Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Ann Hematol ; 103(7): 2355-2364, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38710877

RESUMO

Acute myeloid leukemia (AML) shows multiple chromosomal translocations & point mutations which can be used to refine risk-adapted therapy in AML patients. Ecotropic viral integration site-1 (EVI-1) & myocyte enhancer factor 2 C gene (MEF2C) are key regulatory transcription factors in hematopoiesis and leukemogenesis & both drive immune escape. This prospective study involved 80 adult de novo AML patients recruited from Oncology Center, Mansoura University, between March 2019 and July 2021. The MEF2C and EVI1 expression were measured using a Taqman probe-based qPCR assay. The results revealed that EVI1 and MEF2C expression were significantly elevated in AML patients as compared to control subjects (p = 0.001. 0.007 respectively). Aberrant expressions of EVI1 and MEF2C showed a significant negative correlation with hemoglobin levels (p = 0.034, 0.025 respectively), & bone marrow blasts (p = 0.007, 0.002 respectively). 11q23 translocation was significantly associated with EVI1 and MEF2C (p = 0.004 and 0.02 respectively). Also, t (9;22) was significantly associated with EVI1 and MEF2C (p = 0.01 and 0.03 respectively), higher expression of EVI1 and MEF2C were significantly associated with inferior outcome after induction therapy (p = 0.001 and 0.018 respectively) and shorter overall survival (p = 0.001, 0.014 respectively). In conclusion, EVI1 & MEF2C were significantly expressed in AML cases. EVI1 & MEF2C overexpression were significantly associated with 11q23 rearrangements and t (9;22) and were indicators for poor outcome in adult AML patients; These results could be a step towards personalized therapy in those patients.


Assuntos
Leucemia Mieloide Aguda , Proteína do Locus do Complexo MDS1 e EVI1 , Fatores de Transcrição MEF2 , Translocação Genética , Humanos , Fatores de Transcrição MEF2/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Masculino , Proteína do Locus do Complexo MDS1 e EVI1/genética , Feminino , Adulto , Pessoa de Meia-Idade , Idoso , Cromossomos Humanos Par 11/genética , Estudos Prospectivos , Adulto Jovem , Rearranjo Gênico , Adolescente
2.
Rinsho Ketsueki ; 65(9): 954-960, 2024.
Artigo em Japonês | MEDLINE | ID: mdl-39358295

RESUMO

EVI1 is a zinc finger transcription factor encoded by the MECOM locus and is essential for the development and maintenance of hematopoietic stem cells. However, overexpression of EVI1 in various myeloid malignancies is associated with aggressive clinical behavior and poor outcome. The locus encodes multiple isoforms that are differentially acting and independently regulated. EVI1 interacts with a variety of transcription and epigenetic factors via different domains. It also regulates cell survival, differentiation, and proliferation through a variety of mechanisms, including transcriptional activation and repression, regulation of other transcription factors' activity, and chromatin remodeling. While the mechanism by which 3q26 translocation leads to high EVI1 expression through enhancer hijacking of genes active in myeloid development is now better understood, regulation of EVI1 expression in the absence of chromosomal translocations and in normal hematopoiesis remains unclear. Recent studies have provided insight into the regulatory mechanisms of EVI1 expression and action, which may lead to development of targeted therapies in the near future.


Assuntos
Neoplasias Hematológicas , Hematopoese , Proteína do Locus do Complexo MDS1 e EVI1 , Proteína do Locus do Complexo MDS1 e EVI1/genética , Proteína do Locus do Complexo MDS1 e EVI1/metabolismo , Humanos , Hematopoese/genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patologia , Animais , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Proto-Oncogenes/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica
3.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 45(9): 1061-1067, 2020.
Artigo em Inglês, Zh | MEDLINE | ID: mdl-33051419

RESUMO

OBJECTIVES: To evaluate the expression of myeloid ecotropic viral integration site 1 (Meis1) and vascular endothelial growth factor receptor 2 (VEGFR-2) in early-stage kidney cancers and the clinical significance. METHODS: The cancer tissues and the matched adjacent normal tissues in patients with kidney cancer, who received surgical treatment from April 2005 to September 2018 in the Haikou Hospital Affiliated to Xiangya School of Medicine, Central South University, were collected. The samples included 80 pairs of paraffin specimen, 15 pairs of fresh cancer and the matched adjacent normal tissues from these patients. Real-time PCR and immunohistochemical method were used to detect the expression levels of Meis1 and VEGFR-2 mRNA and protein in kidney tissues and adjacent normal tissues, and the correlation of clinical pathology parameters and the prognosis were analyzed in the patients. RESULTS: The expression levels of Meis1 and VEGFR-2 mRNA and protein in the renal carcinoma tissues were lower than those in the matched adjacent normal tissues (both P<0.01), and the expression levels of Meis1 were positively correlated with that of VEGFR-2 (r=0.681, P<0.01). The analysis of relevant clinical-pathological parameters in the patients showed that: the expression positive rate of Meis1 was significantly related with the pathological type of renal cancer (P<0.01), while the positive rate of Meis1 and VEGFR-2 expression was not related with the gender, age, T stage of patients (all P>0.05), but it was significantly related with the prognosis in the patients (P<0.05). Cox regression analysis showed that: Meis1 was an independent factor for the prognosis of patients (P<0.05). CONCLUSIONS: The mRNA and protein expression levels of Meis1 and VEGFR-2 in the early-stage kidney cancer tissues are significantly decreased compared with those in the adjacent normal tissues. Meis1 may be served as a tumor suppressor to affect the occurrence and development of kidney cancer. Therefore, Meis1 may be used as a biomarker to predict the prognosis of patients with kidney cancer.


Assuntos
Carcinoma de Células Renais , Neoplasias Renais , Proteína Meis1 , Receptor 2 de Fatores de Crescimento do Endotélio Vascular , Humanos , Neoplasias Renais/genética , Proteína Meis1/metabolismo , Prognóstico , Fator A de Crescimento do Endotélio Vascular , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
4.
J Neurooncol ; 145(2): 223-231, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31617054

RESUMO

PURPOSE: Ecotropic viral integration site-1 (EVI1) is a transcription factor that contributes to the unfavorable prognosis of leukemia, some epithelial cancers, and glial tumors. However, the biological function of EVI1 in glioblastoma multiforme (GBM) remains unclear. Based on microarray experiments, EVI1 has been reported to regulate epidermal growth factor receptor (EGFR) transcription. Signal transduction via EGFR plays an essential role in glioblastoma. Therefore, we performed this study to clarify the importance of EVI1 in GBM by focusing on the regulatory mechanism between EVI1 and EGFR transcription. METHODS: We performed immunohistochemical staining and analyzed the EVI1-expression in glioma tissue. To determine the relationship between EVI1 and EGFR, we induced siRNA-mediated knockdown of EVI1 in GBM cell lines. To investigate the region that was essential for the EVI1 regulation of EGFR expression, we conducted promoter reporter assays. We performed WST-8 assay to investigate whether EVI1 affected on the proliferation of GBM cells or not. RESULTS: It was observed that 22% of GBM tissues had over 33% of tumor cells expressing EVI1, whereas no lower-grade glioma tissue had over 33% by immunohistochemistry. In A172 and YKG1 cells, the expression levels of EGFR and EVI1 correlated. Analysis of the EGFR promoter region revealed that the EGFR promoter (from - 377 to - 266 bp) was essential for the EVI regulation of EGFR expression. We showed that EVI1 influenced the proliferation of A172 and YKG1 cells. CONCLUSION: This is the first study reporting the regulation of EGFR transcription by EVI1 in GBM cells.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteína do Locus do Complexo MDS1 e EVI1/metabolismo , Adulto , Idoso , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patologia , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/diagnóstico , Glioblastoma/patologia , Humanos , Proteína do Locus do Complexo MDS1 e EVI1/genética , Masculino , Pessoa de Meia-Idade , RNA Interferente Pequeno/administração & dosagem
5.
Mol Med Rep ; 28(6)2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37830162

RESUMO

Following the publication of this paper, it was drawn to the Editor's attention by a concerned reader that, for the Transwell cell migration and invasion assay experiments shown in Fig. 3 on p. 1650, there were several panels showing overlapping sections of data; moreover, certain of the data shown in this Figure were also strikingly similar to data appearing in different form in Fig. 4 in another article written by different authors at a different research institute [Liu J and Duan X: PA­MSHA induces apoptosis and suppresses metastasis by tumor associated macrophages in bladder cancer cells. Cancer Cell Int 17: 76, 2017].  Owing to the fact that the contentious data in the above article had already been published prior to its submission to Molecular Medicine Reports, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [Molecular Medicine Reports 19: 1645­1653, 2019; DOI: 10.3892/mmr.2018.9796].

