Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
J Hematol Oncol ; 17(1): 36, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783389

ABSTRACT

Oncolytic viruses (OVs) offer a novel approach to treat solid tumors; however, their efficacy is frequently suboptimal due to various limiting factors. To address this challenge, we engineered an OV containing targets for neuron-specific microRNA-124 and Granulocyte-macrophage colony-stimulating factor (GM-CSF), significantly enhancing its neuronal safety while minimally compromising its replication capacity. Moreover, we identified PARP1 as an HSV-1 replication restriction factor using genome-wide CRISPR screening. In models of glioblastoma (GBM) and triple-negative breast cancer (TNBC), we showed that the combination of OV and a PARP inhibitor (PARPi) exhibited superior efficacy compared to either monotherapy. Additionally, single-cell RNA sequencing (scRNA-seq) revealed that this combination therapy sensitized TNBC to immune checkpoint blockade, and the incorporation of an immune checkpoint inhibitor (ICI) further increased the survival rate of tumor-bearing mice. The combination of PARPi and ICI synergistically enhanced the ability of OV to establish durable tumor-specific immune responses. Our study effectively overcomes the inherent limitations of OV therapy, providing valuable insights for the clinical treatment of TNBC, GBM, and other malignancies.


Subject(s)
Oncolytic Virotherapy , Oncolytic Virotherapy/methods , Animals , Humans , Mice , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Glioblastoma/therapy , Glioblastoma/genetics , Oncolytic Viruses/genetics , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/genetics , Female , Poly (ADP-Ribose) Polymerase-1/genetics , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Herpesvirus 1, Human/genetics , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , MicroRNAs/genetics , Xenograft Model Antitumor Assays , CRISPR-Cas Systems
2.
Mol Ther ; 31(11): 3163-3175, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37658603

ABSTRACT

In vivo CRISPR gene therapy holds large clinical potential, but the safety and efficacy remain largely unknown. Here, we injected a single dose of herpes simplex virus 1 (HSV-1)-targeting CRISPR formulation in the cornea of three patients with severe refractory herpetic stromal keratitis (HSK) during corneal transplantation. Our study is an investigator-initiated, open-label, single-arm, non-randomized interventional trial at a single center (NCT04560790). We found neither detectable CRISPR-induced off-target cleavages by GUIDE-seq nor systemic adverse events for 18 months on average in all three patients. The HSV-1 remained undetectable during the study. Our preliminary clinical results suggest that in vivo gene editing targeting the HSV-1 genome holds acceptable safety as a potential therapy for HSK.


Subject(s)
Herpesvirus 1, Human , Keratitis, Herpetic , Humans , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , Keratitis, Herpetic/therapy , Keratitis, Herpetic/drug therapy , Cornea , Herpesvirus 1, Human/genetics
4.
Cell Rep ; 32(4): 107974, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32726637

ABSTRACT

Attenuated DNA repair leads to genomic instability and tumorigenesis. BRCA1/BARD1 are the best-known tumor suppressors that promote homology recombination (HR) and arrest cell cycle. However, it remains ambiguous whether and how their E3 ligase activity regulates HR. Here, we demonstrate that upon genotoxic stress, BRCA1 together with BARD1 catalyzes the K48 polyubiquitination on LARP7, a 7SK RNA binding protein known to control RNAPII pausing, and thereby degrades it through the 26S ubiquitin-proteasome pathway. Depleting LARP7 suppresses the expression of CDK1 complex, arrests the cell at the G2/M DNA damage checkpoint, and reduces BRCA2 phosphorylation, which thereby facilitates RAD51 recruitment to damaged DNA to enhance HR. Importantly, LARP7 depletion observed in breast cancer patients leads to chemoradiotherapy resistance both in vitro and in vivo. Altogether, this study unveils a mechanism by which BRCA1/BARD1 control HR and cell cycle, and highlights LARP7 as a potential target for cancer prevention and therapy.


Subject(s)
BRCA1 Protein/genetics , Ribonucleoproteins/metabolism , Tumor Suppressor Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Adult , BRCA1 Protein/metabolism , CDC2 Protein Kinase/metabolism , Carcinogenesis , Cell Cycle , DNA Damage , DNA Repair , Female , Genomic Instability , HeLa Cells , Homologous Recombination/genetics , Humans , Middle Aged , Recombinational DNA Repair/genetics , Ribonucleoproteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
Proc Natl Acad Sci U S A ; 117(25): 14395-14404, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32513696

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) is up-regulated during granulocytic differentiation of acute promyelocytic leukemia (APL) cells induced by all-trans retinoic acid (ATRA). It has been reported that RIG-I recognizes virus-specific 5'-ppp-double-stranded RNA (dsRNA) and activates the type I interferons signaling pathways in innate immunity. However, the functions of RIG-I in hematopoiesis remain unclear, especially regarding its possible interaction with endogenous RNAs and the associated pathways that could contribute to the cellular differentiation and maturation. Herein, we identified a number of RIG-I-binding endogenous RNAs in APL cells following ATRA treatment, including the tripartite motif-containing protein 25 (TRIM25) messenger RNA (mRNA). TRIM25 encodes the protein known as an E3 ligase for ubiquitin/interferon (IFN)-induced 15-kDa protein (ISG15) that is involved in RIG-I-mediated antiviral signaling. We show that RIG-I could bind TRIM25 mRNA via its helicase domain and C-terminal regulatory domain, enhancing the stability of TRIM25 transcripts. RIG-I could increase the transcriptional expression of TRIM25 by caspase recruitment domain (CARD) domain through an IFN-stimulated response element. In addition, RIG-I activated other key genes in the ISGylation pathway by activating signal transducer and activator of transcription 1 (STAT1), including the modifier ISG15 and several enzymes responsible for the conjugation of ISG15 to protein substrates. RIG-I cooperated with STAT1/2 and interferon regulatory factor 1 (IRF1) to promote the activation of the ISGylation pathway. The integrity of ISGylation in ATRA or RIG-I-induced cell differentiation was essential given that knockdown of TRIM25 or ISG15 resulted in significant inhibition of this process. Our results provide insight into the role of the RIG-I-TRIM25-ISGylation axis in myeloid differentiation.


Subject(s)
Cell Differentiation , Cytokines/metabolism , DEAD Box Protein 58/metabolism , Granulocytes/physiology , Transcription Factors/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Cell Line, Tumor , Cytokines/genetics , Gene Knockdown Techniques , HEK293 Cells , Humans , RNA Stability , RNA, Messenger/metabolism , Receptors, Immunologic , Transcription Factors/genetics , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitins/genetics , Up-Regulation
6.
Cell Death Dis ; 11(1): 75, 2020 01 30.
Article in English | MEDLINE | ID: mdl-32001672

ABSTRACT

The bivalent domain (BD) at promoter region is an unique epigenetic feature poised for activation or repression during cell differentiation in embryonic stem cell. However, the function of BDs in already differentiated cells remains exclusive. By profiling the epigenetic landscape of endothelial cells during VEGFA (vascular endothelial growth factor A) stimulation, we discovered that BDs are widespread in endothelial cells and preferentially marked genes responsive to VEGFA. The BDs responsive to VEGFA have more permissive chromatin environment comparing to other BDs. The initial activation of bivalent genes depends on RNAPII pausing release induced by EZH1 rather than removal of H3K27me3. The later suppression of bivalent gene expression depended on KDM5A recruitment by its interaction with PRC2. Importantly, EZH1 promoted both in vitro and in vivo angiogenesis by upregulating EGR3, whereas KDM5A dampened angiogenesis. Collectively, this study demonstrates a novel dual function of BDs in endothelial cells to control VEGF responsiveness and angiogenesis.


Subject(s)
Endothelial Cells/metabolism , Histones/metabolism , Neovascularization, Physiologic/genetics , Promoter Regions, Genetic/genetics , Vascular Endothelial Growth Factor A/pharmacology , Animals , Cell Movement/drug effects , Cell Movement/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation Sequencing , Early Growth Response Protein 3/metabolism , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endothelial Cells/drug effects , Epigenesis, Genetic , Human Umbilical Vein Endothelial Cells , Humans , Mice , Polycomb Repressive Complex 2/metabolism , Protein Domains/genetics , RNA Polymerase II/metabolism , RNA, Small Interfering , RNA-Seq , Retinoblastoma-Binding Protein 2/metabolism , Up-Regulation
7.
Genome Res ; 29(2): 193-207, 2019 02.
Article in English | MEDLINE | ID: mdl-30670628

ABSTRACT

Cell behaviors are dictated by epigenetic and transcriptional programs. Little is known about how extracellular stimuli modulate these programs to reshape gene expression and control cell behavioral responses. Here, we interrogated the epigenetic and transcriptional response of endothelial cells to VEGFA treatment and found rapid chromatin changes that mediate broad transcriptomic alterations. VEGFA-responsive genes were associated with active promoters, but changes in promoter histone marks were not tightly linked to gene expression changes. VEGFA altered transcription factor occupancy and the distal epigenetic landscape, which profoundly contributed to VEGFA-dependent changes in gene expression. Integration of gene expression, dynamic enhancer, and transcription factor occupancy changes induced by VEGFA yielded a VEGFA-regulated transcriptional regulatory network, which revealed that the small MAF transcription factors are master regulators of the VEGFA transcriptional program and angiogenesis. Collectively these results revealed that extracellular stimuli rapidly reconfigure the chromatin landscape to coordinately regulate biological responses.


Subject(s)
Epigenesis, Genetic , Neovascularization, Physiologic/genetics , Transcription, Genetic , Vascular Endothelial Growth Factor A/physiology , Animals , Cells, Cultured , Chromatin/metabolism , Enhancer Elements, Genetic , Humans , Maf Transcription Factors/metabolism , Male , Mice , Mice, Nude , Promoter Regions, Genetic , Transcription Factors/metabolism
8.
Cell Death Dis ; 8(4): e2744, 2017 04 13.
Article in English | MEDLINE | ID: mdl-28406475

ABSTRACT

Polycomb repressive complex 2 (PRC2), a H3K27me3 methyltransferase complex, promotes the development of many organs by silencing ectopic transcription program. However, currently little is known about the role of PRC2 in blood and vascular development. In this study, we interrogated the function of embryonic ectoderm development (EED), a core PRC2 component, in both endothelial and hematopoietic tissues by inactivating a floxed murine EED allele with Tie2Cre, which catalyzes recombination in endothelial and hematopoietic lineages. Murine EEDfl/fl;Tie2Cre (EEDCKO) embryos died at embryonic day (E) 13.5. We did not observe structural abnormalities of blood vessels or cardiac valves, suggesting that EED is dispensable in endothelial cells for initial steps of vascular development. EEDCKO embryos were pale and had abnormal livers. Flow cytometry of fetal liver cells showed that EED depletion significantly impeded erythroid maturation. There was a corresponding increase in myeloid progenitors and granulocytes and macrophages, suggesting an attenuated differentiation path in myeloid lineages. Moreover, EED depletion impaired the generation of hematopoietic stem cells. Collectively, our study demonstrates that within Tie2Cre-recombined embryonic cells, EED is required for proper erythropoiesis and for formation of hematopoietic progenitor and stem cells, but is dispensable for endothelial lineage commitment and early vascular patterning.


Subject(s)
Erythroid Cells/metabolism , Erythropoiesis/physiology , Fetus/metabolism , Hematopoietic Stem Cells/metabolism , Polycomb Repressive Complex 2/metabolism , Animals , Endothelial Cells/metabolism , Female , Gene Deletion , Male , Mice , Mice, Transgenic , Neovascularization, Physiologic/physiology , Polycomb Repressive Complex 2/genetics
9.
Food Chem ; 194: 20-5, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26471522

ABSTRACT

For transferring the event-specific PCR methods of genetically modified papaya Huanong No.1 to other laboratories, we validated the previous developed PCR assays of Huanong No.1 according to the international standard organization (ISO) guidelines. A total of 11 laboratories participated and returned their test results in this trial. In qualitative PCR assay, the high specificity and limit of detection as low as 0.1% was confirmed. For the quantitative PCR assay, the limit of quantification was as low as 25 copies. The quantitative biases among ten blind samples were within the range between 0.21% and 10.04%. Furthermore, the measurement uncertainty of the quantitative PCR results was calculated within the range between 0.28% and 2.92% for these ten samples. All results demonstrated that the Huanong No.1 qualitative and quantitative PCR assays were creditable and applicable for identification and quantification of GM papaya Huanong No.1 in further routine lab analysis.


Subject(s)
Carica/chemistry , DNA, Plant/analysis , Plants, Genetically Modified/chemistry , Real-Time Polymerase Chain Reaction/methods , DNA, Plant/chemistry , Plants, Genetically Modified/genetics
10.
Proc Natl Acad Sci U S A ; 112(49): 15084-9, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26598702

ABSTRACT

Arsenic is highly effective for treating acute promyelocytic leukemia (APL) and has shown significant promise against many other tumors. However, although its mechanistic effects in APL are established, its broader anticancer mode of action is not understood. In this study, using a human proteome microarray, we identified 360 proteins that specifically bind arsenic. Among the most highly enriched proteins in this set are those in the glycolysis pathway, including the rate-limiting enzyme in glycolysis, hexokinase-1. Detailed biochemical and metabolomics analyses of the highly homologous hexokinase-2 (HK2), which is overexpressed in many cancers, revealed significant inhibition by arsenic. Furthermore, overexpression of HK2 rescued cells from arsenic-induced apoptosis. Our results thus strongly implicate glycolysis, and HK2 in particular, as a key target of arsenic. Moreover, the arsenic-binding proteins identified in this work are expected to serve as a valuable resource for the development of synergistic antitumor therapeutic strategies.


Subject(s)
Arsenic/pharmacology , Carrier Proteins/analysis , Hexokinase/antagonists & inhibitors , Amino Acid Sequence , Apoptosis/drug effects , Arsenic/metabolism , Arsenic Trioxide , Arsenicals/pharmacology , Carrier Proteins/metabolism , Computational Biology , Glycolysis , Humans , Metabolomics , Molecular Sequence Data , Oxides/pharmacology , Proteome
11.
Cell Cycle ; 14(5): 761-71, 2015.
Article in English | MEDLINE | ID: mdl-25590866

ABSTRACT

Cordycepin, an adenosine analog derived from Cordyceps militaris has been shown to exert anti-tumor activity in many ways. However, the mechanisms by which cordycepin contributes to the anti-tumor still obscure. Here our present work showed that cordycepin inhibits cell growth in NB-4 and U937 cells by inducing apoptosis. Further study showed that cordycepin increases the expression of p53 which promotes the release of cytochrome c from mitochondria to the cytosol. The released cytochrome c can then activate caspase-9 and trigger intrinsic apoptosis. Cordycepin also blocks MAPK pathway by inhibiting the phosphorylation of ERK1/2, and thus sensitizes the apoptosis. In addition, our results showed that cordycepin inhibits the expression of cyclin A2, cyclin E, and CDK2, which leads to the accumulation of cells in S-phase. Moreover, our study showed that cordycepin induces DNA damage and causes degradation of Cdc25A, suggesting that cordycepin-induced S-phase arrest involves activation of Chk2-Cdc25A pathway. In conclusion, cordycepin-induced DNA damage initiates cell cycle arrest and apoptosis which leads to the growth inhibition of NB-4 and U937 cells.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , DNA Damage , Deoxyadenosines/pharmacology , Leukemia/pathology , Tumor Suppressor Protein p53/metabolism , Up-Regulation/drug effects , Caspases/metabolism , Cell Line, Tumor , Deoxyadenosines/chemistry , Humans , Leukemia/metabolism , MAP Kinase Signaling System/drug effects , Models, Biological
12.
Mol Cell Proteomics ; 12(10): 2804-19, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23824909

ABSTRACT

Bcl2-associated athanogene 3 (BAG3), a member of the BAG family of co-chaperones, plays a critical role in regulating apoptosis, development, cell motility, autophagy, and tumor metastasis and in mediating cell adaptive responses to stressful stimuli. BAG3 carries a BAG domain, a WW domain, and a proline-rich repeat (PXXP), all of which mediate binding to different partners. To elucidate BAG3's interaction network at the molecular level, we employed quantitative immunoprecipitation combined with knockdown and human proteome microarrays to comprehensively profile the BAG3 interactome in humans. We identified a total of 382 BAG3-interacting proteins with diverse functions, including transferase activity, nucleic acid binding, transcription factors, proteases, and chaperones, suggesting that BAG3 is a critical regulator of diverse cellular functions. In addition, we characterized interactions between BAG3 and some of its newly identified partners in greater detail. In particular, bioinformatic analysis revealed that the BAG3 interactome is strongly enriched in proteins functioning within the proteasome-ubiquitination process and that compose the proteasome complex itself, suggesting that a critical biological function of BAG3 is associated with the proteasome. Functional studies demonstrated that BAG3 indeed interacts with the proteasome and modulates its activity, sustaining cell survival and underlying resistance to therapy through the down-modulation of apoptosis. Taken as a whole, this study expands our knowledge of the BAG3 interactome, provides a valuable resource for understanding how BAG3 affects different cellular functions, and demonstrates that biologically relevant data can be harvested using this kind of integrated approach.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Cell Line, Tumor , Humans , Protein Array Analysis , Protein Interaction Mapping , Proteome
13.
Environ Sci Pollut Res Int ; 20(9): 5913-21, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23263754

ABSTRACT

Phosphorus-bearing materials have been widely applied in immobilization of heavy metals in contaminated soils. However, the study on the stability of the initially P-induced immobilized metals in the contaminated soils is far limited. This work was conducted to evaluate the mobility of Pb, Cu, and Zn in two contrasting contaminated soils amended with phosphate rock tailing (PR) and triple superphosphate fertilizer (TSP), and their combination (P + T) under simulated landfill and rainfall conditions. The main objective was to determine the stability of heavy metals in the P-treated contaminated soils in response to the changing environment conditions. The soils were amended with the P-bearing materials at a 2:1 molar ratio of P to metals. After equilibrated for 2 weeks, the soils were evaluated with the leaching procedures. The batch-based toxicity characteristic leaching procedure (TCLP) was conducted to determine the leachability of heavy metals from both untreated and P-treated soils under simulated landfill condition. The column-based synthetic precipitation leaching procedure (SPLP) were undertaken to measure the downward migration of metals from untreated and P-treated soils under simulated rainfall condition. Leachability of Pb, Cu, and Zn in the TCLP extract followed the order of Zn > Cu > Pb in both soils, with the organic-C- and clay-poor soil showing higher metal leachability than the organic-C- and clay-rich soil. All three P treatments reduced leachability of Pb, Cu, and Zn by up to 89.2, 24.4, and 34.3 %, respectively, compared to the untreated soil, and TSP revealed more effectiveness followed by P + T and then PR. The column experiments showed that Zn had the highest downward migration upon 10 pore volumes of SPLP leaching, followed by Pb and then Cu in both soils. However, migration of Pb and Zn to subsoil and leachate were inhibited in the P-treated soil, while Cu in the leachate was enhanced by P treatment in the organic-C-rich soil. More than 73 % P in the amendments remained in the upper 0-10 cm soil layers. However, leaching of P from soluble TSP was significant with 24.3 % of P migrated in the leachate in the organic-C-poor soil. The mobility of heavy metals in the P-treated soil varies with nature of P sources, heavy metals, and soils. Caution should be taken on the multi-metal stabilization since the P amendment may immobilize some metals while promoting others' mobility. Also, attention should be paid to the high leaching of P from soluble P amendments since it may pose the risk of excessive P-induced eutrophication.


Subject(s)
Copper/chemistry , Lead/chemistry , Phosphorus/chemistry , Soil Pollutants/chemistry , Soil/chemistry , Zinc/chemistry , Hydrogen-Ion Concentration , Rain , Water/chemistry
14.
Acta Biochim Biophys Sin (Shanghai) ; 45(1): 27-35, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23212076

ABSTRACT

Cancer cells have distinct metabolism that highly depends on glycolysis instead of mitochondrial oxidative phosphorylation alone, known as aerobic glycolysis. Pyruvate kinase (PK), which catalyzes the final step of glycolysis, has emerged as a potential regulator of this metabolic phenotype. Expression of PK type M2 (PKM2) is increased and facilitates lactate production in cancer cells, which determines whether the glucose carbons are degraded to pyruvate and lactate or are channeled into synthetic processes. Modulation of PKM2 catalytic activity also regulates the synthesis of DNA and lipids that are required for cell proliferation. However, the mechanisms by which PKM2 coordinates high-energy requirements with high anabolic activities to support cancer cell proliferation are still not completely understood. This review summarizes the biological characteristics of PKM2 and discusses the dual role in cancer metabolism as well as the potential therapeutic applications. Given its pleiotropic effects on cancer biology, PKM2 represents an attractive target for cancer therapy.


Subject(s)
Carrier Proteins/physiology , Membrane Proteins/physiology , Neoplasms/metabolism , Thyroid Hormones/physiology , Humans , Phosphorylation , Thyroid Hormone-Binding Proteins
15.
PLoS One ; 7(11): e49080, 2012.
Article in English | MEDLINE | ID: mdl-23145077

ABSTRACT

Glycosylation is one of the most abundant protein posttranslational modifications. Protein glycosylation plays important roles not only in eukaryotes but also in prokaryotes. To further understand the roles of protein glycosylation in prokaryotes, we developed a lectin binding assay to screen glycoproteins on an Escherichia coli proteome microarray containing 4,256 affinity-purified E.coli proteins. Twenty-three E.coli proteins that bound Wheat-Germ Agglutinin (WGA) were identified. PANTHER protein classification analysis showed that these glycoprotein candidates were highly enriched in metabolic process and catalytic activity classes. One sub-network centered on deoxyribonuclease I (sbcB) was identified. Bioinformatics analysis suggests that prokaryotic protein glycosylation may play roles in nucleotide and nucleic acid metabolism. Fifteen of the 23 glycoprotein candidates were validated by lectin (WGA) staining, thereby increasing the number of validated E. coli glycoproteins from 3 to 18. By cataloguing glycoproteins in E.coli, our study greatly extends our understanding of protein glycosylation in prokaryotes.


Subject(s)
Escherichia coli , Glycoproteins , Glycosylation , Proteome , Deoxyribonuclease I/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Glycoproteins/classification , Glycoproteins/isolation & purification , Lectins/chemistry , Lectins/metabolism , Protein Array Analysis , Wheat Germ Agglutinins/chemistry , Wheat Germ Agglutinins/metabolism
16.
J Immunol ; 181(3): 2155-64, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18641354

ABSTRACT

Interstitial cell migration through extracellular matrix is a hallmark of the inflammation response, tumor invasion, and metastasis. We have established a stable zebrafish transgenic line expressing enhanced GFP under the lysozyme C promoter for visualizing and measuring primitive macrophage migration in vivo. We show that tissue-resident primitive macrophages migrate rapidly through extracellular matrix to the site of acute injury induced by tail transection. Mechanistically, the specific inhibition of JNK, but not p38 and ERK, dramatically abolished the chemotactic migration in a dose-dependent manner, suppressing the trauma-induced recruitment of phosphorylated C-Jun transcription factor to proximal AP-1 sites in the promoter of matrix metalloproteinase 13 (mmp13), a gene specifically expressed in primitive macrophages during embryogenesis and required for the interstitial migration. Furthermore, dexamethasone suppressed the trauma-induced JNK phosphorylation and macrophage migration accompanied by simultaneous up-regulation of mkp-1, a well-known phosphatase capable of inactivating phosphorylated JNK. The results indicate that the JNK-Mmp13 signaling pathway plays an essential role in regulating the innate immune cell migration in response to severe injury in vivo.


Subject(s)
Intestines/cytology , Intestines/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Macrophages/cytology , Macrophages/enzymology , Matrix Metalloproteinase 13/metabolism , Signal Transduction , Acute Disease , Animals , Animals, Genetically Modified , Cell Movement/drug effects , Dual Specificity Phosphatase 1/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucocorticoids/pharmacology , Intestines/embryology , Intestines/injuries , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Macrophages/drug effects , Matrix Metalloproteinase 13/genetics , Molecular Structure , Promoter Regions, Genetic/genetics , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Transcriptional Activation/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
J Biol Chem ; 283(25): 17652-61, 2008 Jun 20.
Article in English | MEDLINE | ID: mdl-18413315

ABSTRACT

The EEN (extra eleven nineteen) gene is one of the fusion partners of mixed-lineage leukemia, located on chromosome 19p13. Here we cloned two een genes (designated as eena and eenb) in zebrafish, which are assigned to linkage groups 8 and 2, respectively. Whole-mount in situ hybridization assay showed that eena and eenb have overlapping but distinct expression patterns during embryogenesis. Ubiquitous or targeted overexpression of eena, but not eenb, into wild-type or transgenic embryos (green fluorescent protein-labeled myeloid progenitors) induced a significant proliferation and ectopic distribution of myeloid progenitors in the yolk sac. Using a morpholino antisense gene knockdown approach, we showed that the number of myeloid progenitors and their downstream mature myelomonocytic cells was significantly decreased in the eena- deficient embryos. Mechanistically, overexpression of eena selectively stimulated ERK phosphorylation and increased the level of transcription factor c-Fos in vitro and in vivo, whereas eena lacking the Src homology 3 domain completely abolished these effects. Furthermore, a MAPK/ERK kinase (MEK) inhibitor, PD98059, blocked the eena-induced cell proliferation and activation of ERK signaling. The results suggest that eena plays an important role in the development of the myeloid cell through activation of the ERK pathway and may provide a valuable reference for future studies of the role of EEN in leukemogenesis.


Subject(s)
Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myeloid Cells/cytology , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Nucleus/metabolism , Cell Proliferation , Enzyme Inhibitors/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Models, Biological , NIH 3T3 Cells , Phosphorylation , Zebrafish , Zebrafish Proteins/physiology
18.
Blood ; 109(2): 769-77, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-16990610

ABSTRACT

The EEN (extra eleven nineteen) gene, located on chromosome 19p13, was cloned as a fusion with MLL from a patient with acute myeloid leukemia (AML) with translocation t(11;19)(q23;p13). In this study, we characterized the genomic structure of the EEN gene, including its 5' regulatory region and transcription start site (TSS). We found that Sp1 could bind to the guanine-cytosine (GC)-stretch of the EEN promoter and was critical for the normal EEN expression, whereas the leukemia-associated fusion protein AML1-ETO could aberrantly transactivate the EEN gene through an AML1 binding site. Of note, overexpressed EEN showed oncogenic properties, such as transforming potential in NIH3T3 cells, stimulating cell proliferation, and increasing the activity of transcriptional factor AP-1. Retroviral transduction of EEN increased self-renewal and proliferation of murine hematopoietic progenitor cells. Moreover, Kasumi-1 and HL60-cell growth was inhibited with down-regulation of EEN by RNAi. These findings demonstrate that EEN might be a common target in 2 major types of AML associated with MLL or AML1 translocations, and overexpression of EEN may play an essential role in leukemogenesis.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Gene Expression Regulation, Leukemic , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Myeloid/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Oncogene Proteins, Fusion/genetics , Transcription, Genetic/genetics , Acute Disease , Animals , Cell Transformation, Neoplastic , HL-60 Cells , Humans , K562 Cells , Mammary Neoplasms, Animal , Mice , Mice, Nude , Neoplasm Transplantation , RUNX1 Translocation Partner 1 Protein , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity , Transcription Factors/metabolism , U937 Cells
19.
Int J Mol Med ; 15(4): 617-26, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15754023

ABSTRACT

Mucin1 (MUC1) promoter has been cloned from the 5' flanking region of the MUC1 gene in breast carcinoma, functionally characterized and applied in gene therapy of breast and esophageal carcinoma. In the present study, we amplified a 786 base pair (bp) MUC1 promoter by two-step nest PCR, and identified the activity and tumor-specificity using an enhanced green fluorescent protein (EGFP) gene as a reporter gene by fluorescence microscopy and flow cytometry analysis in Panc-1, primary normal pancreatic (PNPC), and cervical cancer HeLa cell lines. Subsequently, the human somatostatin receptor subtype 2 (hSSTR2) gene driven by MUC1 promoter was cloned into the pAdTrack to produce recombinant adenovirus AdMUC1-hSSTR2. The anticancer effect of AdMUC1-hSSTR2 was determined in Panc-1. The results demonstrated that there was no AdMUC1-hSSTR2-induced apoptosis, but a significant cell proliferation inhibition even without somatostatin (SST) analogue Octreotide, involved in the up-regulation of the cyclin-dependent kinase (CDK) inhibitors p21 and p27. Moreover, the anticancer effect could not be augmented by the addition of Octreotide, revealing a mechanism that was independent from induction of Octreotide. Therefore, this adenovirus system can be used as a novel, potent and specific tool for gene-targeting therapy in the MUC1 positive pancreatic carcinoma as shown in Panc-1.


Subject(s)
Antigens/genetics , Genetic Therapy , Glycoproteins/genetics , Pancreatic Neoplasms/therapy , Receptors, Somatostatin/genetics , Adenoviridae , Antigens, Neoplasm , Base Sequence , Cytomegalovirus , Genetic Vectors , HeLa Cells , Humans , Molecular Sequence Data , Mucin-1 , Mucins , Pancreatic Neoplasms/genetics , Promoter Regions, Genetic , Receptors, Somatostatin/metabolism
20.
World J Gastroenterol ; 11(6): 831-8, 2005 Feb 14.
Article in English | MEDLINE | ID: mdl-15682475

ABSTRACT

AIM: To investigate the in vitro antitumor effect of adenovirus-mediated small interfering RNAs (siRNAs) on pancreatic cancer and the associated mechanism. METHODS: A 63-nucleotide (nt) oligonucleotide encoding K-ras(val12) and specific siRNA were introduced into pSilencer 3.1-H1, then the H1-RNA promoter and siRNA coding insert were subcloned into pAdTrack to get plasmid pAdTrackH1-K-ras(val12). After homologous recombination in bacteria and transfections of such plasmids into a mammalian packaging cell line 293, siRNA expressing adenovirus AdH1-K-ras(val12) was obtained. Stable suppression of K-ras(val12) was detected by Northern blot and Western blot. Apoptosis in Panc-1 cells was detected by flow cytometry. RESULTS: We obtained adenovirus AdH1-K-ras(val12) carrying the pSilencer 3.1-H1 cassette, which could mediate gene silencing. Through siRNA targeted K-ras(val12), the oncogenic phenotype of cancer cells was reversed. Flow cytometry showed that apoptotic index of Panc-1 cells was significantly higher in the AdH1-K-ras(val12)-treatment group (18.70% at 72 h post-infection, 49.55% at 96 h post-infection) compared to the control groups (3.47%, 3.98% at 72 and 96 h post-infection of AdH1-empty, respectively; 4.21%, 3.78% at 72 and 96 h post-infection of AdH1-p53, respectively) (P<0.05). CONCLUSION: These results demonstrate that adenoviral vectors can be used to mediate RNA interference (RNAi) to induce persistent loss of functional phenotypes. In gene therapy, the selective down-regulation of only the mutant version of a gene allows for highly specific effects on tumor cells, while leaving the normal cells untouched. In addition, the apoptosis of pancreatic cancer cell line Panc-1 can be induced after AdH1-K-ras(val12) infection. This kind of adenovirus based on RNAi might be a promising vector for cancer therapy.


Subject(s)
Gene Silencing , Genes, ras/genetics , Genetic Therapy/methods , Pancreatic Neoplasms/genetics , RNA, Small Interfering/genetics , Adenoviridae/genetics , Base Sequence , Flow Cytometry , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , In Vitro Techniques , Molecular Sequence Data , Phenotype , Plasmids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...