Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
EMBO J ; 42(2): e111268, 2023 01 16.
Article in English | MEDLINE | ID: mdl-36408830

ABSTRACT

Reprogramming of lipid metabolism is emerging as a hallmark of cancer, yet involvement of specific fatty acids (FA) species and related enzymes in tumorigenesis remains unclear. While previous studies have focused on involvement of long-chain fatty acids (LCFAs) including palmitate in cancer, little attention has been paid to the role of very long-chain fatty acids (VLCFAs). Here, we show that depletion of acetyl-CoA carboxylase (ACC1), a critical enzyme involved in the biosynthesis of fatty acids, inhibits both de novo synthesis and elongation of VLCFAs in human cancer cells. ACC1 depletion markedly reduces cellular VLCFA but only marginally influences LCFA levels, including palmitate that can be nutritionally available. Therefore, tumor growth is specifically susceptible to regulation of VLCFAs. We further demonstrate that VLCFA deficiency results in a significant decrease in ceramides as well as downstream glucosylceramides and sphingomyelins, which impairs mitochondrial morphology and renders cancer cells sensitive to oxidative stress and cell death. Taken together, our study highlights that VLCFAs are selectively required for cancer cell survival and reveals a potential strategy to suppress tumor growth.


Subject(s)
Neoplasms , Stearates , Humans , Stearates/metabolism , Fatty Acids/metabolism , Mitochondria/metabolism , Palmitates/metabolism , Neoplasms/genetics , Neoplasms/metabolism
3.
Acta Biochim Biophys Sin (Shanghai) ; 54(2): 252-260, 2022 Feb 25.
Article in English | MEDLINE | ID: mdl-35538024

ABSTRACT

Focal adhesion kinase (FAK), a multi-functional cytoplasmic tyrosine kinase, plays a critical role in cancer migration, proliferation and metastasis via regulating multiple signaling pathways. SY-707 is an anaplastic lymphoma kinase (ALK)/FAK/type 1 insulin-like growth factor receptor (IGF1R) multi-kinase inhibitor which is now being evaluated in phase II clinical trials for ALK positive non-small cell lung cancer (NSCLC). However, the effect of SY-707 on breast cancer is unknown. In this study, we assessed preclinical the anti-growth and anti-metastasis potency of SY-707 in breast cancer cells. ATP content, PE-Annexin V, and would healing assays were used to examine cell proliferation, cell cycle and migration. Then, SD rat and beagle dog models were used to evaluate the pharmacokinetics profile of SY-707, and mouse xenograft model was used to evaluate the anti-cancer activities of SY-707 . We found that breast cancer cells apoptosis were induced by SY-707. Moreover, SY-707 exerted inhibition on cell migration and adhesion in a dose-dependent manner. In T47D xenograft mice, SY-707 had significant anti-tumor activities alone or synergistically with Paclitaxel. Meanwhile, SY-707 also displayed significant suppression on spontaneous metastasis of tumor to the lung in 4T1 murine breast cancer xenograft model. In conclusion, SY-707 has potent anti-proliferation and anti-migration potential in breast cancer and , implying its therapeutic application for the treatment of breast cancer in future clinical trials.


Subject(s)
Breast Neoplasms , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Dogs , Female , Focal Adhesion Protein-Tyrosine Kinases , Humans , Lung Neoplasms/drug therapy , Mice , Neoplasm Metastasis , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, IGF Type 1 , Xenograft Model Antitumor Assays
4.
Nat Commun ; 15(1): 6915, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134530

ABSTRACT

Protein post-translational modifications (PTMs) are crucial for cancer cells to adapt to hypoxia; however, the functional significance of lysine crotonylation (Kcr) in hypoxia remains unclear. Herein we report a quantitative proteomics analysis of global crotonylome under normoxia and hypoxia, and demonstrate 128 Kcr site alterations across 101 proteins in MDA-MB231 cells. Specifically, we observe a significant decrease in K131cr, K156cr and K220cr of phosphoglycerate kinase 1 (PGK1) upon hypoxia. Enoyl-CoA hydratase 1 (ECHS1) is upregulated and interacts with PGK1, leading to the downregulation of PGK1 Kcr under hypoxia. Abolishment of PGK1 Kcr promotes glycolysis and suppresses mitochondrial pyruvate metabolism by activating pyruvate dehydrogenase kinase 1 (PDHK1). A low PGK1 K131cr level is correlated with malignancy and poor prognosis of breast cancer. Our findings show that PGK1 Kcr is a signal in coordinating glycolysis and the tricarboxylic acid (TCA) cycle and may serve as a diagnostic indicator for breast cancer.


Subject(s)
Breast Neoplasms , Citric Acid Cycle , Glycolysis , Phosphoglycerate Kinase , Phosphoglycerate Kinase/metabolism , Phosphoglycerate Kinase/genetics , Humans , Glycolysis/genetics , Cell Line, Tumor , Female , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Lysine/metabolism , Protein Processing, Post-Translational , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Down-Regulation , Mice , Proteomics/methods , Mice, Nude , Gene Expression Regulation, Neoplastic , Mitochondria/metabolism , Cell Hypoxia , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/genetics
5.
J Exp Med ; 220(3)2023 03 06.
Article in English | MEDLINE | ID: mdl-36520461

ABSTRACT

Fatty acid uptake is essential for cell physiological function, but detailed mechanisms remain unclear. Here, we generated an acetyl-CoA carboxylases (ACC1/2) double-knockout cell line, which lacked fatty acid biosynthesis and survived on serum fatty acids and was used to screen for fatty acid uptake inhibitors. We identified a Food and Drug Administration-approved tricyclic antidepressant, nortriptyline, that potently blocked fatty acid uptake both in vitro and in vivo. We also characterized underlying mechanisms whereby nortriptyline provoked lysosomes to release protons and induce cell acidification to suppress macropinocytosis, which accounted for fatty acid endocytosis. Furthermore, nortriptyline alone or in combination with ND-646, a selective ACC1/2 inhibitor, significantly repressed tumor growth, lipogenesis, and hepatic steatosis in mice. Therefore, we show that cells actively take up fatty acids through macropinocytosis, and we provide a potential strategy suppressing tumor growth, lipogenesis, and hepatic steatosis through controlling the cellular level of fatty acids.


Subject(s)
Fatty Liver , Metabolic Diseases , Neoplasms , Mice , Animals , Fatty Acids/metabolism , Antidepressive Agents, Tricyclic/pharmacology , Antidepressive Agents, Tricyclic/therapeutic use , Antidepressive Agents, Tricyclic/metabolism , Nortriptyline/metabolism , Nortriptyline/therapeutic use , Drug Repositioning , Fatty Liver/pathology , Metabolic Diseases/metabolism , Neoplasms/pathology , Liver/metabolism
6.
Nat Cell Biol ; 25(6): 836-847, 2023 06.
Article in English | MEDLINE | ID: mdl-37291265

ABSTRACT

De novo pyrimidine biosynthesis is achieved by cytosolic carbamoyl-phosphate synthetase II, aspartate transcarbamylase and dihydroorotase (CAD) and uridine 5'-monophosphate synthase (UMPS), and mitochondrial dihydroorotate dehydrogenase (DHODH). However, how these enzymes are orchestrated remains enigmatical. Here we show that cytosolic glutamate oxaloacetate transaminase 1 clusters with CAD and UMPS, and this complex then connects with DHODH, which is mediated by the mitochondrial outer membrane protein voltage-dependent anion-selective channel protein 3. Therefore, these proteins form a multi-enzyme complex, named 'pyrimidinosome', involving AMP-activated protein kinase (AMPK) as a regulator. Activated AMPK dissociates from the complex to enhance pyrimidinosome assembly but inactivated UMPS, which promotes DHODH-mediated ferroptosis defence. Meanwhile, cancer cells with lower expression of AMPK are more reliant on pyrimidinosome-mediated UMP biosynthesis and more vulnerable to its inhibition. Our findings reveal the role of pyrimidinosome in regulating pyrimidine flux and ferroptosis, and suggest a pharmaceutical strategy of targeting pyrimidinosome in cancer treatment.


