7 results on '"You MJ"'
Search Results
2. Acute myeloid leukemia with t(4;12)(q12;p13): an aggressive disease with frequent involvement of PDGFRA and ETV6 .
- Author
-
Li J, Xu J, Abruzzo LV, Tang G, Li S, You MJ, Lu G, Jabbour EJ, Deng Q, Bueso-Ramos CE, Medeiros LJ, and Yin CC
- Abstract
We describe the clinical, morphologic, immunophenotypic and molecular genetic features of 15 cases of acute myeloid leukemia (AML) with t(4;12)(q12;p13). There were 9 men and 6 women, with a median age of 50 years (range, 17-76). Most patients had hypercellular bone marrow with a median blast count of 58% and multilineage dysplasia. Flow cytometry analysis showed myeloid lineage with blasts positive for CD13, CD33, CD34, CD38, CD117 and HLA-DR. Interestingly, aberrant CD7 expression was detected in 12/14 cases, and myeloperoxidase was either negative (3/15) or positive in only a small subset of the blasts (12/15). t(4;12)(q12;p13) was detected at time of initial diagnosis in 4 and at relapse or progression in 9 patients. The initial karyotype was unknown in 2 cases. FISH analysis showed PDGFRA-ETV6 rearrangement in all 7 cases assessed. FLT3 ITD was detected in 2/11 cases and IDH2 and JAK2 mutation were each detected in 1/2 cases assessed. There were no mutations of KRAS (0/8), NRAS (0/8), CEBPA (0/3), KIT (0/3), NPM1 (0/3) or IDH1 (0/2). All patients received aggressive multiagent chemotherapy; 7 patients additionally received stem cell transplantation. With a median follow-up of 10 months (range, 6-51), 13 patients died of AML, 1 patient had persistent disease, and 1 patient was lost to follow-up. In summary, AML with t(4;12)(q12;p13) is usually associated with myelodysplasia, aberrant CD7 expression, weak of absent myeloperoxidase expression, frequent PDGFRA-ETV6 fusion, and an aggressive clinical course. The molecular findings suggest that there may be a role for tyrosine kinase inhibitors in patient management., Competing Interests: CONFLICTS OF INTEREST The authors declare no conflicts of interest.
- Published
- 2017
- Full Text
- View/download PDF
3. Chronic high dose of captopril induces depressive-like behaviors in mice: possible mechanism of regulatory T cell in depression.
- Author
-
Park HS, Han A, Yeo HL, Park MJ, You MJ, Choi HJ, Hong CW, Lee SH, Kim SH, Kim B, and Kwon MS
- Abstract
Major depression has various types of symptoms and disease courses with inconsistent response to monoamine-related antidepressants. Thus, monoamine theory may not be the only pathophysiologic pathway relevant to depression. Recently, it has been suggested that regulatory T cell (Treg) is associated with depression. Based on our previous study that showed decreased regulatory T cell (Treg) population following chronic high-dose captopril (CHC, 40 mg/kg/day * 21 days) administration, we examined whether CHC alone can induce depressive-like behaviors in mice even without stressful stimuli. In this study, we found that CHC induced depressive-like behaviors in tail suspension test (TST) and forced swimming test (FST) without systemic illness, while it did not induce anhedonic behavior, anxiety-like behaviors, or sociality-related behavior. The depressive-like behaviors were rescued by either CHC washout or antidepressant. CHC caused reduction in foxp3 and gata3 mRNA expression in the lymph nodes with elevation in plasma IL-1β and IL-6. Interestingly, CHC increased serum angiotensin II level. In the hippocampus, CHC increased TNF-α and IL-6 mRNA expression with microglia activation while reduced glucocorticoid receptor expression. However, CHC did not affect to hippocampal kynurenine pathway, serotonin level, hypothalamic corticotropin-releasing hormone mRNA level, or serum corticosterone level. Consequently, we propose that CHC may induce a specific form of depressive-like behaviors via Treg reduction and microglial activation., Competing Interests: CONFLICTS OF INTEREST We have no conflicts of interest to declare.
- Published
- 2017
- Full Text
- View/download PDF
4. Tumorigenesis promotes Mdm4-S overexpression.
- Author
-
Pant V, Larsson CA, Aryal N, Xiong S, You MJ, Quintas-Cardama A, and Lozano G
- Subjects
- Aged, Animals, Biomarkers, Cell Cycle Proteins, Cell Line, Tumor, Chromosome Aberrations, Disease Models, Animal, Female, Gene Expression Regulation, Neoplastic, Humans, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Leukemia, Myelogenous, Chronic, BCR-ABL Positive mortality, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Male, Mice, Mice, Transgenic, Middle Aged, Mutation, Polymorphism, Single Nucleotide, RNA Splicing, Cell Transformation, Neoplastic genetics, Gene Expression, Nuclear Proteins genetics, Proto-Oncogene Proteins genetics
- Abstract
Disruption of the p53 tumor suppressor pathway is a primary cause of tumorigenesis. In addition to mutation of the p53 gene itself, overexpression of major negative regulators of p53, MDM2 and MDM4, also act as drivers for tumor development. Recent studies suggest that expression of splice variants of Mdm2 and Mdm4 may be similarly involved in tumor development. In particular, multiple studies show that expression of a splice variant of MDM4, MDM4-S correlates with tumor aggressiveness and can be used as a prognostic marker in different tumor types. However, in the absence of prospective studies, it is not clear whether expression of MDM4-S in itself is oncogenic or is simply an outcome of tumorigenesis. Here we have examined the role of Mdm4-S in tumor development in a transgenic mouse model. Our results suggest that splicing of Mdm4 does not promote tumor development and does not cooperate with other oncogenic insults to alter tumor latency or aggressiveness. We conclude that Mdm4-S overexpression is a consequence of splicing defects in tumor cells rather than a cause of tumor evolution.
- Published
- 2017
- Full Text
- View/download PDF
5. SENP1 promotes proliferation of clear cell renal cell carcinoma through activation of glycolysis.
- Author
-
Dong B, Gao Y, Kang X, Gao H, Zhang J, Guo H, You MJ, Xue W, Cheng J, and Huang Y
- Subjects
- Carcinoma, Renal Cell genetics, Carcinoma, Renal Cell mortality, Carcinoma, Renal Cell pathology, Cell Line, Tumor, Cysteine Endopeptidases genetics, Female, Gene Expression Regulation, Enzymologic, Gene Expression Regulation, Neoplastic, Humans, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Kaplan-Meier Estimate, Kidney Neoplasms genetics, Kidney Neoplasms mortality, Kidney Neoplasms pathology, Male, Middle Aged, Neoplasm Staging, Protein Stability, Proteolysis, RNA Interference, Signal Transduction, Sumoylation, Time Factors, Transfection, Tumor Hypoxia, Von Hippel-Lindau Tumor Suppressor Protein genetics, Von Hippel-Lindau Tumor Suppressor Protein metabolism, Carcinoma, Renal Cell enzymology, Cell Proliferation, Cysteine Endopeptidases metabolism, Glycolysis, Kidney Neoplasms enzymology
- Abstract
Metabolic shift toward aerobic glycolysis is a fundamental element contributing to the development and progression of clear cell renal cell carcinoma (ccRCC). We and others previously observed enhanced glycolysis and diminished tricarboxylic acid (TCA) cycle activity in ccRCC tissue. Here, by integrated gene expression and metabolomic analyses of 36 matched pairs of tumor and adjacent normal tissues, we showed that expression of Sentrin/SUMO-specific protease 1 (SENP1) is positively associated with glycolysis levels in ccRCC. Moreover, SENP1 knockdown in RCC4/VHL cells downregulated expression of key glycolytic enzymes under normoxic and hypoxic conditions and inhibited cell proliferation under hypoxic conditions, possibly due to ineffective deSUMOylation and stablization of Hif-1α related to the SENP-1 deficiency. Finally, SENP1 expression correlated positively with tumor pathological grade and was an indicator of poor overall survival and advanced tumor progression in ccRCC. Altered VHL gene function is found in 60-90% ccRCC cases of ccRCC, but therapies targeting VHL-related signaling pathways have been ineffective, spurring exploration of alternative pathological signaling events. Our results provide a possible mechanistic explanation for the role of SENP1 in the initiation and development of ccRCC with normal VHL activity, and identifies SENP1 as a potential treatment target for the disease.
- Published
- 2016
- Full Text
- View/download PDF
6. Decreased TSPAN1 promotes prostate cancer progression and is a marker for early biochemical recurrence after radical prostatectomy.
- Author
-
Xu F, Gao Y, Wang Y, Pan J, Sha J, Shao X, Kang X, Qin J, You MJ, Huang Y, Dong B, and Xue W
- Subjects
- Aged, Animals, Biomarkers, Tumor metabolism, Cell Cycle, Cell Line, Tumor, Cell Movement, Cell Proliferation, Cohort Studies, Disease Progression, Gene Expression Profiling, HEK293 Cells, Humans, Male, Mice, Middle Aged, Neoplasm Metastasis, Neoplasm Recurrence, Local, Phosphatidylinositol 3-Kinases metabolism, Prognosis, Prostatectomy, Prostatic Neoplasms genetics, Prostatic Neoplasms surgery, Proto-Oncogene Proteins c-akt metabolism, Tetraspanins genetics, Gene Expression Regulation, Neoplastic, Prostatic Neoplasms metabolism, Tetraspanins metabolism
- Abstract
Patients with prostate cancer (PCa) have a variable prognosis. It is challenging to recognize the progressive disease. In this study, we focused on TSPAN1, a new member of the tetraspanin family. Its expression was decreased in progressive PCa and was an independent prognosis factor of biochemical recurrence after radical prostatectomy. In vitro, knockdown and overexpression of TSPAN1 in PCa cell lines showed that TSPAN1 could inhibit cell proliferation and migration. TSPAN1 was positive related to PTEN in both clinical specimen and mouse models. The combination of these two markers could increase their prognosis value especially in low risk patients. In vitro TSPAN1 knockdown resulted in increased Akt phosphorylation and caused evident cell cycle transition from G1 to S phase. Our data suggests that TSPAN1 is a valuable marker to recognize more progressive PCa.
- Published
- 2016
- Full Text
- View/download PDF
7. BAF180 regulates cellular senescence and hematopoietic stem cell homeostasis through p21.
- Author
-
Lee H, Dai F, Zhuang L, Xiao ZD, Kim J, Zhang Y, Ma L, You MJ, Wang Z, and Gan B
- Subjects
- Animals, Blotting, Western, Cell Cycle Checkpoints genetics, Cells, Cultured, Cyclin-Dependent Kinase Inhibitor p21 metabolism, DNA-Binding Proteins, Embryo, Mammalian cytology, Fibroblasts cytology, Fibroblasts metabolism, Gene Expression, HMGB Proteins metabolism, Mice, Knockout, Mice, Transgenic, Reverse Transcriptase Polymerase Chain Reaction, Time Factors, Transcription Factors, Cellular Senescence genetics, Cyclin-Dependent Kinase Inhibitor p21 genetics, HMGB Proteins genetics, Hematopoietic Stem Cells metabolism, Homeostasis genetics
- Abstract
BAF180 (also called PBRM1), a subunit of the SWI/SNF complex, plays critical roles in the regulation of chromatin remodeling and gene transcription, and is frequently mutated in several human cancers. However, the role of mammalian BAF180 in tumor suppression and tissue maintenance in vivo remains largely unknown. Here, using a conditional somatic knockout approach, we explored the cellular and organismal functions of BAF180 in mouse. BAF180 deletion in primary mouse embryonic fibroblasts (MEFs) triggers profound cell cycle arrest, premature cellular senescence, without affecting DNA damage response or chromosomal integrity. While somatic deletion of BAF180 in adult mice does not provoke tumor development, BAF180 deficient mice exhibit defects in hematopoietic system characterized by progressive reduction of hematopoietic stem cells (HSCs), defective long-term repopulating potential, and hematopoietic lineage developmental aberrations. BAF180 deletion results in elevated p21 expression in both MEFs and HSCs. Mechanistically, we showed that BAF180 binds to p21 promoter, and BAF180 deletion enhances the binding of modified histones associated with transcriptional activation on p21 promoter. Deletion of p21 rescues cell cycle arrest and premature senescence in BAF180 deficient MEFs, and partially rescues hematopoietic defects in BAF180 deficient mice. Together, our study identifies BAF180 as a critical regulator of cellular senescence and HSC homeostasis, which is at least partially regulated through BAF180-mediated suppression of p21 expression. Our results also suggest that senescence triggered by BAF180 inactivation may serve as a failsafe mechanism to restrain BAF180 deficiency-associated tumor development, providing a conceptual framework to further understand BAF180 function in tumor biology.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.