6.
Int J Lab Hematol ; 45(2): 195-203, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36358022

RESUMO

INTRODUCTION: AML patients with KMT2A-MLLT3 and other 11q23 abnormalities belong to the intermediate and high-risk level groups, respectively. Whether the poor prognostic value of Ecotropic Viral Integration site-1 (EVI1) overexpression suits either the subtypes of KMT2A-MLLT3 or Non-KMT2A-MLLT3 AML patients (intermediate and high risk group) needs to be further investigated. METHODS: We retrospectively analyzed the clinical characteristics of 166 KMT2A-r and KMT2A-PTD AML patients. RESULTS: For the Non-KMT2A-MLLT3 group, patients in the EVI1-high subgroup had shorter OS and DFS and higher CIR than those in the EVI1-low subgroup (p = .027, p = .018, and p = .020, respectively). Additionally, both KMT2A-MLLT3 and Non-KMT2A-MLLT3 patients who received chemotherapy alone had poorer prognosis than patients who also received allogeneic hematopoietic stem cell transplant (allo-HSCT) regardless of their EVI1 expression level (all p < .001). For transplanted patients with KMT2A-MLLT3 or Non-KMT2A-MLLT3 rearrangement, the EVI1-high subgroup had worse prognosis than the EVI1-low subgroup (all p < .05). The 2-year CIR of the KMT2A-MLLT3 and Non-KMT2A-MLLT3 groups with high EVI1 expression was high (52% and 49.6%, respectively). However, for patients with low EVI1 expression, the 2-year CIR of transplanted patients with KMT2A-MLLT3 and Non-KMT2A-MLLT3 was relatively low. CONCLUSIONS: Our study showed that for the Non-KMT2A-MLLT3 group, the EVI1-high group had shorter OS and DFS than the EVI1-low group. High EVI1 expression showed an adverse effect in AML with KMT2A rearrangement in different risk stratification subtypes. For the EVI1-high patients with non-KMT2A-MLLT3 rearrangement, other novel regimens towards relapse should be taken into consideration.


Assuntos
Leucemia Mieloide Aguda , Proteína do Locus do Complexo MDS1 e EVI1 , Humanos , Rearranjo Gênico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Proteína do Locus do Complexo MDS1 e EVI1/genética , Proteína do Locus do Complexo MDS1 e EVI1/uso terapêutico , Prognóstico , Estudos Retrospectivos , Medição de Risco , Integração Viral , Transplante de Células-Tronco Hematopoéticas/métodos , Expressão Gênica
7.
Life Sci ; 255: 117822, 2020 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-32450174

RESUMO

AIM: Proliferation and migration of pulmonary artery smooth muscle cells (PASMCs) are regarded as the primary factors resulting in pulmonary arterial remodeling in pulmonary hypertension (PH). Myeloid ecotropic viral integration site 1 (MEIS1) has been positioned as a negative cardiomyocyte cell cycle regulator and regulates proliferation of multiple kinds of cancer cells. Whether MESI1 is involved in the proliferation and migration of PASMCs deserves to be identified. MAIN METHODS: Sprague Dawley rats were exposed to hypoxia condition (10% O2) for 4 weeks to induce PH and primary rat PASMCs were cultured in hypoxia condition (3% O2) for 48 h to induce proliferation and migration. Immunohistochemistry, immunofluorescence, reverse transcription PCR and Western blot analysis were performed to detect the expressions of target mRNAs and proteins. EDU, CCK8 and wound healing assays were conducted to measure the proliferation and migration of PASMCs. KEY FINDINGS: Hypoxia down-regulated the expression of MEIS1 (both mRNA and protein) in pulmonary arteries and PASMCs. Over-expression of MEIS1 inhibited the proliferation and migration of PASMCs afforded by hypoxia. In contrast, knockdown of MEIS1 under normoxia condition like hypoxia induced the proliferation and migration of PASMCs. MEIS1 mediated hypoxia-induced the proliferation and migration of PASMCs via METTL14/MEIS1/p21 signaling. SIGNIFICANCE: The present study revealed that MEIS1 regulated the proliferation and migration of PASMCs during hypoxia-induced PH. Thus, MEIS1 may be a potential target for PH therapy.


