Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Cell Infect Microbiol ; 13: 1294801, 2023.
Article in English | MEDLINE | ID: mdl-38089817

ABSTRACT

Introduction: The binary PirA/PirB toxin expressed by Vibrio parahaemolyticus (PirABVp) is a virulent complex that causes acute hepatopancreatic necrosis disease (AHPND) in shrimps, affecting the global shrimp farming industry. AHPND is currently diagnosed by detecting pirA and pirB genes by PCR; however, several V. parahaemolyticus strains do not produce the two toxins as proteins. Thus, an immunoassay using antibodies may be the most effective tool for detecting toxin molecules. In this study, we report a sandwich ELISA-based immunoassay for the detection of PirABVp. Methods: We utilized a single-chain variable fragment (scFv) antibody library to select scFvs against the PirA or PirB subunits. Phage display panning rounds were conducted to screen and identify scFv antibodies directed against each recombinant toxin subunit. Selected scFvs were converted into IgGs to develop a sandwich immunoassay to detect recombinant and bacterial PirABVp. Results: Antibodies produced as IgG forms showed sub-nanomolar to nanomolar affinities (KD), and a pair of anti-PirA antibody as a capture and anti-PirB antibody as a detector showed a limit of detection of 201.7 ng/mL for recombinant PirABVp. The developed immunoassay detected PirABVp in the protein lysates of AHPND-causing V. parahaemolyticus (VpAHPND) and showed a significant detectability in moribund or dead shrimp infected with a VpAHPND virulent strain compared to that in non-infected shrimp. Discussion: These results indicate that the developed immunoassay is a reliable method for diagnosing AHPND by detecting PirABVp at the protein level and could be further utilized to accurately determine the virulence of extant or newly identified VpAHPND in the global shrimp culture industry.


Subject(s)
Penaeidae , Toxins, Biological , Vibrio parahaemolyticus , Animals , Vibrio parahaemolyticus/genetics , Penaeidae/microbiology , Enzyme-Linked Immunosorbent Assay , Acute Disease , Necrosis
2.
Cardiovasc Res ; 119(5): 1265-1278, 2023 05 22.
Article in English | MEDLINE | ID: mdl-36534975

ABSTRACT

AIMS: The nuclear factor-κB (NF-κB) signalling pathway plays a critical role in the pathogenesis of multiple vascular diseases. However, in endothelial cells (ECs), the molecular mechanisms responsible for the negative regulation of the NF-κB pathway are poorly understood. In this study, we investigated a novel role for protein tyrosine phosphatase type IVA1 (PTP4A1) in NF-κB signalling in ECs. METHODS AND RESULTS: In human tissues, human umbilical artery ECs, and mouse models for loss of function and gain of function of PTP4A1, we conducted histological analysis, immunostaining, laser-captured microdissection assay, lentiviral infection, small interfering RNA transfection, quantitative real-time PCR and reverse transcription-PCR, as well as luciferase reporter gene and chromatin immunoprecipitation assays. Short hairpin RNA-mediated knockdown of PTP4A1 and overexpression of PTP4A1 in ECs indicated that PTP4A1 is critical for inhibiting the expression of cell adhesion molecules (CAMs). PTP4A1 increased the transcriptional activity of upstream stimulatory factor 1 (USF1) by dephosphorylating its S309 residue and subsequently inducing the transcription of tumour necrosis factor-alpha-induced protein 3 (TNFAIP3/A20) and the inhibition of NF-κB activity. Studies on Ptp4a1 knockout or transgenic mice demonstrated that PTP4A1 potently regulates the interleukin 1ß-induced expression of CAMs in vivo. In addition, we verified that PTP4A1 deficiency in apolipoprotein E knockout mice exacerbated high-fat high-cholesterol diet-induced atherogenesis with upregulated expression of CAMs. CONCLUSION: Our data indicate that PTP4A1 is a novel negative regulator of vascular inflammation by inducing USF1/A20 axis-mediated NF-κB inactivation. Therefore, the expression and/or activation of PTP4A1 in ECs might be useful for the treatment of vascular inflammatory diseases.


Subject(s)
Endothelial Cells , NF-kappa B , Vasculitis , Animals , Humans , Mice , Cell Cycle Proteins/metabolism , Endothelial Cells/metabolism , Inflammation/genetics , Inflammation/metabolism , Membrane Proteins/metabolism , NF-kappa B/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Upstream Stimulatory Factors/metabolism , Vasculitis/genetics , Vasculitis/metabolism
3.
Biosensors (Basel) ; 12(10)2022 Sep 24.
Article in English | MEDLINE | ID: mdl-36290925

ABSTRACT

Staphylococcal enterotoxin B (SEB) is a potent bacterial toxin that causes inflammatory stimulation and toxic shock, thus it is necessary to detect SEB in food and environmental samples. Here, we developed a sensitive immunodetection system using monoclonal antibodies (mAbs). Our study is the first to employ a baculovirus expression vector system (BEVS) to produce recombinant wild-type SEB. BEVS facilitated high-quantity and pure SEB production from suspension-cultured insect cells, and the SEB produced was characterized by mass spectrometry analysis. The SEB was stable at 4 °C for at least 2 years, maintaining its purity, and was further utilized for mouse immunization to generate mAbs. An optimal pair of mAbs non-competitive to SEB was selected for sandwich enzyme-linked immunosorbent assay-based immunodetection. The limit of detection of the immunodetection method was 0.38 ng/mL. Moreover, it displayed higher sensitivity in detecting SEB than commercially available immunodetection kits and retained detectability in various matrices and S. aureus culture supernatants. Thus, the results indicate that BEVS is useful for producing pure recombinant SEB with its natural immunogenic property in high yield, and that the developed immunodetection assay is reliable and sensitive for routine identification of SEB in various samples, including foods.


Subject(s)
Bacterial Toxins , Staphylococcus aureus , Mice , Animals , Baculoviridae , Enterotoxins/analysis , Enzyme-Linked Immunosorbent Assay/methods , Antibodies, Monoclonal
4.
BMB Rep ; 55(3): 142-147, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34674794

ABSTRACT

Human pluripotent stem cells (PSCs) have been utilized as a promising source in regenerative medicine. However, the risk of teratoma formation that comes with residual undifferentiated PSCs in differentiated cell populations is most concerning in the clinical use of PSC derivatives. Here, we report that a monoclonal antibody (mAb) targeting PSCs could distinguish undifferentiated PSCs, with potential teratoma-forming activity, from differentiated PSC progeny. A panel of hybridomas generated from mouse immunization with H9 human embryonic stem cells (hESCs) was screened for ESC-specific binding using flow cytometry. A novel mAb, K312, was selected considering its high stem cell-binding activity, and this mAb could bind to several human induced pluripotent stem cells and PSC lines. Cell-binding activity of K312 was markedly decreased as hESCs were differentiated into embryoid bodies or by retinoic acid treatment. In addition, a cell population negatively isolated from undifferentiated or differentiated H9 hESCs via K312 targeting showed a significantly reduced expression of pluripotency markers, including Oct4 and Nanog. Furthermore, K312-based depletion of pluripotent cells from differentiated PSC progeny completely prevented teratoma formation. Therefore, our findings suggest that K312 is utilizable in improving stem cell transplantation safety by specifically distinguishing residual undifferentiated PSCs. [BMB Reports 2022; 55(3): 142-147].


Subject(s)
Induced Pluripotent Stem Cells , Pluripotent Stem Cells , Teratoma , Animals , Antibodies, Monoclonal/metabolism , Cell Differentiation , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Pluripotent Stem Cells/metabolism
5.
Cell Death Differ ; 28(3): 968-984, 2021 03.
Article in English | MEDLINE | ID: mdl-32989241

ABSTRACT

Gallbladder carcinoma (GBC) exhibits poor prognosis due to local recurrence, metastasis, and resistance to targeted therapies. Using clinicopathological analyses of GBC patients along with molecular in vitro and tumor in vivo analysis of GBC cells, we showed that reduction of Dsg2 expression was highly associated with higher T stage, more perineural, and lymphatic invasion. Dsg2-depleted GBC cells exhibited significantly enhanced proliferation, migration, and invasiveness in vitro and tumor growth and metastasis in vivo through Src-mediated signaling activation. Interestingly, Dsg2 binding inhibited Src activation, whereas its loss activated cSrc-mediated EGFR plasma membrane clearance and cytoplasmic localization, which was associated with acquired EGFR-targeted therapy resistance and decreased overall survival. Inhibition of Src activity by dasatinib enhanced therapeutic response to anti-EGFR therapy. Dsg2 status can help stratify predicted patient response to anti-EGFR therapy and Src inhibition could be a promising strategy to improve the clinical efficacy of EGFR-targeted therapy.


Subject(s)
Carcinoma/drug therapy , Desmoglein 2/metabolism , Drug Resistance, Neoplasm/genetics , Gallbladder Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , src-Family Kinases/metabolism , Animals , Carcinoma/enzymology , Carcinoma/genetics , Carcinoma/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Desmoglein 2/genetics , Disease Progression , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gallbladder Neoplasms/enzymology , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Signal Transduction , Xenograft Model Antitumor Assays , src-Family Kinases/genetics
6.
Biomaterials ; 259: 120265, 2020 11.
Article in English | MEDLINE | ID: mdl-32827795

ABSTRACT

The self-renewal properties of human pluripotent stem cells (hPSCs) contribute to their efficacy in tissue regeneration applications yet increase the likelihood of teratoma formation, thereby limiting their clinical utility. To address this issue, we developed a tool to specifically target and neutralize undifferentiated hPSCs, thereby minimizing tumorigenicity risk without negatively affecting regenerated and somatic tissues. Specifically, we conjugated a monoclonal antibody (K6-1) previously generated in our laboratory against desmoglein 2 (Dsg2), which is highly differentially expressed in undifferentiated hPSCs versus somatic tissues, to the chemotherapeutic agent doxorubicin (DOX). The K6-1-DOX conjugates were selectively targeted and incorporated into Dsg2-positive hPSCs, leading to pH-dependent endosomal release and nuclear localization of DOX with subsequent cytotoxicity via an apoptotic caspase cascade. Conversely, Dsg2-negative fibroblasts showed minimal conjugate uptake or cytotoxicity, suggesting that K6-1-DOX treatment would yield few side effects owing to off-target effects. Selective removal of undifferentiated stem cells was also supported by in vivo studies using a mouse xenograft model, wherein hIgG-DOX- but not K6-1-DOX-pretreated-hPSC injection led to teratoma development. Together, these results validated the ability of the Dsg2-targeted antibody-anticancer drug conjugate to facilitate the safety of stem cell therapies.


Subject(s)
Antineoplastic Agents , Pluripotent Stem Cells , Teratoma , Antibodies, Monoclonal , Doxorubicin/pharmacology , Humans
7.
Sci Rep ; 9(1): 16757, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31727938

ABSTRACT

Cancer stem-like cells (CSCs) can generate solid tumors through the properties of stem cells such as self-renewal and differentiation and they cause drug resistance, metastasis and recurrence. Therefore, establishing CSC lines is necessary to conduct various studies such as on the identification of CSC origin and specific targeted therapies. In this study, we stimulated NIH3T3 fibroblasts to exhibit the characteristics of CSCs using the whole protein lysates of B16F10 melanoma cells. As a result, we induced colony formation that displayed self-renewal and differentiation capacities through anchorage-independent culture and re-attached culture. Moreover, colonies showed drug resistance by being maintained in the G0/G1 state. Colonies expressed various CSC markers and displayed high-level drug efflux capacity. Additionally, colonies clearly demonstrated tumorigenic ability by forming a solid tumor in vivo. These results show that proteins of cancer cells could transform normal cells into CSCs by increasing expression of CSC markers. This study argues the tremendous importance of the extracellular microenvironmental effect on the generation of CSCs. It also provides a simple experimental method for deriving CSCs that could be based on the development of targeted therapy techniques.


Subject(s)
Fibroblasts/cytology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Neoplastic Stem Cells/cytology , Animals , Cell Culture Techniques , Cell Differentiation , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Fibroblasts/metabolism , Fibroblasts/pathology , Mice , NIH 3T3 Cells , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Cells, Cultured , Tumor Microenvironment
8.
Biomed Pharmacother ; 116: 109050, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31170662

ABSTRACT

The genes of Gekkonidae, a lizard, are known to be very similar to human genes. According to previous research, lizard extracts inhibit angiogenesis and show anticancer activity against various cancers. In this regard, this study assessed whether lizard tail extracts (LTE) cause anticancer activity against lung cancer in mouse and human lung cancer cell lines. The results showed that LTE exhibited anticancer activity against lung cancer in vitro and in vivo. In vitro, cell viability and proliferation decreased in two lung cancer cell lines, while annexin V and single-stranded DNA both increased, showing apoptotic activity caused by LTE. We also found that LTE induced apoptosis in a caspase-3/7 cascade-dependent manner and inhibited the phosphorylation of Akt by participating in the PI3k/Akt pathway. In vivo, LTE decreased tumor volume in LLC bearing mice. Our results demonstrate the potential of LTE as a natural-derived anticancer drug to overcome the chemotherapy side effects during cancer treatment and contribute to the discovery of candidate substances.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lizards/metabolism , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tissue Extracts/pharmacology , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/enzymology , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Clone Cells , Enzyme Activation/drug effects , Humans , Lung Neoplasms/enzymology , Mice, Inbred C57BL , Phosphorylation/drug effects , Signal Transduction/drug effects , Tail , Xenograft Model Antitumor Assays
9.
Stem Cell Reports ; 11(1): 115-127, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29910125

ABSTRACT

Pluripotent stem cells (PSCs) represent the most promising clinical source for regenerative medicine. However, given the cellular heterogeneity within cultivation and safety concerns, the development of specific and efficient tools to isolate a pure population and eliminate all residual undifferentiated PSCs from differentiated derivatives is a prerequisite for clinical applications. In this study, we raised a monoclonal antibody and identified its target antigen as desmoglein-2 (DSG2). DSG2 co-localized with human PSC (hPSC)-specific cell surface markers, and its expression was rapidly downregulated upon differentiation. The depletion of DSG2 markedly decreased hPSC proliferation and pluripotency marker expression. In addition, DSG2-negative population in hPSCs exhibited a notable suppression in embryonic body and teratoma formation. The actions of DSG2 in regulating the self-renewal and pluripotency of hPSCs were predominantly exerted through the regulation of ß-catenin/Slug-mediated epithelial-to-mesenchymal transition. Our results demonstrate that DSG2 is a valuable PSC surface marker that is essential for the maintenance of PSC self-renewal.


Subject(s)
Antigens, Surface/metabolism , Desmoglein 2/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Antigens, Surface/genetics , Biomarkers , Cell Differentiation/genetics , Cell Line , Cell Self Renewal/genetics , Cell Separation/methods , Cellular Reprogramming/genetics , Desmoglein 2/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation , Humans , Immunophenotyping , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , beta Catenin/metabolism
10.
J Hepatol ; 63(6): 1429-39, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26292259

ABSTRACT

BACKGROUND & AIMS: Gallbladder carcinoma (GBC) is the most common malignancy of the biliary tract and one of the most lethal forms of human cancer. However, there is limited information about the molecular pathogenesis of GBC. Here, we examined the functional role of the tumor suppressor N-myc downstream-regulated gene 2 (NDRG2) and the underlying molecular mechanisms of disease progression in GBC. METHODS: Clinical correlations between NDRG2 expression and clinicopathological factors were determined by immunohistochemical analysis of tumor tissues from 86 GBC patients. Biological functions of NDRG2 and NDRG2-mediated signaling pathways were determined in GBC cell lines with NDRG2 knockdown or overexpression. RESULTS: Loss of NDRG2 expression was an independent predictor of decreased survival and was significantly associated with a more advanced T stage, higher cellular grade, and lymphatic invasion in patients with GBC. GBC cells with loss of NDRG2 expression showed significantly enhanced proliferation, migration, and invasiveness in vitro, and tumor growth and metastasis in vivo. Loss of NDRG2 induced the expression of matrix metalloproteinase-19 (MMP-19), which regulated the expression of Slug at the transcriptional level. In addition, MMP-19-induced Slug, increased the expression of a receptor tyrosine kinase, Axl, which maintained Slug expression through a positive feedback loop, and stabilized epithelial-mesenchymal transition of GBC cells. CONCLUSIONS: The results of our study help to explain why the loss of NDRG2 expression is closely correlated with malignancy of GBC. These results strongly suggest that NDRG2 could be a favorable prognostic indicator and promising target for therapeutic agents against GBC.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gallbladder Neoplasms/genetics , Gallbladder Neoplasms/pathology , Matrix Metalloproteinases, Secreted/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Aged , Animals , Cell Line, Tumor , Epithelial-Mesenchymal Transition/physiology , Female , Gallbladder Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Models, Biological , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Snail Family Transcription Factors , Tumor Suppressor Proteins/antagonists & inhibitors , Up-Regulation , Axl Receptor Tyrosine Kinase
11.
Oncotarget ; 6(9): 7182-94, 2015 Mar 30.
Article in English | MEDLINE | ID: mdl-25762629

ABSTRACT

Endostatin is an endogenous angiogenesis inhibitor that exhibits potential anti-tumor efficacy in various preclinical animal models. However, its relatively short in vivo half-life and the long-term, frequent administration of high doses limit its widespread clinical use. In this study, we evaluated whether a fusion protein of murine endostatin (mEndo) to a humanized antibody against tumor-associated glycoprotein 72 (TAG-72), which is highly expressed in several human tumor tissues including colon cancer, can extend the serum half-life and improve the anti-tumor efficacy of endostatin by targeted delivery to the tumor mass. The fusion protein (3E8-mEndo) and mEndo showed improved anti-angiogenic activity in vitro and in vivo, predominantly by interfering with pro-angiogenic signaling triggered by vascular endothelial growth factor (VEGF). Moreover, in mice treated with 3E8-mEndo, we observed a markedly prolonged serum half-life and significantly inhibited tumor growth. The improved anti-tumor activity of 3E8-mEndo can be partially explained by increased local concentration in the tumor mass due to targeted delivery of 3E8-mEndo to implanted colon tumors. Collectively, our data clearly indicate that tumor-targeting antibody fusions to endostatin are a powerful strategy that improves the poor pharmacokinetic profile and anti-tumor efficacy of endostatin.


Subject(s)
Angiogenesis Inhibitors/chemistry , Antigens, Neoplasm/chemistry , Antineoplastic Agents/chemistry , Colorectal Neoplasms/drug therapy , Endostatins/chemistry , Glycoproteins/chemistry , Animals , CHO Cells , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colonic Neoplasms/metabolism , Colorectal Neoplasms/immunology , Cricetinae , Cricetulus , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Recombinant Fusion Proteins/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Clin Cancer Res ; 18(12): 3270-80, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22553346

ABSTRACT

PURPOSE: Although recent studies have suggested that chemokine (C-X-C motif) ligand 12 (CXCL12) is important in the progression of various malignancies, its role in gallbladder carcinoma (GBC) remains unknown. We investigated CXCL12 expression in GBC and its biologic and prognostic role in GBC tumorigenesis. EXPERIMENTAL DESIGN: We examined CXCL12 expression in tumor specimens from 72 patients with GBC by immunohistochemistry and analyzed the correlation between CXCL12 expression and clinicopathologic factors or survival. The functional significance of CXCL12 expression was investigated by CXCL12 treatment and suppression of CXCR4, a major receptor of CXCL12, as well as by CXCL12 overexpression in in vitro and in vivo studies. RESULTS: CXCL12 was differentially expressed in GBC tissues. CXCL12 expression was significantly associated with a high histologic grade (P = 0.042) and nodal metastasis (P = 0.015). Multivariate analyses showed that CXCL12 expression (HR, 8.675; P = 0.014) was an independent risk factor for patient survival. CXCL12 significantly increased anchorage-dependent and -independent growth, migration, invasion, adhesiveness, and survival of GBC cells in vitro, and these effects were dependent on CXCR4. Consistent with these results, overexpression of CXCL12 significantly promoted GBC tumorigenicity in a xenograft model. CONCLUSIONS: Our results indicate that GBC cells express both CXCL12 and its receptor CXCR4, and CXCL12 may have a role in GBC progression through an autocrine mechanism. In addition, CXCL12 is a novel independent poor prognostic factor in patients with GBCs. Thus, targeting CXCL12 and CXCR4 may provide a novel therapeutic strategy for GBC treatment.


Subject(s)
Chemokine CXCL12/metabolism , Disease Progression , Gallbladder Neoplasms/metabolism , Gallbladder Neoplasms/physiopathology , Receptors, CXCR4/metabolism , Adult , Aged , Aged, 80 and over , Animals , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic , Chemokine CXCL12/genetics , Female , Gallbladder/metabolism , Gallbladder Neoplasms/pathology , Humans , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Prognosis , RNA Interference , RNA, Small Interfering , Receptors, CXCR4/genetics , Transplantation, Heterologous
13.
Biochem Biophys Res Commun ; 417(2): 665-72, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22166197

ABSTRACT

A two-component system comprising GacS and GacA affects a large number of traits in many Gram-negative bacteria. However, the signals to which GacS responds, the regulation mechanism for GacA expression, and the genes GacA controls are not yet clear. In this study, several phenotypic tests and tobacco-leaf pathogenicity assays were conducted using a gacA deletion mutant strain (BL473) of Pseudomonas syringae pv. tabaci 11528. To determine the regulation mechanism for gacA gene expression and to identify GacA-regulated genes, we conducted quantitative RT-PCR and electrophoretic mobility shift assay (EMSA) experiments. The results indicated that virulence traits related to the pathogenesis of P. syringae pv. tabaci 11528 are regulated coordinately by GacA and iron availability. They also revealed that several systems coordinately regulate gacA gene expression in response to iron concentration and bacterial cell density and that GacA and iron together control the expression of several virulence genes. EMSA results provided genetic and molecular evidence for direct control of virulence genes by GacA.


Subject(s)
Bacterial Proteins/metabolism , Gene Expression Regulation, Bacterial , Genes, Regulator , Nicotiana/microbiology , Plant Diseases/microbiology , Pseudomonas syringae/genetics , Pseudomonas syringae/pathogenicity , Bacterial Proteins/genetics , Electrophoretic Mobility Shift Assay , Genetic Loci , Iron/metabolism , Plant Leaves/microbiology , Promoter Regions, Genetic , Pseudomonas syringae/metabolism , Virulence/genetics
14.
Cancer Lett ; 316(1): 70-6, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22088438

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) is refractory to conventional chemotherapy. We previously generated chemoresistant ICC (SCK(R)) cells and showed that AKT and ERK signaling conferred cisplatin resistance. Here, we report that epidermal growth factor receptor (EGFR) signaling and L1 cell adhesion molecule (L1CAM) conferred cisplatin resistance in SCK(R) cells in an additive fashion. Activation of EGFR connected to AKT and ERK signaling pathways may induce anti-apoptosis and promote cell proliferation, while L1CAM promoted cell proliferation by mainly activating ERK signaling. Inhibition of EGFR activation or L1ACM greatly sensitized the cells to cisplatin. EGFR and L1CAM may be important targets for ICC therapy.


Subject(s)
Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , Cisplatin/pharmacology , ErbB Receptors/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Bile Duct Neoplasms , Bile Ducts, Intrahepatic , Cell Line, Tumor , Cell Proliferation , Cholangiocarcinoma/genetics , Drug Resistance, Neoplasm , ErbB Receptors/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Liver Neoplasms/genetics , Mice , Mice, Nude , Neural Cell Adhesion Molecule L1/antagonists & inhibitors , Neural Cell Adhesion Molecule L1/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...