Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Front Chem ; 11: 1264593, 2023.
Article in English | MEDLINE | ID: mdl-37720718

ABSTRACT

Li-O2 batteries are a promising technology for the upcoming energy storage requirements because of their high theoretical specific energy density of 11,680 Wh kg-1. Currently, the actual capacity of Li-O2 batteries is much lower than this theoretical value. In many studies, perovskites have been applied as catalysts to improve the air electrode reactions in Li-O2 batteries. The effects of structure and doping on the catalytic activity of perovskites are still unclear. La1-xSrxCoO3-δ (x = 0.1, 0.3, and 0.5) and La0.9Sr0.1YbO3-δ mixed with carbon black (Vulcan XC500 or Super P) were used as air electrode catalysts. Electrochemical characterizations were conducted using a Swagelok-type cell. The charge-discharge capacity and cyclic voltammetry (CV) performance were investigated in this study. The La1-xSrxCoO3-δ (x = 0.1, 0.3, and 0.5) is a suitable cathode catalyst for Li-O2 batteries. In this study, the La0.5Sr0.5CoO3-δ/Super P cathode demonstrated the highest discharge capacity (6,032 mAh g-1). This excellent performance was attributed to the large reaction area and enhanced Li2CO3 generation.

2.
Cancer Res ; 80(22): 5035-5050, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32978170

ABSTRACT

Although epithelial cell adhesion molecule (EpCAM) has previously been shown to promote tumor progression, the underlying mechanisms remain largely unknown. Here, we report that the EGF-like domain I within the extracellular domain of EpCAM (EpEX) binds EGFR, activating both AKT and MAPK signaling to inhibit forkhead transcription factor O3a (FOXO3a) function and stabilize PD-L1 protein, respectively. Treatment with the EpCAM neutralizing antibody, EpAb2-6, inhibited AKT and FOXO3a phosphorylation, increased FOXO3a nuclear translocation, and upregulated high temperature requirement A2 (HtrA2) expression to promote apoptosis while decreasing PD-L1 protein levels to enhance the cytotoxic activity of CD8+ T cells. In vivo, EpAb2-6 markedly extended survival in mouse metastasis and orthotopic models of human colorectal cancer. The combination of EpAb2-6 with atezolizumab, an anti-PD-L1 antibody, almost completely eliminated tumors. Moreover, the number of CD8+ T cells in combination-treated tumors was increased compared with atezolizumab alone. Our findings suggest a new combination strategy for cancer immunotherapy in patients with EpCAM-expressing tumors. SIGNIFICANCE: This study shows that treatment with an EpCAM neutralizing antibody promotes apoptosis while decreasing PD-L1 protein to enhance cytotoxic activity of CD8+ T cells.


Subject(s)
B7-H1 Antigen/chemistry , CD8-Positive T-Lymphocytes/immunology , Disease Progression , Epithelial Cell Adhesion Molecule/metabolism , ErbB Receptors/metabolism , Forkhead Box Protein O3/metabolism , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neutralizing/pharmacology , Antigens, Neoplasm/metabolism , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Nucleus/metabolism , Colorectal Neoplasms/mortality , Colorectal Neoplasms/therapy , Cycloheximide/pharmacology , Enzyme Activation , Heterografts , High-Temperature Requirement A Serine Peptidase 2/metabolism , Humans , Mice , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Transplantation , Phosphorylation/drug effects , Programmed Cell Death 1 Receptor/metabolism , Protein Domains , Protein Stability/drug effects , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Up-Regulation
3.
Materials (Basel) ; 12(4)2019 Feb 14.
Article in English | MEDLINE | ID: mdl-30769818

ABSTRACT

The advent of personalized cancer treatment resulted in the shift from the administration of cytotoxic drugs with broad activity spectrum to a targeted tumor-specific therapy. Aligned to this development, the focus of this study revolved around the application of our novel and patented microtube array membrane (MTAM) in the US National Cancer Institute (NCI) developed an HFA (hollow fiber assay) assay; hereinafter known as MTAM/HFA. Electrospun poly-L-lactic acid (PLLA) MTAM was sterilized and loaded with cell lines/patient derived tumor cells (PDTC) and subcutaneously implanted into the backs of BALB/C mice. Anticancer drugs were administered at the respective time points and the respective MTAMs were retrieved and the viability tumor cells within were quantified with the MTT assay. Results revealed that the MTAMs were excellent culture substrate for various cancer cell lines and PDTCs (patient derived tumor cells). Compared to traditional HFA systems that utilize traditional hollow fibers, MTAM/HFA revealed superior drug sensitivity for a wide range of anticancer drug classes. Additionally, the duration for each test was <14 days; all this while capable of producing similar trend outcome to the current gold-standard xenograft models. These benefits were observed in both the in vitro and in vivo stages, making it a highly practical phenotypic-based solution that could potentially be applied in personalized medicine.

4.
Cancer Lett ; 433: 165-175, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29981429

ABSTRACT

Epithelial cell adhesion molecule (EpCAM) is highly expressed in colon cancers, but its role in cancer progression remains to be elucidated. In this work, we found that the extracellular domain of EpCAM (EpEX) activated EGFR and downstream ERK1/2 signaling to promote colon cancer cell migration and proliferation, as well as tumor growth. Mechanistically, we discovered that EpEX-EGFR-ERK1/2 signaling positively regulated intramembrane proteolysis (RIP) of EpCAM and shedding of the intracellular domain (EpICD). Treatment with an EGFR inhibitor ablated the EpEX-induced phosphorylation of ERK1/2 and AKT. Additionally, treatment with inhibitors of either EGFR or MEK decreased EpEX-induced EpICD shedding and further revealed that EpICD is necessary for nuclear accumulation of ß-catenin and the induction of HIF1α target gene expression in vitro and in vivo. Moreover, an anti-EpCAM neutralizing monoclonal antibody, EpAb2-6, inhibited the nuclear translocation of EpICD and ß-catenin and induced apoptosis in colon cancer cells. Importantly, analysis of colorectal cancer tissues showed that nuclear accumulation of EpICD was highly correlated with metastasis and poor prognosis, suggesting that it may play an important functional role in cancer progression. Thus, we provide novel insights into the mechanisms and functions of EpEX-mediated signaling, which may be considered as a promising target for the treatment of colon cancer.


Subject(s)
Cell Nucleus/metabolism , Colonic Neoplasms/pathology , Epithelial Cell Adhesion Molecule/chemistry , Epithelial Cell Adhesion Molecule/metabolism , MAP Kinase Signaling System , Animals , Cell Line, Tumor , Cell Movement , Colonic Neoplasms/metabolism , Disease Progression , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Mice , Neoplasm Transplantation , Phosphorylation , Prognosis , Protein Domains , Up-Regulation
5.
Oncotarget ; 6(28): 24947-68, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26317650

ABSTRACT

Epithelial cell adhesion molecule (EpCAM) is known to be overexpressed in epithelial cancers associated with enhanced malignant potential, particularly colorectal carcinoma (CRC) and head and neck squamous cell carcinoma (HNSCC). However, it is unknown whether progression of malignance can be directly inhibited by targeting EpCAM. Here, we have generated five novel monoclonal antibodies (mAbs) against EpCAM. One of these anti-EpCAM mAbs, EpAb2-6, was found to induce cancer cell apoptosis in vitro, inhibit tumor growth, and prolong the overall survival of both a pancreatic cancer metastatic mouse model and mice with human colon carcinoma xenografts. EpAb2-6 also increases the therapeutic efficacy of irinotecan, fluorouracil, and leucovorin (IFL) therapy in a colon cancer animal model and gemcitabine therapy in a pancreatic cancer animal model. Furthermore, EpAb2-6, which binds to positions Y95 and D96 of the EGF-II/TY domain of EpCAM, inhibits production of EpICD, thereby decreasing its translocation and subsequent signal activation. Collectively, our results indicate that the novel anti-EpCAM mAb can potentially be used for cancer-targeted therapy.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Adhesion Molecules/antagonists & inhibitors , Colonic Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Antigens, Neoplasm/metabolism , Apoptosis/drug effects , Blotting, Western , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Synergism , Epithelial Cell Adhesion Molecule , Flow Cytometry , Fluorouracil/administration & dosage , HCT116 Cells , Humans , Irinotecan , Leucovorin/administration & dosage , Mice, Inbred NOD , Mice, SCID , Molecular Targeted Therapy/methods , Vitamin B Complex/administration & dosage
6.
J Cell Sci ; 125(Pt 20): 4853-64, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22854048

ABSTRACT

Hepatocyte growth factor/scatter factor (HGF) is unique by inducing epithelial cell scattering, a cellular event pivotal to HGF-mediated invasive-growth response essential for embryonic development and metastasis. Krüppel-like factor 4 (KLF4) is a multifunctional zinc-finger transcription factor involved in cell proliferation, differentiation and self-renewal. We herein present the first evidence for the functional connection between KLF4 and HGF-induced cell scattering. In particular, we found that KLF4 was upregulated by HGF in two independent epithelial cell types, HepG2 and MDCK, whereas KLF4 knockdown inhibited HGF-induced E-cadherin suppression and cell scattering. Moreover, enforced nuclear KLF4 expression alone was sufficient to upregulate KLF4, downregulate E-cadherin and trigger scattering. Chromatin immunoprecipitation (ChIP) analysis further revealed that KLF4 induced suppression of E-cadherin transcription by directly binding to the E-cadherin promoter. Additionally, we proved that HGF-induced upregulation of KLF4 transcription and cell scattering require activation of the MEK/ERK signaling pathway and the induction of early growth response 1 (EGR-1). At the mechanistic level, ChIP analysis validated a direct binding of EGR-1 to the KLF4 promoter to induce KLF4 transcription; in turn, EGR-1-induced KLF4 binds to its own promoter, thus creating a positive feedback mechanism to sustain KLF4 expression and the resultant cell scattering. We conclude that KLF4 upregulation by HGF represents a novel mechanism mediating HGF-induced cell scattering and perhaps other associated events such as cell migration and invasion.


Subject(s)
Cell Movement/genetics , Epithelial Cells , Hepatocyte Growth Factor , Kruppel-Like Transcription Factors , Animals , Cadherins/genetics , Cadherins/metabolism , Dogs , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Embryonic Development/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation , Hep G2 Cells , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Humans , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , MAP Kinase Signaling System , Madin Darby Canine Kidney Cells , Neoplasm Invasiveness , Neoplasm Metastasis/genetics , Signal Transduction
7.
Toxicol Appl Pharmacol ; 225(3): 318-28, 2007 Dec 15.
Article in English | MEDLINE | ID: mdl-17881028

ABSTRACT

Undecylprodigiosin (UP) is a bacterial bioactive metabolite produced by Streptomyces and Serratia. In this study, we explored the anticancer effect of UP. Human breast carcinoma cell lines BT-20, MCF-7, MDA-MB-231 and T47D and one nonmalignant human breast epithelial cell line, MCF-10A, were tested in this study. We found that UP exerted a potent cytotoxicity against all breast carcinoma cell lines in a dose- and time-dependent manner. In contrast, UP showed limited toxicity to MCF-10A cells, indicating UP's cytotoxic effect is selective for malignant cells. UP's cytotoxic effect was due to apoptosis, as confirmed by positive TUNEL signals, annexin V-binding, caspase 9 activation and PARP cleavage. Notably, UP-induced apoptosis was blocked by the pan-caspase inhibitor z-VAD.fmk, further indicating the involvement of caspase activity. Moreover, UP caused a marked decrease of the levels of antiapoptotic BCL-X(L), Survivin and XIAP while enhancing the levels of proapoptotic BIK, BIM, MCL-1S and NOXA, consequently favoring induction of apoptosis. Additionally, we found that cells with functional p53 (MCF-7, T47D) or mutant p53 (BT-20, MDA-MB-231) were both susceptible to UP's cytotoxicity. Importantly, UP was able to induce apoptosis in MCF-7 cells with p53 knockdown by RNA interference, confirming the dispensability of p53 in UP-induced apoptosis. Overall, our results establish that UP induces p53-independent apoptosis in breast carcinoma cells with no marked toxicity to nonmalignant cells, raising the possibility of its use as a new chemotherapeutic drug for breast cancer irrespective of p53 status.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism , Annexin A5/drug effects , Annexin A5/metabolism , Antineoplastic Agents/administration & dosage , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Caspase 9/drug effects , Caspase 9/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , In Situ Nick-End Labeling , Inhibitor of Apoptosis Proteins , Membrane Proteins/metabolism , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/drug effects , Neoplasm Proteins/metabolism , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Prodigiosin/administration & dosage , Prodigiosin/analogs & derivatives , Prodigiosin/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Serratia marcescens/chemistry , Survivin , Time Factors , Tumor Suppressor Protein p53/drug effects , X-Linked Inhibitor of Apoptosis Protein/drug effects , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-X Protein/drug effects , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...