Assuntos
Hipertensão Pulmonar/fisiopatologia , Proteína Meis1/genética , Miócitos de Músculo Liso/citologia , Artéria Pulmonar/citologia , Animais , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Hipertensão Pulmonar/genética , Hipóxia , Masculino , Músculo Liso Vascular/citologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Remodelação Vascular/fisiologia
8.
Oncol Lett ; 16(2): 2701-2708, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30013666

RESUMO

Acute myeloid leukemia (AML) is a type of malignant tumor that is caused by malignant clone hematopoietic stem cells. The ecotropic viral integration site 1 (Evi1) is a zinc finger transcription factor, which is highly expressed in AML, and its expression level has been associated with poor prognosis of AML. Previous studies have indicated that Evi1 may regulate cell proliferation, differentiation and apoptosis by inhibiting the membrane-spanning-4-domains subfamily-A member-3 (MS4A3) gene in AML. The aim of the present study was to investigate the role of Evi1 in the progression of AML. The results revealed that Evi1 was overexpressed in leukemia cells compared with normal T lymphocytes. MicroRNAs (miR)-133 and -431 that target Evi1 were investigated, and it was observed that there was a low expression of miR-431 in AML. The transfection of miR-431 was able to decrease the promoter methylation levels of the Evi1 gene in AML cells. The transfection of miR-431 also suppressed the migration and invasion of AML cells. The present study revealed that the transfection of miR-431 mimic was able to downregulate MS4A3 expression in AML cells. Furthermore, the expression levels of transforming growth factor ß (TGFß) and epithelial-to-mesenchymal transition (EMT) markers fibronectin, α-smooth muscle actin, and vimentin were downregulated following the transfection of miR-431 in AML cells. The overexpression of MS4A3 was also able to suppress miR-431-mediated inhibition of the expression of TGFß and EMT markers in AML cells. The addition of TGFß inhibited the downregulation of EMT markers by transfection of miR-431 in AML cells. The transfection of miR-431 suppressed the migration and invasion of AML cells, which was also abolished by the addition of TGFß. In conclusion, the results of the present study indicated that Evi1 may be a potential molecular target of leukemia therapy via MS4A3-mediated TGFß/EMT signaling pathway.

9.
Onco Targets Ther ; 10: 2739-2744, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28603423

RESUMO

AIM: To explore the expression and clinical significance of ecotropic viral integration site-1 (EVI1) of lung squamous cell cancer (SCC). METHODS: The expression of EVI1 in SCC was detected by immunohistochemistry and the validation cohort was divided into EVI1 high-expression group and low-expression group according to the cutoff of immunohistochemical score. The correlations between EVI1 expression and the clinicopathological factors were analyzed by χ2 test. The relation between EVI1 expression and overall survival rate was evaluated by univariate analysis with Kaplan-Meier method. The independent prognostic factor was identified by multivariate analysis with Cox regression model. RESULTS: In this study, the EVI1 high-expression percentage was 32.32% (53/164). EVI1 high expression was significantly associated with a poorer overall 5-year survival rate of SCC (P=0.021). Moreover, EVI1 high expression was identified as an independent prognostic factor of SCC, predicting the unfavorable prognosis (P=0.013). CONCLUSION: High expression of EVI1 was significantly associated with a poorer prognosis and it was identified as an independent prognostic factor of SCC.

10.
Oncotarget ; 7(16): 21887-99, 2016 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-26967394

RESUMO

UNLABELLED: The involvement of the hepatitis B virus X (HBx) protein in epigenetic modifications during hepatocarcinogenesis has been previously characterized. Long noncoding RNAs (lncRNAs), a kind of epigenetic regulator molecules, have also been shown to play crucial roles in HBx-related hepatocellular carcinoma (HCC). In this study, we analyzed the key transcription factors of aberrantly expressed lncRNAs in the livers of HBx transgenic mice by bioinformatics prediction, and found that ecotropic viral integration site 1 (Evi1) was a potential main transcription regulator. Further investigation showed that EVI1 was positively correlated to HBx expression and was frequently up-regulated in HBV-related HCC tissues. The forced expression of HBx in liver cell lines resulted in a significant increase of the expression of EVI1. Furthermore, suppression of EVI1 expression decreased the proliferation of HCC cells overexpressing HBx in vitro and in vivo. CONCLUSION: Our findings suggest that EVI1 is frequently up-regulated and regulates a cluster of lncRNAs in HBV-related hepatocellular carcinoma (HCC). These findings highlight a novel mechanism for HBx-induced hepatocarcinogenesis through transcription factor EVI1 and its target lncRNAs, and provide a potential new approach to predict the functions of lncRNAs.


Assuntos
Carcinoma Hepatocelular/virologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Hepatite B/complicações , Neoplasias Hepáticas/virologia , Proteína do Locus do Complexo MDS1 e EVI1/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Carcinoma Hepatocelular/patologia , Proliferação de Células/genética , Transformação Celular Viral/fisiologia , Vírus da Hepatite B , Humanos , Neoplasias Hepáticas/patologia , Proteína do Locus do Complexo MDS1 e EVI1/genética , Camundongos , Camundongos Transgênicos
11.
Oncol Lett ; 10(3): 1694-1700, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26622734

RESUMO

Chronic neutrophilic leukemia (CNL) is a rare type of myeloproliferative neoplasm, characterized by sustained neutrophilia, splenomegaly, bone marrow granulocytic hyperplasia (without evidence of dysplasia) and an absence of the Philadelphia chromosome. Thus far, ~150 cases of CNL have been described in the literature; however, none have demonstrated overexpression of the ecotropic viral integration site-1 (EVI-1, also known as MECOM) gene. The present study describes a case that fulfilled the World Health Organization diagnostic criteria for CNL, and was associated with overexpression of EVI-1, as well as novel concurrent mutations of colony stimulating factor 3 receptor (CSF3R) and SET binding protein-1 (SETBP1). In addition, the current study briefly reviewed the relevant literature regarding novel genetic findings associated with the diagnosis and treatment of CNL. To the best of our knowledge, this is the first case report of CNL with associated EVI-1 overexpression, and concurrent CSF3R and SETBP1 mutations.

12.
Exp Ther Med ; 8(1): 85-90, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24944602

RESUMO

Acute leukemia is a malignant clonal hematopoietic stem cell disease. In the current study, the effects of arsenic trioxide (ATO) on the ecotropic viral integration site-1 (EVI-1) gene were investigated in the THP1 cell line. THP-1 cells were treated with different concentrations of ATO (0, 1, 3 and 5 µM) for 24, 48 or 72 h, then tested for cell viability by CCK-8 kit, cell morphology by cytospin smear, cell apoptosis by flow cytometry, EVI-1 mRNA expression by reverse transcription polymerase chain reaction (RT-PCR) and protein quantity by western blot. ATO treatment was shown to inhibit proliferation and induce apoptosis in THP1 cells in a dose- and time-dependent manner. ATO downregulated the mRNA and protein expression of EVI-1 in the THP1 cell line. In addition, ATO significantly decreased the expression of antiapoptotic proteins, B-cell lymphoma 2 (Bcl-2) and B cell lymphoma-extra large (Bcl-xL), but markedly increased the expression of proapoptotic proteins, including c-Jun N-terminal kinase (JNK), phosphorylated-JNK, Bax, full length caspase-3 and cleaved caspase-3. These results indicated that ATO inhibited the proliferation and induced apoptosis in THP1 cells partially via blocking the inhibitory effects of EVI-1 on the JNK signaling pathway with the involvement of apoptosis-associated proteins, including Bax, Bcl-2, Bcl-xL and caspase-3. These novel observations may be used to elucidate the mechanism by which ATO induces apoptosis in acute leukemia cells, and provide rationales to develop a personalized medicine strategy for ATO via targeting EVI-1 positive neoplasm.

13.
Int J Biochem Cell Biol ; 47: 113-7, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24361302

RESUMO

The ubiquitin-conjugating enzymes 2C (UBE2C) is an integral component of the ubiquitin proteasome system. UBE2C consists of a conserved core domain containing the catalytic Cys residue and an N-terminal extension. The core domain is required for ubiquitin adduct formation by interacting with the ubiquitin-fold domain in the E1 enzyme, and contributes to the E3 enzyme binding. UBE2C N-terminal extension regulates E3 enzyme activity as a part of an intrinsic inhibitory mechanism. UBE2C is required for the destruction of mitotic cyclins and securin, which are essential for spindle assembly checkpoint and mitotic exit. The UBE2C mRNA and/or protein levels are aberrantly increased in many cancer types with poor clinical outcomes. Accumulation of UBE2C stimulates cell proliferation and anchorage-independent growth. UBE2C transgenic mice are prone to develop spontaneous tumors and carcinogen-induced tumor with evidence of chromosome aneuploidy.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias/enzimologia , Enzimas de Conjugação de Ubiquitina/metabolismo , Sequência de Aminoácidos , Animais , Domínio Catalítico , Humanos , Modelos Moleculares , Enzimas de Conjugação de Ubiquitina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA