Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cell Death Dis ; 15(5): 378, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816356

ABSTRACT

Lung adenocarcinoma (LUAD) remains a leading cause of cancer-related mortality worldwide. Understanding the dysregulated epigenetics governing LUAD progression is pivotal for identifying therapeutic targets. CBX4, a chromobox protein, is reported to be upregulated in LUAD. This study highlights the dual impact of CBX4 on LUAD proliferation and metastasis through a series of rigorous in vitro and in vivo experiments. Further investigation into the underlying mechanism through high-throughput ChIP-seq and RNA-seq reveals that CBX4 functions in promoting LUAD proliferation via upregulating PHGDH expression and subsequent serine biosynthesis, while concurrently suppressing LUAD metastasis by inhibiting ZEB2 transcription. CBX4 facilitates PHGDH transcription through the interaction with GCN5, inducing heightened histone acetylation on the PHGDH promoter. Simultaneously, the inhibition of ZEB2 transcription involves CBX4-mediated recruitment of canonical PRC1 (cPRC1), establishing H2K119ub on the ZEB2 promoter. These findings underscore CBX4's pivotal role as a regulator of LUAD progression, emphasizing its diverse transcriptional regulatory functions contingent upon interactions with specific epigenetic partners. Understanding the nuanced interplay between CBX4 and epigenetic factors sheds light on potential therapeutic avenues in LUAD.


Subject(s)
Adenocarcinoma of Lung , Disease Progression , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Polycomb-Group Proteins , Animals , Humans , Mice , A549 Cells , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Adenocarcinoma of Lung/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Ligases , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Mice, Nude , Polycomb-Group Proteins/metabolism , Polycomb-Group Proteins/genetics , Promoter Regions, Genetic/genetics , Transcription, Genetic , Zinc Finger E-box Binding Homeobox 2/metabolism , Zinc Finger E-box Binding Homeobox 2/genetics
2.
Mol Ther Nucleic Acids ; 34: 102063, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38028203

ABSTRACT

Ferroptosis is an iron-catalyzed form of regulated cell death that results from the accumulation of lipid peroxidation products and reactive oxygen species to a lethal content. However, the transcriptional regulation of ferroptosis is not well understood. Sorafenib, a standard drug for hepatocellular carcinoma (HCC), induces ferroptosis in HCC cells. In this study, we conducted a CRISPR-Cas9 library screening targeting epigenetic factors and identified coactivator-associated arginine methyltransferase 1 (CARM1) as a critical inhibitor of ferroptosis. CARM1 depletion intensified Sorafenib-induced ferroptosis, resulting in decreased cell viability, reduced cellular glutathione level, increased lipid peroxidation, and altered mitochondrial crista structure. Additionally, we investigated a CARM1 inhibitor (CARM1i) as a potential ferroptosis inducer. Combining the CARM1i with Sorafenib enhanced the induction of ferroptosis. Notably, both CARM1 knockdown and CARM1i showed cooperative effects with Sorafenib in inhibiting HCC growth in mice. The underlying mechanism involves CARM1-catalyzed H3R26me2a on the promoter of glutathione peroxidase 4, leading to its transcriptional activation and subsequent ferroptosis inhibition. Furthermore, Sorafenib treatment induced the transcription of CARM1 through the MDM2-p53 axis. In summary, our findings establish CARM1 as a critical ferroptosis inhibitor and highlight the potential of CARM1is as novel ferroptosis inducers, providing promising therapeutic strategies for HCC treatment.

