Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Genes (Basel) ; 13(12)2022 12 03.
Article in English | MEDLINE | ID: mdl-36553549

ABSTRACT

Pyroptosis is an inflammatory form of programmed necrotic cell death, but its potential prognostic value in acute myeloid leukemia (AML) remains unclear. On the basis of available AML data from TCGA and TARGET databases, a 10-gene signature model was constructed to effectively predict AML prognosis by performing LASSO Cox regression analysis, which showed that patients with a low-risk score had a significantly better prognosis than that of the high-risk group, and receiver operator characteristic (ROC) analysis achieved superior performance in the prognostic model. The model was further well-verified in an external GEO cohort. Multivariable Cox regression analysis showed that, in addition to age, the risk score was an independent poor survival factor for AML patients, and a nomogram model was constructed with high accuracy. Moreover, the high-risk group generally had higher cytolytic activity and increased levels of infiltrating immune cells, including tumor-infiltrating lymphocytes (TILs) and regulatory T cells (Tregs), which could be related to the expression of immune checkpoint genes. Additionally, low-risk AML patients may have a better response from traditional chemotherapeutic drugs. In conclusion, a pyroptosis-related gene signature can independently predict the prognosis of AML patients with sufficient predictive power, and pyroptosis plays an important role in the immune microenvironment of AML, which may be used to develop a new effective therapeutic method for AML in the future.


Subject(s)
Leukemia, Myeloid, Acute , Pyroptosis , Humans , Pyroptosis/genetics , Prognosis , Nomograms , Apoptosis , Leukemia, Myeloid, Acute/genetics , Tumor Microenvironment
2.
Spine (Phila Pa 1976) ; 46(22): E1192-E1201, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34714793

ABSTRACT

STUDY DESIGN: Retrospective analysis. OBJECTIVE: The aim of this study was to develop and validate a competing-risk-based prognostic model and a nomogram for predicting the three- and five-year probability of cancer-specific death (CSD) in patients with spinal and pelvic chondrosarcoma. SUMMARY OF BACKGROUND DATA: The issue of competing risk has rarely been addressed and discussed in survival analysis of bone sarcoma. In addition, the Fine and Gray model, a more accurate method for survival analysis in the context of competing risk, has also been less reported in prognostic study of chondrosarcoma. METHODS: A total of 623 patients with spinal or pelvic chondrosarcoma were identified from the SEER database and were divided into a training and a validation cohort. These two cohorts were used to develop and validate a prognostic model to predict the 3- and 5-year probability of CSD, considering non-CSD as competing risk. The C-index, calibration plot, and decision curve analysis were used to assess the predictive performance and clinical utility of the model. RESULTS: Older age (subdistribution hazards ratio [SHR]: 1.02, 95% confidence interval [CI]: 1.01∼1.03; P = 0.013), high grade (SHR: 2.68, 95% CI: 1.80∼3.99; P < 0.001), regional involvement (SHR: 1.66, 95% CI: 1.06∼2.58; P = 0.026), distant metastasis (SHR: 5.18, 95% CI: 3.11∼8.62; P < 0.001) and radical resection (SHR: 0.38, 95% CI: 0.24∼0.60; P < 0.001) were significantly associated with the incidence of CSD. These factors were used to build a competing-risk-based model and a nomogram to predict CSD. The C-index, calibration plot, and decision curve analysis indicated that the nomogram performs well in predicting CSD and is suitable for clinical use. CONCLUSION: A competing-risk based prognostic model is developed to predict the probability of CSD of patients with spinal and pelvic chondrosarcoma. This nomogram performs well and is suitable for clinical use.Level of Evidence: 4.