Subject(s)
Ferroptosis , Neoplasms , Dihydroorotate Dehydrogenase , AMP-Activated Protein Kinases , Pyrimidines/pharmacology , Cell Proliferation
7.
EPMA J ; 13(3): 519-534, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36061829

ABSTRACT

Background: Pancreatic cancer presents extremely poor prognosis due to the difficulty of early diagnosis, low resection rate, and high rates of recurrence and metastasis. Immune checkpoint blockades have been widely used in many cancer types but showed limited efficacy in pancreatic cancer. The current study aimed to evaluate the landscape of tumor microenvironment (TME) of pancreatic cancer and identify the potential markers of prognosis and immunotherapy efficacy which might contribute to improve the targeted therapy strategy and efficacy in pancreatic cancer in the context of predictive, preventive, and personalized medicine (PPPM). Methods: In the current study, a total of 382 pancreatic samples from the datasets of Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) were selected. LM22 gene signature matrix was applied to quantify the fraction of immune cells based on "CIBERSORT" algorithm. Weighted Gene Co-expression Network Analysis (WGCNA) and Molecular Complex Detection (MCODE) algorithm was applied to confirm the hub-network of immune-resistance phenotype. A nomogram model based on COX and Logistic regression was constructed to evaluate the prognostic value and the predictive value of hub-gene in immune-response. The role of transmembrane protein 92 (TMEM92) in regulating cell proliferation was evaluated by MTS assay. Western blot and Real-time PCR were applied to assess the biological effects of PD-L1 inhibition by TMEM92. Moreover, the effect of TMEM92 in immunotherapy was evaluated with PBMC co-culture and by MTS assay. Results: Two tumor-infiltrating immune cell (TIIC) phenotypes were identified and a weighted gene co-expression network was constructed to confirm the 167 gene signatures correlated with immune-resistance TIIC subtype. TMEM92 was further identified as a core gene of 167 gene signature network based on MCODE algorithm. High TMEM92 expression was significantly correlated with unfavorable prognosis, characterizing by immune resistance. A nomogram model and external validation confirmed that TMEM92 was an independent prognostic factor in pancreatic cancer. An elevated tumor mutation burden (TMB), mostly is consistent with commonly mutations of KRAS and TP53, was found in the high TMEM92 group. The predictive role of TMEM92 in immunotherapeutic response was also confirmed by IMvigor210 datasets. In addition, the specific biological roles of TMEM92 in cancer was explored in vitro. The results showed that abnormal overexpression of TMEM92 was significantly associated with the poor survival rate of pancreatic cancer. Moreover, we demonstrated that TMEM92 inhibit tumour immune responses of the anti-PD-1 antibody with PBMC co-culture. Conclusion: The current study explored for the first time the immune-resistance phenotype of pancreatic cancer and identified TMEM92 as an innovative marker in predicting clinical outcomes and immunotherapeutic efficacy. These findings not only help to recognize high-risk and immune-resistance population which could be supplied targeted prevention, but also provide personalized medical services by intervening TMEM92 function to improve the prognosis of pancreatic cancer. In addition, the biological role of TMEM92 might reveal the potential molecular mechanisms of pancreatic cancer and lead to a novel sight for development of a PPPM approach for pancreatic cancer management. Supplementary Information: The online version contains supplementary material available at 10.1007/s13167-022-00287-0.

8.
Nat Commun ; 13(1): 7031, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36396642

ABSTRACT

An enhanced NADH/NAD+ ratio, termed reductive stress, is associated with many diseases. However, whether a downstream sensing pathway exists to mediate pathogenic outcomes remains unclear. Here, we generate a soluble pyridine nucleotide transhydrogenase from Escherichia coli (EcSTH), which can elevate the NADH/NAD+ ratio and meantime reduce the NADPH/NADP+ ratio. Additionally, we fuse EcSTH with previously described LbNOX (a water-forming NADH oxidase from Lactobacillus brevis) to resume the NADH/NAD+ ratio. With these tools and by using genome-wide CRISPR/Cas9 library screens and metabolic profiling in mammalian cells, we find that accumulated NADH deregulates PRPS2 (Ribose-phosphate pyrophosphokinase 2)-mediated downstream purine biosynthesis to provoke massive energy consumption, and therefore, the induction of energy stress. Blocking purine biosynthesis prevents NADH accumulation-associated cell death in vitro and tissue injury in vivo. These results underscore the pathophysiological role of deregulated purine biosynthesis in NADH accumulation-associated disorders and demonstrate the utility of EcSTH in manipulating NADH/NAD+ and NADPH/NADP+.


Subject(s)
Escherichia coli , NAD , Animals , NADP/metabolism , NAD/metabolism , Oxidation-Reduction , Escherichia coli/metabolism , Cell Death , Mammals/metabolism
9.
Biochem Biophys Res Commun ; 408(1): 84-8, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21458412

ABSTRACT

Human CAP10-like protein 46 kDa (hCLP46) is the homolog of Rumi, which is the first identified protein O-glucosyltransferase that modifies Notch receptor in Drosophila. Dysregulation of hCLP46 occurs in many hematologic diseases, but the role of hCLP46 remains unclear. Knockdown of hCLP46 by RNA interference resulted in decreased protein levels of endogenous Notch1, Notch intracellular domain (NICD) and Notch target gene Hes-1, suggesting the impairment of the Notch signaling. However, neither cell surface Notch expression nor ligand binding activities were affected. In addition, down-regulated expression of hCLP46 inhibited the proliferation of U937 cells, which was correlated with increased cyclin-dependent kinase inhibitor (CDKI) CDKN1B (p27) and decreased phosphorylation of retinoblastoma (RB) protein. We showed that lack of hCLP46 results in impaired ligand induced Notch activation in mammalian cell, and hCLP46 regulates the proliferation of U937 cell through CDKI-RB signaling pathway, which may be important for the pathogenesis of leukemia.


Subject(s)
Cell Proliferation , Proteins/physiology , Receptors, Notch/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , CDC2 Protein Kinase/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Gene Knockdown Techniques , Glucosyltransferases , Homeodomain Proteins/metabolism , Humans , Jurkat Cells , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , Proteins/genetics , RNA Interference , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Notch/genetics , Retinoblastoma Protein/metabolism , Signal Transduction , Transcription Factor HES-1 , U937 Cells
10.
Cancer Med ; 10(11): 3689-3699, 2021 06.
Article in English | MEDLINE | ID: mdl-33960694

ABSTRACT

The marked overexpression of cyclin-dependent kinase 5 (CDK5) or Notch1 receptor, which plays critical roles in pancreatic ductal adenocarcinoma (PDAC) development, has been detected in numerous PDAC cell lines and tissues. Although, a previous study has demonstrated that CDK5 inhibition disrupts Notch1 functions in human umbilical vein endothelial cells, the mechanism underlying Notch1 activation regulated by CDK5 remains unclear. Herein, we identified a physical interaction between CDK5 and Notch1 in PDAC cells, with the Notch1 peptide phosphorylated by CDK5/p25 kinase. CDK5 blockade resulted in the profound inhibition of Notch signaling. Accordingly, CDK5 inhibition sensitized PDAC cell proliferation and migration following Notch inhibition. In conclusion, CDK5 positively regulates Notch1 function via phosphorylation, which in turn promotes cell proliferation and migration. The combinational inhibition of CDK5 and Notch signaling may be an effective strategy in the treatment of PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cyclin-Dependent Kinase 5/metabolism , Pancreatic Neoplasms/metabolism , Receptor, Notch1/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclin-Dependent Kinase 5/antagonists & inhibitors , Cyclin-Dependent Kinase 5/genetics , Dipeptides/pharmacology , Gene Silencing , Humans , Immunoprecipitation , Pancreatic Neoplasms/pathology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Roscovitine/pharmacology , Signal Transduction
11.
J Exp Med ; 217(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-31961917

ABSTRACT

Cancer cells often proliferate under hypoxia and reprogram their metabolism. However, how to find targets to effectively block the hypoxia-associated metabolic pathways remains unclear. Here, we developed a tool to conveniently calculate electrons dissipated in metabolic transformations. Based on the law of conservation of electrons in chemical reactions, we further built up an electron balance model for central carbon metabolism, and it can accurately outline metabolic plasticity under hypoxia. Our model specifies that glutamine metabolism reprogrammed for biosynthesis of lipid and/or proline actually acts as the alternative electron bin to enable electron transfer in proliferating cells under hypoxia. Inhibition of both proline biosynthesis and lipogenesis can synergistically suppress cancer cell growth under hypoxia and in vivo tumor onset. Therefore, our model helps to reveal combinations of potential targets to inhibit tumor growth by blocking hypoxia-rewired metabolism and provides a useful tool for future studies on cancer metabolism.


Subject(s)
Cell Proliferation/physiology , Lipogenesis/physiology , Neoplasms/metabolism , Proline/biosynthesis , Proline/metabolism , A549 Cells , Animals , Cell Hypoxia/physiology , Cell Line, Tumor , Female , Glutamine/metabolism , HeLa Cells , Hep G2 Cells , Humans , MCF-7 Cells , Metabolic Networks and Pathways/physiology , Mice , Mice, Nude
12.
J Hum Genet ; 54(8): 457-60, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19590515

ABSTRACT

Normal function of the dopaminergic system is necessary for speech fluency. There was evidence that the activities of dopamine transporter (DAT) and dopamine D2 receptor (DRD2) could be altered in people with speech disfluency. This study aims to ascertain the possible correlation between two dopaminergic genes (SLC6A3 and DRD2) and disorder of speech fluency, and to determine the allelic frequencies of the five single-nucleotide polymorphisms (SNPs) (rs2617604, rs28364997, rs28364998 in SLC6A3 and rs6275, rs6277 in DRD2) among Han Chinese patients with this disorder. A sample of 112 patients with speech disfluency and 112 gender-matched controls were included in this case-control study. The results show that the presence of C allele at rs6277 in DRD2 gene is associated with increased susceptibility to the disorder, whereas T allele is protective. Haplotype 939T/957T is also a protective factor.


Subject(s)
Asian People/genetics , Dopamine Plasma Membrane Transport Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Receptors, Dopamine D2/genetics , Stuttering/genetics , Adult , Case-Control Studies , China , Female , Genetic Predisposition to Disease , Genotype , Haplotypes/genetics , Humans , Male , Polymerase Chain Reaction , Young Adult
13.
Nat Commun ; 10(1): 201, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30643150

ABSTRACT

Under hypoxia, most of glucose is converted to secretory lactate, which leads to the overuse of glutamine-carbon. However, under such a condition how glutamine nitrogen is disposed to avoid over-accumulating ammonia remains to be determined. Here we identify a metabolic flux of glutamine to secretory dihydroorotate, which is indispensable to glutamine-carbon metabolism under hypoxia. We found that glutamine nitrogen is necessary to nucleotide biosynthesis, but enriched in dihyroorotate and orotate rather than processing to its downstream uridine monophosphate under hypoxia. Dihyroorotate, not orotate, is then secreted out of cells. Furthermore, we found that the specific metabolic pathway occurs in vivo and is required for tumor growth. The identified metabolic pathway renders glutamine mainly to acetyl coenzyme A for lipogenesis, with the rest carbon and nitrogen being safely removed. Therefore, our results reveal how glutamine carbon and nitrogen are coordinatively metabolized under hypoxia, and provide a comprehensive understanding on glutamine metabolism.


Subject(s)
Glutamine/metabolism , Metabolic Networks and Pathways , Metabolome , Neoplasms/metabolism , Orotic Acid/analogs & derivatives , Acetyl Coenzyme A/metabolism , Ammonia/metabolism , Ammonia/toxicity , Animals , Carbon/chemistry , Carbon/metabolism , Cell Hypoxia , Cell Line, Tumor , Cell Survival , Female , Glucose/metabolism , Glutamine/chemistry , HEK293 Cells , Humans , Lactic Acid/metabolism , Lipogenesis , Metabolomics , Mice , Mice, Nude , Neoplasms/blood , Neoplasms/mortality , Neoplasms/pathology , Nitrogen/chemistry , Nitrogen/metabolism , Nucleotides/biosynthesis , Orotic Acid/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
14.
OMICS ; 21(5): 266-274, 2017 05.
Article in English | MEDLINE | ID: mdl-28481732

ABSTRACT

Colon cancer patients have major unmet needs in terms of robust diagnostics and molecular biomarkers for personalized therapeutics. We have previously reported that human CAP10-like protein 46 kDa (hCLP46) is overexpressed in human acute myelogenous leukemia, T acute lymphoblastic leukemia, and leukemia cell lines. We extend this line of biomarker and diagnostic discovery research by investigating hCLP46 expression in colorectal cancer (CRC) tissues and examine the possibility of hCLP46 as a candidate biomarker for diagnosis and prognosis of CRC. Using a tissue microarray analysis approach, we found that hCLP46 is (1) overexpressed in 90 CRC tissues compared with 90 matched noncancerous tissues and (2) positively correlated with higher tumor-node-metastasis (TNM) stage, lymph node metastasis, and shorter survival time. Moreover, in vitro experiments demonstrated that downregulation of hCLP46 in CRC cells results in proliferation arrest and adhesion enhancement, while apoptosis is unchanged. Further transcriptome profile analysis corroborated that the adhesion pathway is related to hCLP46 downregulation. This report for the first time, to the best of our knowledge, demonstrates that hCLP46 promotes tumor malignancy in CRC cells. We suggest that hCLP46 is warranted for further research as a candidate biomarker for clinical phenotypes related to colon cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Glucosyltransferases/metabolism , Adult , Biomarkers, Tumor/genetics , Caco-2 Cells , Colonic Neoplasms/genetics , Colonic Neoplasms/mortality , Down-Regulation , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glucosyltransferases/genetics , HCT116 Cells , Humans , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Neoplasm Staging , Precision Medicine , Prognosis
17.
Genet Test Mol Biomarkers ; 14(1): 127-33, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20143914

ABSTRACT

AIMS: We earlier identified a novel gene human CAP10-like protein 46 KD (hCLP46) from human acute myelogenous leukemia (AML) transformed from myelodysplastic syndrome CD34(+) cells, but the function of this gene remains unclear. In this study, a real-time polymerase chain reaction-based assay was developed to quantify expression of hCLP46 in the peripheral blood of AML and T-acute lymphoblastic leukemia (T-ALL) primary samples and in six leukemic cell lines. Also, we investigated expression of CDKN2A/B and the apoptosis in U937 cells when hCLP46 is downregulated in vitro. RESULTS: Our findings showed that hCLP46 was overexpressed in AML, T-ALL, and the leukemic cell lines. Suppressing hCLP46 overexpression had no effect on expression of CDKN2A/B and apoptosis of U937 cells. CONCLUSION: Considering that hCLP46 has the capability of modifying the Notch pathway, our finding adds weight to the importance of Notch signaling in hematopoiesis and suggests that overexpression of hCLP46 might be an early event in the pathogenesis of AML and T-ALL.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Proteins/genetics , Apoptosis/genetics , Base Sequence , Cell Line, Tumor , DNA Primers/genetics , Gene Expression , Genes, p16 , Glucosyltransferases , Hematopoiesis/genetics , Hematopoiesis/physiology , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/physiopathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/physiopathology , Proteins/antagonists & inhibitors , RNA, Small Interfering/genetics , Receptors, Notch/physiology , Signal Transduction , Transfection , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL