Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Nanoscale ; 16(22): 10618-10627, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38764380

ABSTRACT

The CoSn(OH)6 perovskite hydroxide is a structure stable and inexpensive electrocatalyst for the oxygen evolution reaction (OER). However, the OER activity of CoSn(OH)6 is still unfavorable due to its limited active sites. In this work, an Fe3+ doping strategy is used to optimize the d-band state of the CoSn(OH)6 perovskite hydroxide. The CoSn(OH)6 catalyst with slightly Fe3+ doped nanocubes is synthesized by a facile hydrothermal method. Structure characterization shows that Fe3+ ions are incorporated into the crystal structure of CoSn(OH)6. Owing to the regulation of the electronic structure, CoSn(OH)6-Fe1.8% exhibits an OER overpotential of 289 mV at a current density of 10 mA cm-2 in OER electrochemical tests. In situ Raman spectroscopy shows that no obvious re-construction occurred during the OER for both CoSn(OH)6 and CoSn(OH)6-Fe1.8%. DFT calculations show that the introduction of Fe3+ into CoSn(OH)6 can shift the d-band center to a relatively high position, thus promoting the OER intermediates' adsorption ability. Further DFT calculations suggest that incorporation of an appropriate amount of Fe3+ into CoSn(OH)6 significantly reduces the rate-determining Gibbs free energy during the OER. This work offers valuable insights into tuning the d-band center of perovskite hydroxide materials for efficient OER applications.

2.
ACS Nano ; 17(24): 25091-25100, 2023 Dec 26.
Article in English | MEDLINE | ID: mdl-38054420

ABSTRACT

Electrochemical NO3- reduction reaction (NO3RR) is a promising technique for green NH3 synthesis. Tungsten oxide (WO3) has been regarded as an effective electrocatalyst for electrochemical NH3 synthesis. However, the weak adsorption and the sluggish hydrogenation of oxynitride intermediates (NOx, e.g., *NO3 and *NO2) over WO3 materials hinder the efficiency of converting NO3- to NH3. Herein, we design a heterostructure of tungsten nitride (WN) and WO3 (WN/WO3) nanosheets to optimize *NO3 and *NO2 adsorptions and facilitate *NO2 hydrogenations to achieve a highly efficient electrochemical NO3RR to produce NH3. Theoretical calculations predict that locally introducing WN into WO3 will shorten the distance between adjacent W atoms, resulting in *NO3 and *NO2 being strongly adsorbed on W active sites in the form of bidentate ligands instead of the relatively weak monodentate ligands. Furthermore, WN facilitates H2O dissociation to supply the requisite protons, which is beneficial for *NO2 hydrogenations. Inspired by theoretical prediction, WN/WO3 nanosheets are successfully fabricated through a high-temperature nitridation process. The transmission electron microscopy, X-ray photoelectron spectroscopy, and X-ray absorption near-edge spectroscopy investigations confirm that the amorphous WN has been locally introduced in situ into WO3 nanosheets to form a composite heterostructure. The as-prepared WN/WO3 nanosheets exhibit a high Faraday efficiency of 88.9 ± 7.2% and an appreciable yield rate of 8.4 mg h-1 cm-2 toward NH3 production, which is much higher than that of individual WO3 and WN. The enhanced adsorption and hydrogenation behaviors of *NOx over WN/WO3 are characterized by in situ Fourier-transform infrared spectroscopy, consistent with the theoretical predictions. This work develops facile and effective heterostructure nanomaterials to tune the adsorption and hydrogenation of NOx for boosting the efficiency from NO3- to NH3.

3.
J Pharm Biomed Anal ; 115: 418-30, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26280925

ABSTRACT

Mogroside V, a cucurbitane-type saponin, is not only the major bioactive constituent of traditional Chinese medicine Siraitiae Fructus, but also a widely used sweetener. To clarify its biotransformation process and identify its effective forms in vivo, we studied its metabolism in a human intestinal bacteria incubation system, a rat hepatic 9000g supernatant (S9) incubation system, and rats. Meanwhile, the distribution of mogroside V and its metabolites was also reported firstly. Seventy-seven new metabolites, including 52 oxidation products formed by mono- to tetra- hydroxylation/dehydrogenation, were identified with the aid of HPLC in tandem with ESI ion trap (IT) TOF multistage mass spectrometry (HPLC-ESI-IT-TOF-MS(n)). Specifically, 14 metabolites were identified in human intestinal bacteria incubation system, 4 in hepatic S9 incubation system, 58 in faeces, 29 in urine, 14 in plasma, 34 in heart, 33 in liver, 39 in spleen, 39 in lungs, 42 in kidneys, 45 in stomach, and 51 in small intestine. The metabolic pathways of mogroside V were proposed and the identified metabolic reactions were deglycosylation, hydroxylation, dehydrogenation, isomerization, glucosylation, and methylation. Mogroside V and its metabolites were distributed unevenly in the organs of treated rats. Seven bioactive metabolites of mogroside V were identified, among which mogroside IIE was abundant in heart, liver, spleen and lung, suggesting that it may contribute to the bioactivities of mogroside V. Mogroside V was mainly excreted in urine, whereas its metabolites were mainly excreted in faeces. To our knowledge, this is the first report that a plant constituent can be biotransformed into more than 65 metabolites in vivo. These findings will improve understanding of the in vivo metabolism, distribution, and effective forms of mogroside V and congeneric molecules.


Subject(s)
Chromatography, High Pressure Liquid , Drugs, Chinese Herbal/pharmacokinetics , Spectrometry, Mass, Electrospray Ionization , Sweetening Agents/pharmacokinetics , Triterpenes/pharmacokinetics , Administration, Oral , Animals , Biotransformation , Drugs, Chinese Herbal/administration & dosage , Feces/microbiology , Gastrointestinal Microbiome , Glycosylation , Humans , Hydroxylation , Intestines/microbiology , Male , Methylation , Microsomes, Liver/metabolism , Molecular Structure , Rats, Sprague-Dawley , Sweetening Agents/administration & dosage , Tissue Distribution , Triterpenes/administration & dosage
4.
Oncotarget ; 6(19): 17738-52, 2015 Jul 10.
Article in English | MEDLINE | ID: mdl-26036634

ABSTRACT

Lapatinib has been used in combination with capecitabine or paclitaxel to treat patients with progressive HER2-overexpressing metastatic breast cancer (MBC). Unfortunately, an increased incidence of hepatotoxicity had been reported in the combinational therapy. The aim of this study was to investigate the potential mechanisms of this combinational therapy. We found that the patients receiving lapatinib and paclitaxel treatment showed a higher incidence of hepatobiliary system disorders than those receiving paclitaxel alone. Lapatinib was shown to increase the accumulation of doxorubicin in ABCB1-overexpressing hepatocellular cancer cells and normal liver tissues without altering the protein level of ABCB1. Pharmacokinetic studies revealed that lapatinib could increase the systematic exposure of paclitaxel and doxorubicin. Moreover, the in vivo experiments showed that the levels of alanine aminotransferase and serious hepatocyte injury in the group of lapatinib plus chemotherapeutic agent were significantly higher than those in the group of single chemotherapeutic agent such as paclitaxel or doxorubicin. Our study thus revealed for the first time that the higher incidence of hepatotoxicity during this combinational treatment was due to the increased drug accumulation in hepatocytes mediated by the inhibition of ABCB1 by lapatinib. Appropriate dose adjustment may be needed to optimize the combination therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Breast Neoplasms/drug therapy , Hepatocytes/drug effects , Quinazolines/adverse effects , ATP Binding Cassette Transporter, Subfamily B/drug effects , Animals , Blotting, Western , Cell Line, Tumor , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Interactions , Female , Humans , Immunohistochemistry , Lapatinib , Mice , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Quinazolines/administration & dosage , Quinazolines/pharmacokinetics , Reverse Transcriptase Polymerase Chain Reaction
5.
Am J Cancer Res ; 5(3): 1000-16, 2015.
Article in English | MEDLINE | ID: mdl-26045982

ABSTRACT

Despite a variety of novel therapeutic agents, such as bortezomib, thalidomide and topotecan, multiple myeloma (MM) remains an incurable disease, thus the development of new chemotherapeutical agents is of high priority. We found HM910, a novel camptothecin (CPT) derivative, exhibited potent inhibition of MM cell growth in vitro and in xenografts of nude mice. Mechanistically, HM910 reduced the mitochondrial transmembrane potential (ΔΨm) via an increase in reactive oxygen species (ROS), which eventually resulting in the release of cytochrome c and the activation of mitochondrial-dependent apoptotic pathway. On the other hand, HM910 significantly triggered cell cycle arrest in G1 phase via downregulating the expressions of cyclin dependent kinase (CDK) 4 and 6, resulting in down-regulation of cyclin D1. Therefore, HM910 maybe a promising candidate for treating MM patients and is currently in phase I clinical trial in China.

6.
Oncotarget ; 5(23): 11971-85, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25436978

ABSTRACT

Multidrug resistance (MDR) to chemotherapeutic drugs is a formidable barrier to the success of cancer chemotherapy. Expressions of ATP-binding cassette (ABC) transporters contribute to clinical MDR phenotype. In this study, we found that afatinib, a small molecule tyrosine kinase inhibitor (TKI) targeting EGFR, HER-2 and HER-4, reversed the chemoresistance mediated by ABCG2 in vitro, but had no effect on that mediated by multidrug resistance protein ABCB1 and ABCC1. In addition, afatinib, in combination with topotecan, significantly inhibited the growth of ABCG2- overexpressing cell xenograft tumors in vivo. Mechanistic investigations exhibited that afatinib significantly inhibited ATPase activity of ABCG2 and downregulated expression level of ABCG2, which resulted in the suppression of efflux activity of ABCG2 in parallel to the increase of intracellular accumulation of ABCG2 substrate anticancer agents. Taken together, our findings may provide a new and useful combinational therapeutic strategy of afatinib with chemotherapeutical drug for the patients with ABCG2 overexpressing cancer cells.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Neoplasm Proteins/antagonists & inhibitors , Neoplasms, Experimental/drug therapy , Quinazolines/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , Afatinib , Animals , Blotting, Western , Cell Line, Tumor , Female , Flow Cytometry , Humans , Immunohistochemistry , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/metabolism , Real-Time Polymerase Chain Reaction , Topotecan/pharmacology , Xenograft Model Antitumor Assays
7.
Mol Med ; 20: 390-9, 2014 Sep 08.
Article in English | MEDLINE | ID: mdl-25105301

ABSTRACT

Lapatinib, a tyrosine kinase inhibitor, is used in the treatment of advanced or metastatic breast cancer overexpressing human epidermal receptor 2 (HER2). Lapatinib can modulate the function of ATP-binding cassette (ABC) transporters (ABCB1 and ABCG2), which are the major mechanism responsible for multidrug resistance (MDR) in cancer. In this study, we investigated the effect of lapatinib on multidrug resistance-associated protein 1 (MRP1 [ABCC1]), MRP2 (ABCC2), MRP4 (ABCC4) and lung relative resistance protein (LRP) drug efflux pumps. We demonstrated that lapatinib could enhance the efficacy of conventional chemotherapeutic agents in MRP1-overexpressing cells in vitro and in vivo, but no effect in MRP2-, MPR4- and LRP-overexpressing cells. Furthermore, lapatinib significantly increased the accumulation of rhodamine 123 (Rho123) and doxorubicin (DOX) in MRP1-overexpressing cells. However, lapatinib did not alter the protein or mRNA expression levels of MRP1. Further studies showed that the level of phosphorylation of AKT and extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) were not altered at the indicated concentrations of lapatinib. In conclusion, lapatinib enhanced the efficacy of conventional chemotherapeutic agents in MRP1-overexpressing cells by inhibiting MRP1 transport function without altering the level of AKT or ERK1/2 phosphorylation. These findings will encourage the clinical research of lapatinib combined with conventional chemotherapeutic drugs in MRP1-overexpressing cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Quinazolines/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Biological Transport/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Doxorubicin/pharmacology , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Humans , Lapatinib , Mice , Mice, Nude , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , NIH 3T3 Cells , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/therapeutic use , Tumor Burden/drug effects , Vincristine/pharmacology , Vincristine/therapeutic use
8.
Biochem Pharmacol ; 91(2): 144-56, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25058526

ABSTRACT

The overexpression of ATP-binding cassette (ABC) transporters often leads to the development of multidrug resistance (MDR), which is the major factor contributing to the failure of chemotherapy. The objective of this study was to investigate the enhancement of CEP-33779, a small-molecule inhibitor of Janus kinase 2 (JAK2), on the efficacy of conventional chemotherapeutic agents in MDR cells with overexpression of P-glycoprotein (ABCB1), multidrug resistance-associated protein 1 (ABCC1) and breast cancer resistance protein (ABCG2). Our results showed that CEP-33779, at nontoxic concentrations, significantly sensitized ABCB1 overexpressing MDR cells to its anticancer substrates. CEP-33779 significantly increased intracellular accumulation and decreased the efflux of doxorubicin by inhibiting the ABCB1 transport function. Furthermore, CEP-33779 did not alter the expression of ABCB1 both at protein and mRNA levels but did stimulate the activity of ABCB1 ATPase. CEP-33779 was predicted to bind within the large hydrophobic cavity of homology modeled ABCB1. In addition, the down-regulation of JAK2 by shRNA altered neither the expression of ABCB1 nor the cytotoxic effect of chemotherapeutic agents in ABCB1-overexpressing cells. Significantly, CEP-33779 enhanced the efficacy of vincristine against the ABCB1-overexpressing and drug resistant KBv200 cell xenograft in nude mice. In conclusion, we conclude that CEP-33779 enhances the efficacy of substrate drugs in ABCB1-overexpressing cells by directly inhibiting ABCB1 transport function. The findings encouraged to further study on the combination therapy of CEP-33779 with conventional chemotherapeutic agents in ABCB1 mediated-MDR cancer patients.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Pyridines/pharmacology , Triazoles/pharmacology , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/metabolism , Heterografts , Humans , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental , Real-Time Polymerase Chain Reaction , Rhodamine 123/metabolism , Topotecan/administration & dosage , Topotecan/metabolism
9.
Cancer Res ; 74(16): 4431-45, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-24972892

ABSTRACT

Cancer stem cells (CSC) have garnered significant attention as a therapeutic focus, based on evidence that they may represent an etiologic root of treatment-resistant cells. Indeed, expression of the multidrug resistance protein ATP-binding cassette subfamily G member 2 (ABCG2) confers chemoresistance to CSCs, where it serves as a potential biomarker and therapeutic target. Here, we show that afatinib, a small-molecule inhibitor of the tyrosine kinases EGFR, HER2, and HER4, preferentially eliminated side population cells with CSC character, in both cell lines and patient-derived leukemia cells, by decreasing ABCG2 expression. In these cells, afatinib also acted in parallel to suppress self-renewal capacity and tumorigenicity. Combining afatinib with the DNA-damaging drug topotecan enhanced the antitumor effect of topotecan in vitro and in vivo. Mechanistic investigations suggested that ABCG2 suppression by afatinib did not proceed by proteolysis through the ubiquitin-dependent proteosome, lysosome, or calpain. Instead, we found that afatinib increased DNA methyltransferase activity, thereby leading to methylation of the ABCG2 promoter and to a decrease in ABCG2 message level. Taken together, our results advocate the use of afatinib in combination with conventional chemotherapeutic drugs to improve efficacy by improving CSC eradication.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplastic Stem Cells/drug effects , Quinazolines/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/biosynthesis , ATP-Binding Cassette Transporters/genetics , Afatinib , Animals , Cell Line, Tumor , DNA Methylation , Drug Synergism , Humans , Leukemia/drug therapy , Leukemia/genetics , Leukemia/metabolism , Leukemia/pathology , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic , Protein Kinase Inhibitors/pharmacology , Quinazolines/administration & dosage , Random Allocation , Signal Transduction , Topotecan/administration & dosage , Topotecan/pharmacology , Xenograft Model Antitumor Assays
10.
Am J Cancer Res ; 4(2): 148-60, 2014.
Article in English | MEDLINE | ID: mdl-24660104

ABSTRACT

Multidrug resistance (MDR) mediated by ATP-binding cassette (ABC) transporters through efflux of antineoplastic agents from cancer cells is a major obstacle to successful cancer chemotherapy. The inhibition of these ABC transporters is thus a logical approach to circumvent MDR. There has been intensive research effort to design and develop novel inhibitors for the ABC transporters to achieve this goal. In the present study, we evaluated the ability of UMMS-4 to modulate P-glycoprotein (P-gp/ABCB1)-, breast cancer resistance protein (BCRP/ABCG2)- and multidrug resistance protein (MRP1/ABCC1)-mediated MDR in cancer cells. Our findings showed that UMMS-4, at non-cytotoxic concentrations, apparently circumvents resistance to ABCB1 substrate anticancer drugs in ABCB1-overexpressing cells. When used at a concentration of 20 µmol/L, UMMS-4 produced a 17.53-fold reversal of MDR, but showed no effect on the sensitivity of drug-sensitive parental cells. UMMS-4, however, did not significantly alter the sensitivity of non-ABCB1 substrates in all cells and was unable to reverse ABCG2- and ABCC1-mediated MDR. Additionally, UMMS-4 profoundly inhibited the transport of rhodamine 123 (Rho 123) and doxorubicin (Dox) by the ABCB1 transporter. Furthermore, UMMS-4 did not alter the expression of ABCB1 at the mRNA and protein levels. In addition, the results of ATPase assays showed that UMMS-4 stimulated the ATPase activity of ABCB1. Taken together, we conclude that UMMS-4 antagonizes ABCB1-mediated MDR in cancer cells through direct inhibition of the drug efflux function of ABCB1. These findings may be useful for the development of safer and more effective MDR modulator.

SELECTION OF CITATIONS
SEARCH DETAIL
...