Subject(s)
Chondrosarcoma , Aged , Chondrosarcoma/diagnosis , Chondrosarcoma/surgery , Humans , Nomograms , Prognosis , Retrospective Studies , SEER Program
3.
Ann Pharmacother ; 54(6): 517-525, 2020 06.
Article in English | MEDLINE | ID: mdl-31855061

ABSTRACT

Background: The addition of bevacizumab to neoadjuvant chemotherapy improves the pathological complete response rate of human epidermal growth factor 2 (HER2)-negative breast cancer patients. However, the characteristics of adverse events associated with the use of bevacizumab should receive more attention from clinicians. Objective: This meta-analysis aimed to detect the adverse events of adding bevacizumab to neoadjuvant chemotherapy compared with neoadjuvant chemotherapy alone in HER2-negative breast cancer patients. Methods: PubMed, Cochrane Library, Web of Science, and EMBASE databases were systematically accessed to find eligible studies from January 1, 2000, to October 20, 2019. Reference lists were searched for additional studies. Pooled risk ratios for adverse events of bevacizumab were meta-analyzed. Results: Overall, 6 of 829 initially identified studies met the inclusion criteria, with 4681 patients randomized (2321 in the bevacizumab plus neoadjuvant chemotherapy group and 2360 in the neoadjuvant chemotherapy group). The incidence of grade ≥3 hypertension, left-ventricular dysfunction, mucositis, febrile neutropenia, infection, pain, hand-foot syndrome, hemorrhage, and neutropenia significantly increased in patients treated with bevacizumab plus neoadjuvant chemotherapy. However, adding bevacizumab to neoadjuvant chemotherapy was not associated with increasing the incidences of grade ≥3 proteinuria, dyspnea, heart failure, peripheral neurotoxicity, thrombosis, thrombocytopenia, fatigue, leucopenia, vomiting, nausea, and diarrhea. Conclusion and Relevance: Adding bevacizumab to neoadjuvant chemotherapy to treat HER2-negative breast cancer patients increased adverse events. However, most adverse events are clinically manageable. Patients, therefore, need to be monitored carefully for hypertension, left-ventricular dysfunction, mucositis, febrile neutropenia, infection, pain, hand-foot syndrome, hemorrhage, and neutropenia when treated with bevacizumab and neoadjuvant chemotherapy simultaneously.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/adverse effects , Breast Neoplasms/drug therapy , Neoadjuvant Therapy/adverse effects , Receptor, ErbB-2/genetics , Adult , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/administration & dosage , Bevacizumab/therapeutic use , Drug Monitoring , Drug-Related Side Effects and Adverse Reactions/prevention & control , Female , Humans , Middle Aged
4.
Aging (Albany NY) ; 11(12): 3969-3992, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31219799

ABSTRACT

The angiotensin II type I receptor (AGTR1) has a strong influence on tumor growth, angiogenesis, inflammation and immunity. However, the role of AGTR1 on lymph node metastasis (LNM) in breast cancer, which correlates with tumor progression and patient survival, has not been examined. AGTR1 was highly expressed in lymph node-positive tumor tissues, which was confirmed by the Oncomine database. Next, inhibition of AGTR1 reduced tumor growth and LNM in orthotopic xenografts by bioluminescence imaging (BLI). Losartan, an AGTR1-specific inhibitor, decreased the chemokine pair CXCR4/SDF-1α levels invivo and inhibited AGTR1-induced cell migration and invasion invitro. Finally, the molecular mechanism of AGTR1-induced cell migration and LNM was assessed by knocking down AGTR1 in normal cells or CXCR4 in AGTR1high cells. AGTR1-silenced cells treated with losartan showed lower CXCR4 expression. AGTR1 overexpression caused the upregulation of FAK/RhoA signaling molecules, while knocking down CXCR4 in AGTR1high cells downregulated these molecules. Collectively, AGTR1 promotes LNM by increasing the chemokine pair CXCR4/SDF-1α and tumor cell migration and invasion. The potential mechanism of AGTR1-mediated cell movement relies on activating the FAK/RhoA pathway. Our study indicated that inhibiting AGTR1 may be a potential therapeutic target for LNM in early-stage breast cancer.


Subject(s)
Breast Neoplasms/pathology , Chemokine CXCL12/metabolism , Lymph Nodes/pathology , Receptor, Angiotensin, Type 1/metabolism , Receptors, CXCR4/metabolism , Animals , Breast Neoplasms/drug therapy , Cell Movement , Chemokine CXCL12/genetics , Female , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Losartan/administration & dosage , Losartan/pharmacology , Lymphatic Metastasis , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasms, Experimental , Receptor, Angiotensin, Type 1/genetics , Receptors, CXCR4/genetics , Signal Transduction , Up-Regulation , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
5.
Aging (Albany NY) ; 10(10): 2570-2584, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30312171

ABSTRACT

Calcium activated Chloride Channel A4 (CLCA4), as a tumor suppressor, was reported to contribute to the progression of several malignant tumors, yet little is known about the significance of CLCA4 in invasion and prognosis of hepatocellular carcinoma (HCC). CLCA4 expression was negatively correlated with tumor size, vascular invasion and TNM stage. Kaplan-Meier analysis showed that CLCA4 was an independent predictor for overall survival (OS) and time to recurrence (TTR). In addition, CLCA4 status could act as prognostic predictor in different risk of subgroups. Moreover, combination of CLCA4 and serum AFP could be a potential predictor for survival in HCC patients. Furthermore, CLCA4 may inhibit cell migration and invasion by suppressing epithelial-mesenchymal transition (EMT) via PI3K/ATK signaling. Knockdown of CLCA4 significantly increased the migration and invasion of HCC cells and changed the expression pattern of EMT markers and PI3K/AKT phosphorylation. An opposite expression pattern of EMT markers and PI3K/AKT phosphorylation was observed in CLCA4-transfected cells. Additionally, immunohistochemistry and RT-PCR results further confirmed this correlation. Taken together, CLCA4 contributes to migration and invasion by suppressing EMT via PI3K/ATK signaling and predicts favourable prognosis of HCC. CLCA4/AFP expression may help to distinguish different risks of HCC patients after hepatectomy.


Subject(s)
Carcinoma, Hepatocellular/enzymology , Cell Movement , Cell Proliferation , Chloride Channels/metabolism , Epithelial-Mesenchymal Transition , Liver Neoplasms/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Chloride Channels/genetics , Female , Humans , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Phosphorylation , Prognosis , Signal Transduction
6.
Medicine (Baltimore) ; 97(30): e11588, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30045289

ABSTRACT

RATIONALE: Primary cardiac tumors are very rare, and angiosarcoma accounts for about 33% of all primary malignant cardiac tumors. Primary cardiac epithelioid angiosarcoma is a highly aggressive and difficult to diagnose tumor, with early systemic metastasis and poor prognosis. PATIENT CONCERNS: A 35-year-old Han male experienced sudden severe palpitation and moderate dyspnea. The patient received a whole body F-18 fluoro-deoxyglucose positron emission tomography (18F-FDG PET)/computed tomography (CT) scan, the scan showed a large mass in the right atrium (RA) and numerous pulmonary nodules in both lungs. DIAGNOSES: The patient was diagnosed as right atrial epithelioid angiosarcoma with multiple pulmonary metastasis by pulmonary biopsy through CT-guided percutaneous transthoracic fine needle aspiration. INTERVENTIONS: The patient received a cycle of chemotherapy with docetaxel and gemcitabine, followed by another cycle with epirubicin and ifosfamide. OUTCOMES: The chemotherapy was ineffective. After the two cycles, the bilateral pleural effusion steadily increased, the patient had severe dyspnea and palpitation, and died three weeks later, with an overall survival of 2.5 months. LESSONS: Primary angiosarcoma of heart is a very rare and aggressive disease, and its diagnosis and treatment are difficult. Most patients may have systemic metastasis at diagnosis, and have a very short survival without surgical resection. Hence, early diagnosis and surgical resection is extremely important to treat this disease.


Subject(s)
Heart Neoplasms/diagnostic imaging , Hemangioendothelioma, Epithelioid/diagnostic imaging , Hemangiosarcoma/diagnostic imaging , Lung Neoplasms/diagnostic imaging , Lung/diagnostic imaging , Positron Emission Tomography Computed Tomography/methods , Adult , Biopsy/methods , Dyspnea/diagnostic imaging , Dyspnea/etiology , Fluorodeoxyglucose F18 , Heart Atria/diagnostic imaging , Heart Atria/pathology , Heart Neoplasms/complications , Heart Neoplasms/pathology , Hemangioendothelioma, Epithelioid/complications , Hemangioendothelioma, Epithelioid/secondary , Hemangiosarcoma/complications , Hemangiosarcoma/secondary , Humans , Lung/pathology , Lung Neoplasms/complications , Lung Neoplasms/secondary , Male
7.
Blood ; 131(14): 1556-1567, 2018 04 05.
Article in English | MEDLINE | ID: mdl-29439954

ABSTRACT

We recently found that FOXO1 repression contributes to the oncogenic program of classical Hodgkin lymphoma (cHL). Interestingly, FOXO3A, another member of the FOXO family, was reported to be expressed in the malignant Hodgkin and Reed-Sternberg cells of cHL at higher levels than in non-Hodgkin lymphoma subtypes. We thus aimed to investigate mechanisms responsible for the maintenance of FOXO3A as well as the potential role of FOXO3A in cHL. Here, we show that high FOXO3A levels in cHL reflect a B-cell-differentiation-specific pattern. In B cells, FOXO3A expression increases during the process of centroblast to plasma cell (PC) differentiation. FOXO3A levels in cHL were found higher than in germinal center B cells, but lower than in terminally differentiated PCs. This intermediate FOXO3A expression in cHL might manifest the "abortive PC differentiation" phenotype. This assumption was further corroborated by the finding that overexpression of FOXO3A in cHL cell lines induced activation of the master PC transcription factor PRDM1α. As factors attenuating FOXO3A expression in cHL, we identified MIR155 and constitutive activation of extracellular signal-regulated kinase. Finally, we demonstrate the importance of FOXO3A expression in cHL using an RNA interference approach. We conclude that tightly regulated expression of FOXO3A contributes to the oncogenic program and to the specific phenotype of cHL.


Subject(s)
Cell Differentiation , Forkhead Box Protein O3/biosynthesis , Gene Expression Regulation, Neoplastic , Hodgkin Disease/metabolism , Neoplasm Proteins/biosynthesis , Plasma Cells/metabolism , Cell Line, Tumor , Cell Survival , Forkhead Box Protein O3/genetics , Hodgkin Disease/genetics , Hodgkin Disease/pathology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/genetics , Plasma Cells/pathology , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
8.
Oncotarget ; 7(24): 36854-36864, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27166193

ABSTRACT

Although Hodgkin and Reed-Sternberg (HRS) cells of classical Hodgkin lymphoma (cHL) derived from germinal or post germinal B cells, they have lost the B cell phenotype in the process of lymphomagenesis. The phenomenon can be at least partially explained by repression of B-cell-specific transcription factors including TCF3, early B-cell factor 1 (EBF1), SPI1/PU.1, and FOXO1, which are down-regulated by genetic and epigenetic mechanisms. The unique phenotype has been suspected to be advantageous for survival of HRS cells. Ectopic expression of some of these transcription factors (EBF1, PU.1, FOXO1) indeed impaired survival of cHL cells. Here we show that forced expression of TCF3 causes cell death and cell cycle arrest in cHL cell lines. Mechanistically, TCF3 overexpression modulated expression of multiple pro-apoptotic genes including BIK, APAF1, FASLG, BOK, and TNFRSF10A/DR4. We conclude that TCF3 inactivation contributes not only to extinguishing of B cell phenotype but also to cHL oncogenesis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinogenesis/metabolism , Hodgkin Disease/pathology , B-Lymphocytes/metabolism , Cell Line, Tumor , Hodgkin Disease/metabolism , Humans
9.
Oncotarget ; 7(18): 25516-27, 2016 May 03.
Article in English | MEDLINE | ID: mdl-27027440

ABSTRACT

OBJECTIVES: Recent studies suggest that an elevated preoperative platelet to lymphocyte ratio (PLR) may be considered a poor prognostic biomarker in patients with colorectal cancer (CRC). The aim of this study was to evaluate the prognostic impact of PLR in patients with CRC. METHODS: We enrolled 1314 patients who underwent surgery for CRC between 2005 and 2011. Preoperative PLR level was stratified into quintiles for Kaplan-Meier analysis and multivariable Cox proportional hazard regression models. RESULTS: Higher PLR quintiles were significantly associated with poorer overall survival (P = 0.002). Multivariate analysis showed that PLR was an independent risk factor for overall survival (OS) (P = 0.034). Patients in PLR quintile 5 had lower overall survival than in quintile 1 (hazard ratio (HR) = 1.701, 95% confidence interval (CI): 1.267-2.282, P < 0.001). Although patients in PLR quintile 5 had significantly lower disease-free survival (DFS) than in quintile 1 (HR = 1.522, 95% CI: 1.114-2.080, P = 0.008), this association was not significant after multivariable adjustment (P = 0.075). In the subgroup analysis, PLR remained an independent factor in terms of advanced tumor stage (III, IV), male sex, carcinoembryonic antigen (≤ 5 ng/ml), age (> 65 years) and body mass index (≤ 25) (P < 0.05 for all measurements). The results remained unchanged when the PLR was analyzed as a dichotomous variable by applying different cut-off values of 150, 185, 220. CONCLUSIONS: Elevated preoperative PLR was independently associated with an increased risk of mortality in patients with CRC. The utility of PLR may help to improve prognostic predictors.


Subject(s)
Biomarkers, Tumor/blood , Colorectal Neoplasms/blood , Lymphocyte Count , Platelet Count , Adult , Aged , Colorectal Neoplasms/mortality , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Proportional Hazards Models
10.
Sci Rep ; 5: 16835, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26568463

ABSTRACT

FOXO transcription factors especially FOXO1 have profound roles in bone development and remodeling. The regulation of cells of the osteoblast lineage by FOXOs is suggested to be stage-specific or context dependent. Intriguingly, recent studies on the role played by FOXOs in osteoclastogenesis reached different conclusion. Bartell et al. showed that FOXOs restrained osteoclastogenesis and bone resorption partially by upregulation of the H2O2-inactivating enzyme catalase. Wang et al. demonstrated that FOXO1 activated osteoclast formation. In the present study, we confirmed the results of Bartell et al. that FOXO1 expression was reduced upon stimulation of RANKL; FOXO1 inhibition promoted and FOXO1 activation repressed, osteoclast differentiation and activity; the inhibitory effect of FOXO1 on osteoclastogenesis was partially mediated by ROS since treatment with ROS scavengers cancelled the effect of FOXO1 inhibition on osteoclastogenesis. We further investigated the mechanisms responsible for repressed osteoclastogenesis by FOXO1. We found that FOXO1 inhibition modulated MAPKs, NF-κB and AP-1. Finally, we proved that the inhibitory effect of FOXO1 on osteoclast formation was partially mediated by MYC suppression by showing that MYC repression almost totally abrogated the effect of FOXO1 inhibition on osteoclastogenesis. To conclude, our study confirmed FOXO1 as a cell-autonomous inhibitor of osteoclastogenesis.


Subject(s)
Forkhead Transcription Factors/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Forkhead Box Protein O1 , Forkhead Transcription Factors/agonists , Forkhead Transcription Factors/antagonists & inhibitors , Free Radical Scavengers/pharmacology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Osteogenesis/drug effects , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Quinolones/pharmacology , RANK Ligand/pharmacology , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor AP-1/genetics , Up-Regulation/drug effects
11.
Blood ; 124(20): 3118-29, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25232062

ABSTRACT

The survival of classical Hodgkin lymphoma (cHL) cells depends on activation of NF-κB, JAK/STAT, and IRF4. Whereas these factors typically induce the master regulator of plasma cell (PC) differentiation PRDM1/BLIMP-1, levels of PRDM1 remain low in cHL. FOXO1, playing a critical role in normal B-cell development, acts as a tumor suppressor in cHL, but has never been associated with induction of PC differentiation. Here we show that FOXO1 directly upregulates the full-length isoform PRDM1α in cHL cell lines. We also observed a positive correlation between FOXO1 and PRDM1 expression levels in primary Hodgkin-Reed-Sternberg cells. Further, we show that PRDM1α acts as a tumor suppressor in cHL at least partially by blocking MYC. Here we provide a link between FOXO1 repression and PRDM1α downregulation in cHL and identify PRDM1α as a tumor suppressor in cHL. The data support a potential role for FOXO transcription factors in normal PC differentiation.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic , Hodgkin Disease/genetics , Hodgkin Disease/pathology , Plasma Cells/pathology , Repressor Proteins/metabolism , Cell Differentiation , Cell Line, Tumor , Down-Regulation , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Hodgkin Disease/metabolism , Humans , Plasma Cells/cytology , Plasma Cells/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-myc/metabolism , Reed-Sternberg Cells/metabolism , Reed-Sternberg Cells/pathology , Repressor Proteins/genetics , Tumor Cells, Cultured , Up-Regulation
12.
Oncotarget ; 5(14): 5392-402, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-24977668

ABSTRACT

Recently we have shown that the transcription factor FOXO1, highly expressed in B cells, is downregulated in classical Hodgkin lymphoma (cHL). As primary mediastinal B cell lymphoma (PMBL) has similarities with the cHL transcription program we investigated FOXO1 expression in this entity. By using immunohistochemistry we found that FOXO1 was absent or expressed at low levels in 19 of 20 primary PMBL cases. PMBL cell lines reproduce the low FOXO1 expression observed in primary cases. By analyzing gene expression profiling data we found that FOXO1 expression inversely correlated with JAK2 in PMBL cases. Targeting JAK2 activity by the small molecular weight inhibitor TG101348 resulted in upregulation of FOXO1 mRNA and protein expression in MedB-1 and U2940 cell lines, and the MYC inhibitor 10058-F4 increased FOXO1 mRNA in MedB-1 cells. Moreover, in MedB-1 cells FOXO1 expression was strongly upregulated by the inhibitor of DNA methylation 5-aza-2-deoxycytidine and by the histone deacetylase inhibitor trichostatin A. Since FOXO1 promoter was unmethylated, this effect is most likely indirect. FOXO1 activation in the FOXO1-negative Med-B1 cell line led to growth arrest and apoptosis, which was accompanied by repression of MYC and BCL2L1/BCLxL. Thus, FOXO1 repression might contribute to the oncogenic program and phenotype of PMBL.


Subject(s)
Forkhead Transcription Factors/genetics , Lymphoma, B-Cell/genetics , Mediastinal Neoplasms/genetics , Apoptosis/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , DNA Methylation/drug effects , Decitabine , Down-Regulation , Epigenomics , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Humans , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Mediastinal Neoplasms/metabolism , Mediastinal Neoplasms/pathology , Signal Transduction
13.
J Pathol ; 229(5): 775-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23341364

ABSTRACT

Burkitt lymphoma (BL) is caused by translocation of the MYC gene to an immunoglobulin locus resulting in its constitutive expression depending on the activity of the immunoglobulin (Ig) enhancer elements. Treatment of BL cell lines with epigenetic modifiers is known to repress B-cell-specific genes and to up-regulate B-cell-inappropriate genes including the transcription repressor ID2 expression. We found that the DNA methyltransferase inhibitor decitabine/5-aza-2-deoxycytidine (5-aza-dC) represses the MYC oncogene on RNA and protein levels by inducing ID2. Down-regulation of MYC was associated with repression of transcriptional activity of the Ig locus and with inhibition of proliferation. The induction of ID2 can be in part explained by activation of the transcription factor NF-κB. We conclude that up-regulation of ID2 contributes to anti-tumour activity of 5-aza-dC via repression of Ig locus activity and consequently MYC expression.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Burkitt Lymphoma/genetics , DNA Modification Methylases/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/genetics , Translocation, Genetic/drug effects , Azacitidine/pharmacology , Burkitt Lymphoma/enzymology , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Decitabine , Dose-Response Relationship, Drug , Epigenetic Repression , Gene Expression Regulation, Neoplastic , Humans , Immunoglobulin M/genetics , Immunoglobulin M/metabolism , Inhibitor of Differentiation Protein 2/genetics , Inhibitor of Differentiation Protein 2/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Transcription, Genetic , Transfection , Up-Regulation
14.
Blood ; 119(15): 3503-11, 2012 Apr 12.
Article in English | MEDLINE | ID: mdl-22343918

ABSTRACT

The FOXO transcription factors control proliferation and apoptosis in different cell types. Their activity is regulated by posttranslational modifications, mainly by the PI3K-PKB pathway, which controls nuclear export and degradation. We show that FOXO1 is highly expressed in normal germinal center B cells as well as in non-Hodgkin lymphomas, including follicular lymphoma, diffuse large B-cell lymphoma, mucosa-associated lymphoid tissue non-Hodgkin lymphoma, B-cell chronic lymphocytic leukemia, and mantle cell lymphoma. In contrast, in 31 of 32 classical Hodgkin lymphoma (cHL) cases, Hodgkin and Reed-Sternberg cells were FOXO1 negative. Neoplastic cells of nodular lymphocyte-predominant Hodgkin lymphoma were negative in 14 of 20 cases. FOXO1 was down-regulated in cHL cell lines, whereas it was expressed in non-Hodgkin lymphoma cell lines at levels comparable with normal B cells. Ectopic expression of a constitutively active FOXO1 induced apoptosis in cHL cell lines and blocked proliferation, accompanied with cell-cycle arrest in the G(0)/G(1) phase. We found that, in cHL cell lines, FOXO1 is inactivated by multiple mechanisms, including constitutive activation of AKT/PKB and MAPK/ERK kinases and up-regulation of microRNAs miR-96, miR-182, and miR-183. These results suggest that FOXO1 repression contributes to cHL lymphomagenesis.


Subject(s)
Forkhead Transcription Factors/physiology , Genes, Tumor Suppressor , Hodgkin Disease/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Deletion , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Genetic Loci/genetics , Hodgkin Disease/pathology , Humans , MicroRNAs/genetics , MicroRNAs/physiology , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Oncogene Protein v-akt/physiology , Tissue Distribution
15.
Blood ; 116(9): 1469-78, 2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20519630

ABSTRACT

The transcription factor KLF4 may act both as an oncogene and a tumor suppressor in a tissue-depending manner. In T- and pre-B-cell lymphoma, KLF4 was found to act as tumor suppressor. We found the KLF4 promoter methylated in B-cell lymphoma cell lines and in primary cases of B-cell lymphomas, namely, follicular lymphoma, diffuse large B-cell lymphoma, Burkitt lymphoma, and in classic Hodgkin lymphoma (cHL) cases. Promoter hypermethylation was associated with silencing of KLF4 expression. Conditional overexpression of KLF4 in Burkitt lymphoma cell lines moderately retarded proliferation, via cell-cycle arrest in G(0)/G(1). In the cHL cell lines, KLF4 induced massive cell death that could partially be inhibited with Z-VAD.fmk. A quantitative reverse-transcribed polymerase chain reaction array revealed KLF4 target genes, including the proapoptotic gene BAK1. Using an shRNA-mediated knock-down approach, we found that BAK1 is largely responsible for KLF4-induced apoptosis. In addition, we found that KLF4 negatively regulates CXCL10, CD86, and MSC/ABF-1 genes. These genes are specifically up-regulated in HRS cells of cHL and known to be involved in establishing the cHL phenotype. We conclude that epigenetic silencing of KLF4 in B-cell lymphomas and particularly in cHL may favor lymphoma survival by loosening cell-cycle control and protecting from apoptosis.


Subject(s)
Burkitt Lymphoma/metabolism , Genes, Tumor Suppressor , Hodgkin Disease/metabolism , Kruppel-Like Transcription Factors/physiology , Lymphoma, Follicular/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Apoptosis , B-Lymphocytes/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Child , Child, Preschool , DNA Methylation , DNA, Neoplasm/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hodgkin Disease/genetics , Hodgkin Disease/pathology , Humans , Kruppel-Like Factor 4 , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...