3.
J Cell Physiol ; 238(11): 2710-2723, 2023 11.
Article in English | MEDLINE | ID: mdl-37733753

ABSTRACT

Dysregulation of polycomb group (PcG) proteins that mediate epigenetic gene silencing contributes to tumorigenesis. As core components of the polycomb repressive complex 1 (PRC1), chromobox (CBX) proteins recognize H3K27me3 to recruit PRC1 to maintain a repressive transcriptional state. However, the individual biological functions of these CBX proteins in tumorigenesis warrant in-depth investigation. In this study, we analyzed the mRNA expression of CBX family genes across multiple cancers using The Cancer Genome Atlas data and found different expression patterns of the five CBX genes in different types of cancer. This analyses together with the result of immunohistochemistry indicated that CBX8 expression was significantly higher in lung adenocarcinoma (LUAD) tissues compared to adjacent nontumor tissues. Overexpression approaches demonstrated that CBX8 facilitated LUAD cell proliferation and migration in vitro. Consistently, CBX8 knockdown reduced LUAD cell proliferation and migration in both cell culture and mouse models. RNA sequencing combined with real-time RT-PCR assays revealed CDKN2C and SCEL as target genes of CBX8. Furthermore, chromatin immunoprecipitation assays indicated that CBX8 directly bound to the promoters of CDKN2C and SCEL to establish H2AK119ub. CBX8 depletion reduced the enrichment of H2AK119ub on CDKN2C and SCEL promoters. Moreover, depletion of CDKN2C and SCEL restored the repressed growth and invasion ability of LUAD cells caused by CBX8 knockdown. These findings demonstrate that CBX8 promotes LUAD growth and metastasis through the transcriptional repression of CDKN2C and SCEL. Our study uncovers the oncogenic role of CBX8 in LUAD progression and provides a new target for the diagnosis and therapy of LUAD.


Subject(s)
Adenocarcinoma of Lung , Carrier Proteins , Cyclin-Dependent Kinase Inhibitor p18 , Gene Expression Regulation, Neoplastic , Lung Neoplasms , Polycomb Repressive Complex 1 , Animals , Humans , Mice , A549 Cells , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/secondary , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , Transcription, Genetic , Cyclin-Dependent Kinase Inhibitor p18/genetics , Carrier Proteins/genetics
4.
Int J Rheum Dis ; 26(3): 464-470, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36575870

ABSTRACT

BACKGROUND: To explore the value of human data from the Zhuang population via predicting the diameter of the hamstring tendon autograft in anterior cruciate ligament (ACL) reconstruction and determining the feasibility of preoperative ultrasound for prediction. METHODS: In total, 24 Zhuang patients who underwent ACL reconstruction with a 4-strand semitendinosus and gracilis tendon autograft (4 S-STG) were enrolled in this study. Before the operation, the affected semitendinosus tendon (ST) was examined by ultrasonography, and its length, diameter, cross-sectional area, and circumference were measured. The patients' basic information and body data, ie, height, weight, body mass index, lower limb length injured, and thigh circumference injured, were recorded. Their ST and gracilis tendon lengths and diameters and 4 S-STG diameter were measured during the operation. A correlation analysis was conducted between the ultrasound measurement results and human data and intraoperative tendon measurements. RESULTS: The ST diameter measured by ultrasound was correlated with the ST length and ST diameter measured during operation, and the ST circumference measured by ultrasound was correlated with the ST diameter measured during operation. The patients' body weight can be used to distinguish a 4 S-STG diameter of ≥8 mm (P < .01, mean difference = 11.59). The area under the receiver operating characteristic curve of body weight was 0.829. The final graft diameter ≥8 mm could be predicted with a body weight of 61.5 kg as the cutoff point; the sensitivity and specificity were 72.2% and 83.3%, respectively. CONCLUSION: In Zhuang patients undergoing ACL reconstruction with 4 S-STG, body weight more accurately predicted graft diameter than preoperative semitendinosus diameter.


Subject(s)
Hamstring Tendons , Humans , Hamstring Tendons/transplantation , Autografts , Transplantation, Autologous , Tendons , Body Weight
5.
Mol Cancer Res ; 20(11): 1611-1622, 2022 11 03.
Article in English | MEDLINE | ID: mdl-35894945

ABSTRACT

Polycomb group proteins are often dysregulated in cancer, leading to disruption of epigenetic landscapes and acquisition of cancer hallmarks. Chromobox 8 (CBX8) is a core component of canonical polycomb repressive complex 1; however, its role in transcriptional regulation and in ovarian carcinoma progression has not been extensively investigated. In this study, we find that CBX8 is upregulated in ovarian cancer. Overexpression and knockdown approaches show that CBX8 facilitates the growth and migration of CAOV3, A2780, and SKOV3 cells in vitro. Consistently, depletion of CBX8 suppresses the growth and metastasis of ovarian carcinoma in vivo. Mechanistically, RNA-sequencing assays together with functional rescue experiments identify a tumor suppressor, SUSD2, as the functional target of CBX8 in ovarian carcinoma cells. Significantly, FLAG affinity coupled with mass spectrometry discovers that CBX8 interacts with a subunit of inhibitor of acetyltransferases (INHAT), SET, which also promotes the growth and migration of A2780 cells. CBX8 and SET cobind to the promoter of SUSD2 to establish H2AK119ub1 and prevent the acetylation of histone H3, resulting in transcriptional suppression of SUSD2. IMPLICATIONS: Our study uncovers a novel mechanism CBX8 explores to execute gene repression, and provides new therapeutic targets for ovarian carcinoma.


Subject(s)
Carcinoma , Liver Neoplasms , Membrane Glycoproteins , Ovarian Neoplasms , Polycomb Repressive Complex 1 , Female , Humans , Cell Line, Tumor , Liver Neoplasms/genetics , Membrane Glycoproteins/genetics , Ovarian Neoplasms/genetics , Polycomb Repressive Complex 1/genetics , Polycomb-Group Proteins/genetics
6.
Front Endocrinol (Lausanne) ; 13: 868105, 2022.
Article in English | MEDLINE | ID: mdl-35528018

ABSTRACT

Objective: This study aimed to investigate the inhibition of human important phase II metabolic enzyme sulfotransferases (SULTs) by phthalate monoesters, which are important metabolites of phthalate esters (PAEs). Method: Recombinant SULT-catalyzed metabolism of p-nitrophenol (PNP) was employed as the probe reactions of SULTs to investigate the inhibition of 8 kinds of phthalate monoesters towards SULT isoforms. An in vitro incubation system was utilized for preliminary screening, and 100 µM of phthalate monoesters was used. Inhibition kinetics were carried out to determine the inhibition of SULTs by phthalate monoesters. Result: Multiple phthalate monoesters have been demonstrated to exert strong inhibition potential towards SULT1A1, SULT1B1, and SULT1E1, and no significant inhibition of phthalate monoesters towards SULT1A3 was found. The activity of SULT1A1 was strongly inhibited by mono-hexyl phthalate (MHP), mono-octyl phthalate (MOP), mono-benzyl phthalate (MBZP), and mono-ethylhexyl phthalate (MEHP). Monobutyl phthalate (MBP), MHP, MOP, mono-cyclohexyl phthalate (MCHP), and MEHP significantly inhibited the activity of SULT1B1. MHP, MOP, and MEHP significantly inhibited the activity of SULT1E1. MOP was chosen as the representative phthalate monoester to determine the inhibition kinetic parameters (Ki) towards SULT1B1 and SULT1E1. The inhibition kinetic parameters (Ki) were calculated to be 2.23 µM for MOP-SULT1B1 and 5.54 µM for MOP-SULT1E1. In silico docking method was utilized to understand the inhibition mechanism of SULT1B1 by phthalate monoesters. Conclusions: All these information will be beneficial for understanding the risk of phthalate monoester exposure from a new perspective.


Subject(s)
Esters , Sulfotransferases , Humans , Phthalic Acids , Protein Isoforms , Sulfotransferases/metabolism
7.
Mol Ther Nucleic Acids ; 27: 670-684, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35070495

ABSTRACT

The disruption of epigenetic regulation is common in tumors; the abnormal expression of epigenetic factors leads to cancer occurrence and development. In this study, to investigate the potential function of histone methylation regulators in lung adenocarcinoma (LUAD), we performed differential expression analysis using RNA-seq data downloaded from The Cancer Genome Atlas (TCGA) database, and identified CBX2 and EZH2 as obviously upregulated histone methylation regulators. CBX2 knockdown significantly inhibited LUAD cell growth and metastasis in vitro and in vivo. The combined high expression of CBX2 and EZH2 was an indicator of poor prognosis in LUAD. The inhibition of both CBX2 and EZH2 exerted cooperative suppressive effects on the growth and metastasis of LUAD cells. Mechanistically, we revealed that CBX2 and EZH2 downregulated several PPAR signaling pathway genes and tumor suppressor genes through binding to their promoter cooperatively or separately. Furthermore, knockdown of CBX2 improved the therapeutic efficiency of EZH2 inhibitor on A549 cells. Our study reveals the cooperative oncogenic role of CBX2 and EZH2 in promoting LUAD progression, thereby providing potential targets for LUAD diagnosis and therapy.

8.
Toxicol Lett ; 358: 40-47, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35063619

ABSTRACT

Myricetin is a dietary flavonol and possesses multiple bioactivities, which making it an excellent nutritional supplement and a new drug candidate. However, whether myricetin and other homologous dietary flavonols affect the activities of UDP-glucuronosyltransferases (UGT) enzymes and facilitated food-drug interactions remains unclear. Our results demonstrated that myricetin displayed broad-spectrum inhibition against human UGTs. Myricetin exhibited strong inhibitory effects against UGT1A1, 1A3, 1A6, 1A7, 1A10 (IC50 < 10 µM) with non-competitive inhibition type, while serving as a moderate inhibitor against UGT1A9 and 2B7 (IC50 range from 25 to 29 µM) with competitive and mixed inhibition type, respectively. In Silico docking was carried out to explore the binding models and free energies of myricetin towards inhibitory UGTs. The potential risks of food-drug interactions after myricetin consumption were predicted by combining the in vitro inhibitory data and physiological data. The quantitative prediction in vivo of inhibition on gastrointestinal UGTs by myricetin showed that the inhibition against UGT1A1, 1A3, 1A6, 1A7, 1A9, 1A10 and 2B7 would likely occur with high risk. The follow-up findings demonstrated that morin, kaempferol, quercetin and galangin, the four homologous dietary flavonols, shared similar inhibition patterns towards UGTs. These findings altogether demonstrate that myricetin and homologous dietary flavonols have potent and broad-spectrum inhibitory effects against most human UGTs, thus suggest that much caution should be exercised when flavonols-rich foods or supplements are co-administered with UGT substrate drugs.


Subject(s)
Flavonols , Microsomes, Liver , Flavonoids , Glucuronosyltransferase/metabolism , Humans , Uridine Diphosphate/metabolism
9.
Biochem Pharmacol ; 192: 114736, 2021 10.
Article in English | MEDLINE | ID: mdl-34411567

ABSTRACT

Reprogramming of energy metabolism is a hallmarkofcancer, and the pentose phosphate pathway (PPP) is a major glucose metabolic pathway important for meeting the cellular demands of biosynthesis and anti-oxidant defense. Our previous study showed that phosphoinositide 3-kinase enhancer-activating Akt (PIKE-A) plays an important role in glioblastoma cell survival and growth under cellular energy stress condition. However, the crucial functions of PIKE-A in cancer energy metabolism are poorly understood.In the present study, we show that PIKE-A promotes DNA biosynthesis, NADPH production and inhibits reactive oxygen species (ROS) production, leading to increasing proliferation and growth of glioblastoma cell and suppressing cellular senescence. Mechanistically, PIKE-A binds to STAT3 and stimulates its phosphorylation mediated by tyrosine kinase Fyn, which enhances transcription of the rate-limitting enzyme glucose-6-phosphate dehydrogenase (G6PD) in the PPP. Finally, targeting PIKE-A-G6PD axis sensitizes glioblastoma to temozolomide (TMZ)treatment. This study reveals that STAT3 is a novel binding partner of PIKE-A which recruits Fyn to phosphorylate STAT3, contributing to the expression of G6PD, leading to promoting tumor growth and suppressing cellular senescence. Thus, the PIKE-A/STAT3/G6PD axis strongly links the PPP to carcinogenesis and may become a promising cancer therapeutic target.


Subject(s)
Cell Proliferation/physiology , GTP-Binding Proteins/biosynthesis , GTPase-Activating Proteins/biosynthesis , Glioblastoma/metabolism , Glucosephosphate Dehydrogenase/biosynthesis , Pentose Phosphate Pathway/physiology , STAT3 Transcription Factor/biosynthesis , Animals , Cell Line, Tumor , Gene Expression Regulation, Enzymologic , Gene Knockout Techniques/methods , Glioblastoma/pathology , Glucosephosphate Dehydrogenase/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Nude , Phosphorylation/physiology
10.
J Exp Med ; 218(9)2021 09 06.
Article in English | MEDLINE | ID: mdl-34236404

ABSTRACT

Obesity-induced secretory disorder of adipose tissue-derived factors is important for cardiac damage. However, whether platelet-derived growth factor-D (PDGF-D), a newly identified adipokine, regulates cardiac remodeling in angiotensin II (AngII)-infused obese mice is unclear. Here, we found obesity induced PDGF-D expression in adipose tissue as well as more severe cardiac remodeling compared with control lean mice after AngII infusion. Adipocyte-specific PDGF-D knockout attenuated hypertensive cardiac remodeling in obese mice. Consistently, adipocyte-specific PDGF-D overexpression transgenic mice (PA-Tg) showed exacerbated cardiac remodeling after AngII infusion without high-fat diet treatment. Mechanistic studies indicated that AngII-stimulated macrophages produce urokinase plasminogen activator (uPA) that activates PDGF-D by splicing full-length PDGF-D into the active PDGF-DD. Moreover, bone marrow-specific uPA knockdown decreased active PDGF-DD levels in the heart and improved cardiac remodeling in HFD hypertensive mice. Together, our data provide for the first time a new interaction pattern between macrophage and adipocyte: that macrophage-derived uPA activates adipocyte-secreted PDGF-D, which finally accelerates AngII-induced cardiac remodeling in obese mice.


Subject(s)
Lymphokines/metabolism , Obesity/physiopathology , Platelet-Derived Growth Factor/metabolism , Urokinase-Type Plasminogen Activator/metabolism , Ventricular Remodeling/physiology , Adipocytes/metabolism , Adipocytes/pathology , Angiotensin II/pharmacology , Animals , Heart/drug effects , Hypertension/genetics , Hypertension/physiopathology , Lymphokines/genetics , Macrophages/metabolism , Macrophages/pathology , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mice, Transgenic , Myocardium/pathology , Obesity/metabolism , Platelet-Derived Growth Factor/genetics , Urokinase-Type Plasminogen Activator/genetics
12.
Oncotarget ; 9(3): 4258-4266, 2018 Jan 09.
Article in English | MEDLINE | ID: mdl-29423120

ABSTRACT

p62, as a scaffolding/adaptor protein, is involved in multiple physiological processes include inflammation, autophagy and mitosis. However, the influence of p62 in cancer patients has not been comprehensively investigated. Moreover, the prognostic value of p62 for the survival of patients with solid tumors remains controversial. In this present meta-analysis, twenty suitable articles were identified from PubMed, EMBASE and Web of Science, Nature databases, including 4271 patients. A random-effect or fixed-effect model was adopted to correlate p62 expression with different outcome measured in entire tumors. Combined with results of hazard ratios (HRs) and 95% confidence intervals (CIs), we concluded that higher expression of p62 is associated with poorer overall survival (OS) (HR: 2.22, 95% CI: 1.82-2.71, P < 0.05), disease-free survival (DFS) (HR = 2.48, 95% CI: 1.78-3.46, P < 0.05) and even certain clinicopathological parameters, such as lymph node metastasis (RR = 1.21, 95% CI: 1.06-1.37) and clinical stages (RR = 1.27, 95% CI: 1.12-1.45), in cancer patients. Consequently, our data showed that p62 might be an effective poor prognostic factor for patients with various solid tumors.

13.
J Biol Chem ; 290(25): 15670-15678, 2015 Jun 19.
Article in English | MEDLINE | ID: mdl-25907557

ABSTRACT

Herpes simplex virus 1 (HSV-1) is the most prevalent human virus and causes global morbidity because the virus is able to infect multiple cell types. Remarkably, HSV infection switches between lytic and latent cycles, where T cells play a critical role. However, the precise way of virus-host interactions is incompletely understood. Here we report that HSV-1 productively infected Jurkat T-cells and inhibited antigen-induced T cell receptor activation. We discovered that HSV-1-encoded Us3 protein interrupted TCR signaling and interleukin-2 production by inactivation of the linker for activation of T cells. This study unveils a mechanism by which HSV-1 intrudes into early events of TCR-mediated cell signaling and may provide novel insights into HSV infection, during which the virus escapes from host immune surveillance.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Membrane Proteins/immunology , Protein Serine-Threonine Kinases/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , TNF Receptor-Associated Factor 6/immunology , Viral Proteins/immunology , Adaptor Proteins, Signal Transducing/genetics , Herpes Simplex/genetics , Herpes Simplex/pathology , Herpesvirus 1, Human/genetics , Humans , Immune Evasion/genetics , Interleukin-2/genetics , Interleukin-2/immunology , Jurkat Cells , Membrane Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Antigen, T-Cell , Signal Transduction/genetics , T-Lymphocytes/pathology , T-Lymphocytes/virology , TNF Receptor-Associated Factor 6/genetics , Viral Proteins/genetics
14.
Biochem Biophys Res Commun ; 456(1): 434-9, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25482447

ABSTRACT

Parthenolide (PTL) is a sesquiterpene lactone isolated from feverfew and exhibits potent antitumor activity against various cancers. Many studies indicate that PTL treatment leads to apoptosis, however, the mechanism has not been defined. Here, we observed that cells underwent autophagy shortly after PTL treatment. Inhibition of autophagy by knocking out autophagy associated gene atg5 blocked PTL-induced apoptosis. Surprisingly, PTL decreased the level of translation initiation factor eIF4E binding protein 1 (4E-BP1) in correlation with autophagy. Ectopic expression or shRNA knockdown of 4E-BP1 further verified the effect of 4E-BP1 on PTL-induced autophagy. Meanwhile, PTL elevated the cellular reactive oxygen species (ROS) which located upstream of the depletion of 4E-BP1, and contributed to the consequent autophagy. This study revealed 4E-BP1 as a trigger for PTL-induced autophagy and may lead to therapeutic strategy to enhance the efficacy of anticancer drugs.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Autophagy/drug effects , Carrier Proteins/metabolism , Phosphoproteins/metabolism , Sesquiterpenes/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Cycle Proteins , Eukaryotic Initiation Factors , Fibroblasts/metabolism , HEK293 Cells , HL-60 Cells , HeLa Cells , Humans , Mice , Phagosomes/metabolism , Phosphorylation/drug effects , Plasmids , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
15.
Sci Rep ; 4: 5346, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24939233

ABSTRACT

Controllable gene expression is always a challenge and of great significance to biomedical research and clinical applications. Recently, various approaches based on extra-engineered light-sensitive proteins have been developed to provide optogenetic actuators for gene expression. Complicated biomedical techniques including exogenous genes engineering, transfection, and material delivery are needed. Here we present an all-optical method to regulate gene expression in targeted cells. Intrinsic or exogenous genes can be activated by a Ca(2+)-sensitive transcription factor nuclear factor of activated T cells (NFAT) driven by a short flash of femtosecond-laser irradiation. When applied to mesenchymal stem cells, expression of a differentiation regulator Osterix can be activated by this method to potentially induce differentiation of them. A laser-induced "Ca(2+)-comb" (LiCCo) by multi-time laser exposure is further developed to enhance gene expression efficiency. This noninvasive method hence provides an encouraging advance of gene expression regulation, with promising potential of applying in cell biology and stem-cell science.


Subject(s)
Cell Differentiation/radiation effects , Gene Expression/radiation effects , Lasers , Mesenchymal Stem Cells/radiation effects , Blotting, Western , Cell Differentiation/genetics , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Mesenchymal Stem Cells/metabolism , Microscopy, Confocal , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Sp7 Transcription Factor , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
16.
Cell Signal ; 26(10): 2202-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24880064

ABSTRACT

Vav proteins are guanine nucleotide exchange factors (GEFs) that activate a group of small G proteins (GTPases). Vav1 is predominantly expressed in hematopoietic cells, whereas Vav2 and Vav3 are ubiquitously distributed in almost all human tissues. All three Vav proteins contain conserved structural motifs and associate with a variety of cellular activities including proliferation, migration, and survival. Previous observation with Jurkat leukemia T cells showed that Vav1 possessed anti-apoptotic activity by enhancing Bcl-2 transcription. However the mechanism has not been unveiled. Here, we explored the effectors of Vav1 in promoting Bcl-2 expression in Jurkat cells and revealed that Rac2-Akt was specifically evoked by the expression of Vav1, but not Vav2 or Vav3. Although all three Vav isoforms existed in Jurkat cells, Rac2 was distinguishably activated by Vav1 and that led to enhanced Bcl-2 expression and cell survival. Akt was modulated downstream of Vav1-Rac2, and the activation of Akt was indispensable in the enhanced transcription of Bcl-2. Intriguingly, neither Vav2 nor Vav3 was able to activate Rac2-Akt pathway as determined by gene silencing approach. Our data illustrated a unique role of Vav1 in T leukemia survival by selectively triggering Rac2-Akt axis and elevating the expression of anti-apoptotic Bcl-2.


Subject(s)
Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-vav/metabolism , rac GTP-Binding Proteins/metabolism , Apoptosis , HEK293 Cells , Humans , Jurkat Cells , Leukemia/metabolism , Leukemia/pathology , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-vav/antagonists & inhibitors , Proto-Oncogene Proteins c-vav/genetics , RNA Interference , RNA, Small Interfering/metabolism , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/genetics , RAC2 GTP-Binding Protein
17.
PLoS One ; 9(6): e99052, 2014.
Article in English | MEDLINE | ID: mdl-24905577

ABSTRACT

Vav1, a guanine nucleotide exchange factor (GEF) for Rho family GTPases, is a hematopoietic protein involved in a variety of cellular events. In recent years, aberrant expression of Vav1 has been reported in non-hematopoietic cancers including human breast cancer. It remains to be answered how Vav1 is expressed and what Vav1 does in its non-resident tissues. In this study, we aimed to explore the mechanism for Vav1 expression in breast cancer cells in correlation with estrogen-ER pathway. We not only verified the ectopic expression of Vav1 in human breast cancer cell lines, but also observed that Vav1 expression was induced by 17ß-estradiol (E2), a typical estrogen receptor (ER) ligand, in ER-positive cell lines. On the other hand, Tamoxifen, a selective estrogen receptor modulator (SERM), and ICI 182,780, an ER antagonist, suppressed the expression of Vav1. The estrogen receptor modulating Vav1 expression was identified to be α form, not ß. Furthermore, treatment of E2 increased the transcription of vav1 gene by enhancing the promoter activity, though there was no recognizable estrogen response element (ERE). Nevertheless, two regions at the vav1 gene promoter were defined to be responsible for E2-induced activation of vav1 promoter. Chromatin immunoprecipitation (ChIP) and co-immunoprecipitation (Co-IP) analyses suggested that ERα might access to the vav1 promoter via interacting with transcription factors, c-Myb and ELF-1. Consequently, the enhanced expression of Vav1 led to the elevation of Cyclin D1 and the progression of cell cycle. The present study implies that estrogen-ER modulates the transcription and expression of Vav1, which may contribute to the proliferation of cancerous cells.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-vav/genetics , Transcriptional Activation , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Female , Humans , Promoter Regions, Genetic , Up-Regulation
18.
Modern Hospital ; (6): 120-121, 2014.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-499580

ABSTRACT

Cultivation and selection of advanced excellent medical models are important links for the hospital cultural construction .Cultivation of appropriate models should focus on selection of models , popularization of mod-els, establishment of platformsin order to make models play a role of demonstrating and guiding and become impor-tant carriers of spreading health core values so as to provide spiritual support for hospital development .

19.
J Biol Chem ; 288(6): 3777-85, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23271736

ABSTRACT

Vav1 is a guanine nucleotide exchange factor (GEF) specifically expressed in hematopoietic cells. It consists of multiple structural domains and plays important roles in T cell activation. The other highly conserved isoforms of Vav family, Vav2 and Vav3, are ubiquitously expressed in human tissues including lymphocytes. All three Vav proteins activate Rho family small GTPases, which are involved in a variety of biological processes during T cell activation. Intensive studies have demonstrated that Vav1 is indispensable for T cell receptor (TCR)-mediated signal transduction, whereas Vav2 and Vav3 function as GEFs that overlap with Vav1 on TCR-induced cytoskeleton reorganization. T cells lacking Vav1 exhibited severe defect in TCR-mediated calcium elevation, indicating that the co-existing Vav2 and Vav3 did not compensate Vav1 in calcium signaling. What is the functional particularity of Vav1 in lymphocytes? In this study, we identified the N-terminal 20 amino acids of Vav1 in the calponin homology (CH) domain to be essential for its interaction with calmodulin (CaM) that leads to TCR-induced calcium mobilization. Substitution of the 1-20 amino acids of Vav1 with those of Vav2 or Vav3 abolished the association with CaM, and the N-terminal mutations of Vav1 failed to potentiate normal TCR-induced calcium mobilization, that in turn, suspended nuclear factor of activated T cells (NFAT) activation and IL-2 production. This study highlights the importance of the N-terminal 20 aa of Vav1 for CaM binding, and provides new insights into the distinguished and irreplaceable role of Vav1 in T cell activation and signal transduction.


Subject(s)
Calcium Signaling/physiology , Lymphocyte Activation/physiology , Proto-Oncogene Proteins c-vav/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Calmodulin/genetics , Calmodulin/metabolism , HeLa Cells , Humans , Interleukin-2/biosynthesis , Interleukin-2/genetics , Mutation , Protein Binding/physiology , Protein Isoforms , Protein Structure, Tertiary , Proto-Oncogene Proteins c-vav/genetics , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/cytology
20.
J Clin Microbiol ; 49(4): 1537-41, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21325551

ABSTRACT

Human bocavirus 2 (HBoV2) is a parvovirus that has been recently identified in stool samples from children. Any association between the virus and clinical disease is unclear. A rapid, reliable diagnostic method is necessary to address this issue. In this study, we developed a sensitive and specific HBoV2 quantitative real-time PCR assay that targets the HBoV2 NP-1 gene, based on the TaqMan method. The assay could reproducibly detect 10 copies of a recombinant DNA plasmid containing a partial region of the HBoV2 genome, with a dynamic range of 8 log units (10(1) to 10(8) copies). A clinical evaluation detected HBoV2 in 85 (24.6%) of 345 children with gastroenteritis, with viral loads ranging from 1.67 × 10(2) to 4.27 × 10(9) copies per ml of stool specimen.


Subject(s)
Clinical Laboratory Techniques/methods , Human bocavirus/isolation & purification , Parvoviridae Infections/diagnosis , Polymerase Chain Reaction/methods , Virology/methods , Child, Preschool , DNA Primers/genetics , DNA, Viral/chemistry , DNA, Viral/genetics , Female , Human bocavirus/classification , Human bocavirus/genetics , Humans , Infant , Male , Molecular Sequence Data , Parvoviridae Infections/virology , Sensitivity and Specificity , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL