Furthermore, treatment with toll-like receptor 4 (TLR4) inhibitors successfully restored the regulatory capacity of BMSCs in ameliorating macrophage polarization imbalance and effectively inhibited the activation of the MyD88-dependent pathway. Meanwhile, infusion of si-TLR4-BMSCs reversed HMGB1-induced platelet dysfunction and reduced over-polarization to M1-like macrophages in the ITP mouse model. Consequently, targeting the HMGB1-TLR4 pathway could be a potential approach to restore the immunoregulatory function of BMSCs.

2.
Cell Transplant ; 32: 9636897231213271, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38059278

RESUMO

Mesenchymal stem cells (MSCs) have become a promising therapeutic method. More safety data are needed to support clinical studies in more diseases. The aim of this study was to investigate the short- and long-term safety of human bone marrow-derived MSCs (hBMMSCs) in mice. In the present study, we injected control (saline infusion only), low (1.0 × 106/kg), medium (1.0 × 107/kg), and high (1.0 × 108/kg) concentrations of hBMMSCs into BALB/c mice. The safety of the treatment was evaluated by observing changes in the general condition, hematology, biochemical indices, pathology of vital organs, lymphocyte subsets, and immune factor levels on days 14 and 150. In the short-term toxicity test, no significant abnormalities were observed in the hematological and biochemical parameters between the groups injected with hBMMSCs, and no significant damage was observed in the major organs, such as the liver and lung. In addition, no significant differences were observed in the toxicity-related parameters among the groups in the long-term toxicity test. Our study also demonstrates that mice infused with different doses of hBMMSCs do not show abnormal immune responses in either short-term or long-term experiments. We confirmed that hBMMSCs are safe through a 150-day study, demonstrating that this is a safe and promising therapy and offering preliminary safety evidence to promote future clinical applications of hBMMSCs in different diseases.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Humanos , Camundongos , Animais , Células-Tronco Mesenquimais/fisiologia , Medula Óssea , Fígado , Transplante de Células-Tronco Mesenquimais/efeitos adversos , Transplante de Células-Tronco Mesenquimais/métodos , Células da Medula Óssea
3.
J Cancer ; 13(3): 1019-1030, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154467

RESUMO

N6-methyladenosine (m6A) is the most abundant internal modification in mammalian mRNA and recent studies have highlighted the importance of m6A levels in tumor development. In this study, we investigated the expression of methyltransferase-like 3 (METTL3) and 14 (METTL14), components of the RNA m6A methyltransferase complex, in samples from 89 patients with acute myeloid leukemia (AML), and followed the survival of 75 of these patients. Our results show that METTL3 and METTL14 are highly expressed in most of the patients with AML (except those with APL), and high levels of METTL3 and/or METTL14 correlated to shorter survival in the patients. In leukemia cell lines K562 and kasumi-1, both METTL3 and METTL14 promote cell proliferation and cell cycle, and the knockdown of METTL3 and METTL14 inhibits proliferation, and induces apoptosis and differentiation. Notably, the knockdown of METTL3 and METTL14 in K562 cell line leads to several changes in the expression of p53 signal pathway, including the upregulation of p53, cyclin dependent kinase inhibitor 1A (CDKN1A/p21), and downregulation of mdm2. Importantly, the m6A level of mdm2 mRNA was significant lower after knock-down of METTL3 and METTL14 examined by m6A-RIP and mdm2 qPCR assay, and the half-life of mdm2 under actinomycin-D treatment became shorter. Taken together, our study demonstrates that the lower m6A levels of mdm2 mRNA mediated by the knockdown of METTL3 and METTL14 could lead to the low stability of mdm2 mRNA transcripts and low expression of MDM2, in the end, activate p53 signal pathway. Both METTL3 and METTL14 play an oncogenic role in AML by targeting mdm2/p53 signal pathway.

4.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 29(5): 1631-1636, 2021 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-34627452

RESUMO

OBJECTIVE: To investigate the effect of high mobility group protein 1(HMGB1) on the proliferation and cytokine expression of human bone marrow mesenchymal stem cells (MSC). METHODS: Different concentrations of recombinant human HMGB1 protein (100, 200, 400, 800 and 1000 ng/ml) were incubated with MSC for 24, 48, 72 h and the proliferation of MSC were detected respectively by using the CCK-8 method and flow cytometry. The best concentrations of HMGB1 incubated with MSC was determined (200 ng/ml, 1000 ng/ml), and the flow cytomerty was used to determine the effect of HMGB1 on the proliferation of MSC. The mRNA expression levels of IL-10, TGF- ß1, TSG-6 and IFN-γ in MSC incubated with HMGB1 protein were detected by real-time quantitative PCR and ELISA. RESULTS: The result of MSC identification and flow cytometry showed that the CD105, CD73 and CD90 were expressed, but did not expression CD45, CD34, CD11b, CD19 and HLA-DR; CCK-8 showed that HMGB1 at the concentrations of 100 ng/ml, 200 ng/ml and 400 ng/ml could promote the proliferation of MSC incubated for 24, 48 and 72 h as compared with the control group (P<0.05), and the most effective concentration was 200 ng/ml; flow cytometry showed that the compared with the control group, HMGB1 200 ng/ml could induce MSC from G1 phase to S phase to promote the proliferation of MSC; QPCR showed that the mRNA expression of MSC cytokines IL-10, TGF-ß1, TSG-6 increased while IFN-γ decreased at the concentration of 200 ng/ml HMGB1 as compared with the control group. ELISA experiments showed that the HMGB1 200 ng/ml acting on MSC for 48 h could significantly promoted the secretion of IL-10, TGF-ß 1 and TSG-6(P<0.05), while IFN-γ showed no significant difference as compared with control group. CONCLUSION: Recombinant human HMGB1 can promote the proliferation and secretion of MSC in healthy people.


Assuntos
Proteína HMGB1 , Células-Tronco Mesenquimais , Células da Medula Óssea , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Humanos
5.
Biochem Pharmacol ; 188: 114538, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33831397

RESUMO

Acute myeloid leukemia (AML) with FLT3 internal tandem duplication (FLT3-ITD) has a dismal prognosis. FLT3 inhibitors have been developed to treat patients with FLT3-ITD AML; however, when used alone, their efficacy is insufficient. FLT3 inhibitors combined with chemotherapy may be a promising treatment for FLT3-ITD AML. Homoharringtonine (HHT) is a classical anti-leukaemia drug with high sensitivity to FLT3-ITD AML cells. Here, we showed that HHT synergizes with a selective next-generation FLT3 inhibitor, quizartinib, to inhibit cell growth/viability and induce cell-cycle arrest and apoptosis in FLT3-ITD AML cells in vitro, significantly inhibit acute myeloid leukemia progression in vivo, and substantially prolong survival of mice-bearing human FLT3-ITD AML. Mechanistically, HHT and quizartinib cooperatively inhibit FLT3-AKT and its downstream targets GSK3ß, c-Myc, and cyclin D1, cooperatively up-regulate the pro-apoptosis proteins Bim and Bax, and down-regulate the anti-apoptosis protein Mcl1. Most strikingly, HHT and quizartinib cooperatively reduce the numbers of side-population (SP) and aldehyde dehydrogenase (ALDH)-positive cells, which reportedly are rich in LSCs. In conclusion, HHT combined with quizartinib may be a promising treatment strategy for patients with FLT3-ITD AML.


Assuntos
Benzotiazóis/administração & dosagem , Mepesuccinato de Omacetaxina/administração & dosagem , Leucemia Mieloide Aguda/tratamento farmacológico , Compostos de Fenilureia/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Adolescente , Adulto , Idoso de 80 Anos ou mais , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos/métodos , Sinergismo Farmacológico , Feminino , Humanos , Leucemia Mieloide Aguda/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Tirosina Quinase 3 Semelhante a fms/metabolismo
6.
J Cancer Res Clin Oncol ; 147(1): 33-47, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32880751

RESUMO

PURPOSE: Acute myeloid leukemia (AML) is a heterogenous disease and the survival of AML patients is largely attributed to the improvement of supportive treatment. Wilms' tumor 1-associated protein (WTAP) is a nuclear protein functions in many physiological and pathological processes. Although its expression and function in many malignant diseases have been reported, its prognostic and epigenetic roles in AML are largely unknown. METHODS: Peripheral blood or bone marrow samples were collected from AML patients. The WTAP expression was detected by western blot. WTAP expression level and patients clinical features were analyzed using statistical methods. WTAP knockdown AML cells were constructed. The experiments on proliferation, tumorigenic ability, cell cycle, and apoptosis were performed. Transcriptome sequencing was performed and analyzed. M6A methylation level was measured and m6A-RIP was performed to quantify m6A methylation level of MYC mRNA. RNA stability assay was performed to measure the half-life of mRNA. RESULTS: WTAP was overexpressed in AML patients and was an independent poor-risk factor in AML (p = 0.0140). Moreover, we found that WTAP regulated proliferation, tumorigenesis, cell cycle, and differentiation of AML cells. Furthermore, WTAP made AML cells resistant to daunorubicin. In further investigations, m6A methylation level was downregulated when knocking down WTAP, and c-Myc was upregulated due to the decreased m6A methylation of MYC mRNA. CONCLUSION: High WTAP expression predicts poor prognosis in AML and WTAP plays an epigenetic role in AML.


Assuntos
Adenosina/análogos & derivados , Proteínas de Ciclo Celular/metabolismo , Metilação de DNA , Regulação Neoplásica da Expressão Gênica , Leucemia Mieloide Aguda/patologia , Proteínas Proto-Oncogênicas c-myc/genética , Fatores de Processamento de RNA/metabolismo , RNA Mensageiro/genética , Adenosina/química , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Estudos de Casos e Controles , Ciclo Celular , Proteínas de Ciclo Celular/genética , Diferenciação Celular , Proliferação de Células , Feminino , Seguimentos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Pessoa de Meia-Idade , Prognóstico , Proteínas Proto-Oncogênicas c-myc/química , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Processamento de RNA/genética , RNA Mensageiro/metabolismo , Taxa de Sobrevida , Células Tumorais Cultivadas , Adulto Jovem
7.
Expert Rev Hematol ; 13(3): 289-297, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31990602

RESUMO

Objective: Therapeutic advances based on risk stratification and implementation of excellent supportive care measures have significantly improved outcomes for childhood acute myeloid leukemia (AML) over the past 30 years. However, approximately half of all childhood AML cases relapse. Therefore, precise risk stratification is needed for predicting relapse potential.Methods: RNA-seq data of TARGET-AML and corresponding clinical information of pediatric and adolescent AML cases were downloaded from The Cancer Genome Atlas. Clinical information of 156 patients with gene expression data was extracted. The effects of PAX7 expression profiles on overall survival (OS) and event-free survival (EFS) were analyzed.Results: Positive expression of PAX7 indicated shorter OS and EFS, especially in patients older than 14 years. Furthermore, positive PAX7 expression also predicted shorter OS and EFS in intermediate- and low-risk group patients, compared to patients with negative PAX7 expression. In addition, patients who have received allogenic hematopoietic stem cell transplantation (allo-HSCT) in the first complete remission had better outcome than those who did not receive HSCT.Conclusions: Positive PAX7 expression in pediatric and adolescent AML patients indicates a poor outcome. Hence, the detection of PAX7 expression profiles is helpful for further stratification of intermediate- and low-risk groups.


Assuntos
Bases de Dados de Ácidos Nucleicos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/mortalidade , Proteínas de Neoplasias/biossíntese , Fator de Transcrição PAX7/biossíntese , Adolescente , Criança , Pré-Escolar , Intervalo Livre de Doença , Feminino , Regulação Leucêmica da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/genética , Masculino , Proteínas de Neoplasias/genética , Fator de Transcrição PAX7/genética , Taxa de Sobrevida
8.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 50(3): 420-424, 2019 May.
Artigo em Chinês | MEDLINE | ID: mdl-31631611

RESUMO

OBJECTIVE: To investigate the expression of cystic fibrosis transmembrane conductance regulator (CFTR) protein in patients with acute leukemia and its relationship to clinical features and prognosis of acute leukemia. METHODS: A total of115 patients with acute leukemia were enrolled in the experimental group and 20 healthy individuals were used as control. Peripheral blood or bone marrow samples were collected, and mononuclear cells were isolated. The expression of CFTR protein was detected by Western blot. The relationships of CFTR protein expression to clinical features and prognosis was analyzed. RESULTS: The expression of CFTR protein was not detected in peripheral blood mononuclear cells of normal control, while it was positive in more than half of acute leukemias including acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL), but negative in the patients with acute promyelocytic leukemia (M3). In the patients with AML, there was no difference in peripheral white blood cells (WBC), peripheral blast cells, platelet and hemoglobin (HGB) between CFTR-positive and CFTR-negative patients. There was no relationship between the expression of CFTR protein and gene mutations such as NPM1, CEBPA, FLT3-ITD, and C-Kit. Complete remission (CR) rate after two course in CFTR-negative patients was slightly higher than that in positive patients. The survival time of CFTR-negative patients was little longer than that of positive patients, but the difference was not statistically significant. CONCLUSIONS: The expression of CFTR protein seems not associated with clinical features, treatment response and prognosis in the patients with acute leukemia.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/genética , Leucemia Mieloide Aguda/genética , Humanos , Leucemia Mieloide Aguda/diagnóstico , Leucócitos Mononucleares , Mutação , Nucleofosmina , Prognóstico
9.
PLoS One ; 12(9): e0185053, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28953917

RESUMO

BACKGROUND: The recent application of gene-sequencing technology has identified many new somatic mutations in patients with myelodysplastic syndromes (MDS). Among them, serine and arginine rich splicing factor 2 (SRSF2) mutations belonging to the RNA splicing pathway were of interest. Many studies have already reported the potential prognostic value of SRSF2 mutations in MDS patients, with controversial results. Therefore, a meta-analysis was performed to investigate their prognostic impact on MDS. METHODS: Databases, including PubMed, Embase and the Cochrane Library, were searched for relevant studies published up to 14 October 2016. Overall survival (OS) was selected as the primary endpoint, and acute myeloid leukemia (AML) transformation was the secondary endpoint. We extracted the corresponding hazard ratios (HRs) and their 95% confidence intervals (CIs) for OS and AML transformation from multivariate Cox proportional hazards models. The combined HRs with their 95% CIs were calculated using fixed or random effect models. RESULTS: A total of 10 cohort studies, covering 1864 patients with de novo MDS and 294 patients with SRSF2 mutations, were included in the final meta-analysis. Our results indicated that SRSF2 mutations had an adverse prognostic impact on OS (p<0.0001) and AML transformation (p = 0.0005) in the total population. Among the MDS patients with low or intermediate-1 risk defined according to the International Prognostic Scoring System (IPSS), SRSF2 mutations predicted a shorter OS (p = 0.009) and were more likely to transform to AML (p = 0.007). CONCLUSIONS: This meta-analysis indicates an independent, adverse prognostic impact of SRSF2 mutations on OS and AML transformation in patients with de novo MDS. This also applies to the subgroup of low- or intermediate-1-IPSS risk MDS. The identification of mutations in SRSF2 can improve current risk stratification and help make treatment decisions.


Assuntos
Mutação , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/genética , Fatores de Processamento de Serina-Arginina/genética , Humanos , Prognóstico
10.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 48(3): 389-393, 2017 May.
Artigo em Chinês | MEDLINE | ID: mdl-28616912

RESUMO

OBJECTIVES: To explore the possible roles of glucose transport 5 (Glut5) in imatinib resistance in the Ph+ acute lymphoblastic leukemia cell (Ph+ ALL). METHODS: The gene chip technique was used to detect different gene expression between Ph+ ALL cell line SUP-B15/R (imatinib resistant cell line) and SUP-B15/S (imatinib sensitive cell line), the gene of solute carrier family 2 member 5 (SLC2A5) and its coded protein Glut5 were screened out and were reconfirmed by qPCR and Western blot assay. The imatinib half maximal inhibitory concentration (IC50) to SUP-B15/S cells with or without fructose treatment was further detected by MTT assay, simultaneously signal pathway gene was detected by qPCR assay. RESULTS: Metabolism related gene SLC2A5 was screened out with gene chip technique and the Western blot assay and qPCR confirmed the high expression of SLC2A5 gene and its coded protein Glut5 in SUP-B15/R cells. IC50 values of imatinib to SUP-B15/S cells after treatment with 25 µmol/L fructose were increased from (44.50±2.38) µmol/L to (64.71±1.69) µmol/L, in the meanwhile, PI3K and AKT mRNA level also increased in fructose treated SUP-B15/S cells compared to the control. CONCLUSIONS: High expression of SLC2A5 and Glut5 protein in SUP-B15/R cells leads to increased fructose absorption, and further activates PI3K/AKT pathway which cause the SUP-B15 cell resistance to imatinib.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Transportador de Glucose Tipo 5/metabolismo , Mesilato de Imatinib/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Linhagem Celular Tumoral , Humanos , Fosfatidilinositol 3-Quinases/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo
11.
Oncotarget ; 8(15): 24437-24448, 2017 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-28445932

RESUMO

Cystic fibrosis transmembrane conductance regulator (CFTR) is classified as an anion channel transporter of Cl- and HCO3-. Through interactions with its PDZ domain, CFTR is capable of regulating other proteins, such as protein phosphatase 2A (PP2A). The aberrant expression and mutation of CFTR have been observed in several tumor, but not in philadelphia chromosome-positive(Ph+) acute leukemia, including Ph+ B cell acute lymphoblastic leukemia(Ph+ B-ALL) and chronic myelogenous leukemia blast crisis phases (CML-BC). In this study, we demonstrated the mean expression level of CFTR in Ph+ acute leukemia cells was markedly higher than that in Ph- B-ALL and CML-chronic phase cells. CFTRinh-172, a classic CFTR inhibitor, down-regulated the expression of CFTR, p-BCR-ABL and classical Wnt/ß-catenin signaling in Ph+ acute leukemia cells, while imatinib had no effect on CFTR. Importantly, reduced efficacy of CFTRinh-172 was closely associated with elevated PP2A phosphatase activity. Furthermore, we confirmed an interaction between CFTR and the PP2AA subunit in K562 cells. In addition, we demonstrated CFTR and PP2AA interact in the cytosol, resulting in PP2A complex inactivation and increased degradation of PP2A substrates via the lysosomal/proteasome pathway. In conclusion, our results showed CFTR was highly expressed in Ph+ acute leukemia, which protected and maintained the continuous activation of BCR-ABL and the canonical Wnt/ß-catenin signaling pathway by decreasing PP2A phosphatase activity. According to this working model of the CFTR-PP2A-BCR-ABL axis, targeting the CFTR protein will activate PP2A and may offer a new treatment strategy for Ph+ acute leukemia, especially for patients exhibiting high levels of CFTR expression.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/biossíntese , Leucemia Mieloide Aguda/metabolismo , Proteína Fosfatase 2/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Proteínas de Fusão bcr-abl/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Cromossomo Filadélfia , Transdução de Sinais
12.
Leuk Res ; 57: 9-19, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28235656

RESUMO

The aberrant expression or mutation of CFTR has been shown to be involved in several tumors, but how mutations or dysfunction of CFTR may increase the risk of malignancies in various tissues remains unclear. Here, we report the interaction between CFTR and HDAC2, and its involvement in the development of Ph+ leukemia. We first detected the physical interaction and co-localization of CFTR and HDAC2 in Ph+ leukemia cell lines. And treatment with CFTRinh-172, a CFTR inhibitor, reduced the expression of HDAC2 protein in K562 and SUP-B15 cells, which could be partially recovered by MG132, a proteasome inhibitor. Additionally, high expression levels of HDAC2 protein were observed in CFTR cDNA transfected HEK-293 and Ba/F3 cells. Next, we found that HDAC2 bound in the promoter region of the PTEN gene, and treatment with HDAC2 inhibitor or CFTRinh-172 resulted in an increase in PTEN mRNA and protein levels and a decrease in PDK1 and mTOR (downstream signaling of PTEN) activity. Finally, the MTT assay revealed that CFTRinh-172 could strengthen the anti-proliferation effect of HDAC2 inhibitor on Ph+ leukemia cells. Altogether, this study provides strong evidence that high-expression CFTR plays an important role in the development of Ph+ leukemia through the HDAC2-mediated PTEN pathway.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Histona Desacetilase 2/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Linhagem Celular Tumoral , Células HEK293 , Humanos , Células K562 , Redes e Vias Metabólicas , PTEN Fosfo-Hidrolase/genética , Regiões Promotoras Genéticas , Transfecção
13.
Leuk Res ; 42: 59-67, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26818574

RESUMO

Niemann-Pick disease type C1 (NPC1) is involved in cholesterol trafficking and may normally function as a transmembrane efflux pump. Previous studies showed that its dysfunction can lead to cholesterol and daunorubicin accumulation in the cytoplasmic endosomal/lysosomal system, lead to Niemann-Pick disease and resistance to anticancer drugs. In the present study, NPC1 was shown by microarray analysis to be more highly expressed in the Ph+ acute lymphoblastic leukemia cell line SUP-B15/RI, an imatinib-resistant variant of SUP-B15/S cells without bcr-abl gene mutation established in our lab. Further investigation revealed a defect in the functional capacity of the NPC1 protein demonstrated by filipin staining accompanied by a lower intracellular imatinib mesylate(IM) concentration by high-performance liquid chromatography in SUP-B15/RI compared with SUP-B15/S cells. Furthermore, U18666A, an inhibitor of NPC1 function, was used to block cholesterol trafficking to imitate the NPC1 defect in SUP-B15/S cells, leading to higher NPC1 expression, stronger filipin fluorescence, lower intracellular IM concentrations and greater resistance against IM. Samples from non-mutated relapsed Ph+ ALL patients also showed higher NPC1 expression compared with IM-sensitive patients. Our experiment may reveal a new mechanism of IM resistance in Ph+ ALL.


Assuntos
Proteínas de Transporte/genética , Resistencia a Medicamentos Antineoplásicos/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Glicoproteínas de Membrana/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Antineoplásicos/farmacologia , Western Blotting , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Imunofluorescência , Humanos , Mesilato de Imatinib/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Proteína C1 de Niemann-Pick , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase
14.
PLoS One ; 10(8): e0136746, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26317515

RESUMO

The eukaryotic translation initiation factor 4E (eIF4E), which is the main composition factor of eIF4F translation initiation complex, influences the growth of tumor through modulating cap-dependent protein translation. Previous studies reported that ribavirin could suppress eIF4E-controlled translation and reduce the synthesis of onco-proteins. Here, we investigated the anti-leukemic effects of ribavirin alone or in combination with tyrosine kinase inhibitor imatinib in Philadelphia chromosome positive (Ph+) leukemia cell lines SUP-B15 (Ph+ acute lymphoblastic leukemia cell line, Ph+ ALL) and K562 (chronic myelogenous leukemia cell line, CML). Our results showed that ribavirin had anti-proliferation effect; it down-regulated the phosphorylation levels of Akt, mTOR, 4EBP1, and eIF4E proteins in the mTOR/eIF4E signaling pathway, and MEK, ERK, Mnk1 and eIF4E proteins in ERK/Mnk1/eIF4E signaling pathway; reduced the expression of Mcl-1 (a translation substrates of eIF4F translation initiation complex) at protein synthesis level not mRNA transcriptional level; and induced cell apoptosis in both SUP-B15 and K562. 7-Methyl-guanosine cap affinity assay further demonstrated that ribavirin remarkably increased the eIF4E binding to 4EBP1 and decreased the combination of eIF4E with eIF4G, consequently resulting in a major inhibition of eIF4F complex assembly. The combination of ribavirin with imatinib enhanced antileukemic effects mentioned above, indicating that two drugs have synergistic anti-leukemic effect. Consistent with the cell lines, similar results were observed in Ph+ acute lymphoblastic primary leukemic blasts; however, the anti-proliferative role of ribavirin in other types of acute primary leukemic blasts was not obvious, which indicated that the anti-leukemic effect of ribavirin was different in cell lineages.


Assuntos
Antineoplásicos/farmacologia , Mesilato de Imatinib/farmacologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Ribavirina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Fator de Iniciação 4E em Eucariotos/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Cromossomo Filadélfia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Serina-Treonina Quinases TOR/metabolismo
15.
Anticancer Drugs ; 26(9): 913-22, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26186064

RESUMO

In recent years, there have been considerable research advances on the antileukemic mechanisms of the antidiabetic drug metformin. Our current studies have shown that metformin suppresses cell viability, induces apoptosis, and downregulates the mTORC1 signaling pathway both in the Ph+ALL cell line and primary blasts from Ph+ ALL patients, as well as the CML cell lines K562 (imatinib-sensitive) and K562R (imatinib-resistance). We have also shown that metformin activates the ERK pathway in Ph+ALL cells, SUP-B15, a side effect that can be overcome by U0126 (MEK1/2 inhibitor) or imatinib. Moreover, this activation of ERK signaling in SUP-B15 induces autophagy. Inhibition of the autophagic process by 3-MA, promoting the death of these cells, suggests that autophagy may be a cytoprotective factor in cell survival after metformin treatment. Finally, metformin is shown to potentiate the anticancer efficacy of imatinib in Ph+ALL and CML cells, resensitizing the CML imatinib-resistance cells to imatinib. Overall, our data suggest that metformin represents a promising and attractive agent for Ph+ALL or CML therapy.


Assuntos
Antineoplásicos/farmacologia , Leucemia Linfocítica Crônica de Células B/patologia , Metformina/farmacologia , Cromossomo Filadélfia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Autofagia/efeitos dos fármacos , Benzamidas/farmacologia , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Humanos , Mesilato de Imatinib , Leucemia Linfocítica Crônica de Células B/genética , Sistema de Sinalização das MAP Quinases , Piperazinas/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Pirimidinas/farmacologia , Células Tumorais Cultivadas
16.
Anticancer Drugs ; 26(4): 410-21, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25535978

RESUMO

PP242 is a novel dual mammalian target of rapamycin (mTOR) inhibitor that simultaneously inhibits mTORC1 and mTORC2, and its antileukemia effect has been sufficiently investigated here. The human acute leukemia cell lines and primary blasts were treated with PP242 alone or in combination with daunorubicin (DNR). Cell proliferation was examined using an MTT assay. The phosphorylation expression of the Akt/mTORC1/eIF4E signaling pathway was assessed by western blot analysis. The assembly of the eIF4F translation initiation complex was examined using a 7-methyl-guanosine cap affinity assay. PP242 significantly induced cytotoxicity in human acute leukemia cells, especially in combination with DNR. The phosphorylation levels of eIF4E (p-eIF4E) at Ser209 influence the antileukemia roles of PP242. As expected, the antiproliferative effects of PP242 on leukemia cells with low p-eIF4E expression, such as the acute promyelocytic leukemia NB4 cell line and AML-M3 primary blasts, were poor. Surprisingly, the effects of PP242 in leukemia cells with high p-eIF4E expression, such as the acute myelomonocytic leukemia THP-1 cell line and M4-M5 primary blasts, were also weak. In contrast, PP242 exerted a significant antiproliferative effect in the Ph+ acute lymphoblastic leukemia SUP-B15 cell line and the mantle cell lymphoma JEKO-1 cell line, which had intermediate p-eIF4E levels. PP242 inhibited the translation of the antiapoptotic protein Mcl-1 by downregulating the Akt/mTORC1/eIF4E signaling pathway. More importantly, DNR activated the Akt/mTORC1/eIF4E signaling pathway, whereas PP242 effectively eliminated this deleterious side effect of DNR and synergistically enhanced the anticancer ability of DNR treatment. PP242, especially in combination with DNR, exerts significant antileukemia effects.


Assuntos
Antineoplásicos/farmacologia , Daunorrubicina/farmacologia , Indóis/farmacologia , Leucemia/patologia , Purinas/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Fator de Iniciação 4E em Eucariotos/metabolismo , Humanos , Leucemia Mielomonocítica Aguda/patologia , Leucemia Promielocítica Aguda/patologia , Linfoma de Célula do Manto/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA