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.
Int J Biol Macromol ; 221: 121-134, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36049568

ABSTRACT

Photothermal and photodynamic therapies (PTT/PDT) have been widely accepted as noninvasive therapeutic methods for cancer treatment. However, tumor hypoxia and insufficient delivery of photoactive compounds to cancer cells can reduce the efficacy of phototherapy. Herein, we first synthesized thiolated hyaluronic acid (THA) and then conjugated it with catalase (CAT) onto chlorin e6 (Ce6)-adsorbed small gold nanorods (Ce6@sAuNRs) with near-infrared (NIR)/visible light activated photothermal/photodynamic effects. The conjugation of THA and CAT on Ce6@sAuNRs resulted in a red-shift of the longitudinal LSPR absorption band of sAuNRs up to 1000 nm and maintained the excellent enzymatic activity of catalase. Modification of Ce6@sAuNRs with THA resulted in efficient internalization of the nanocomposite into MCF-7/ADR multidrug-resistant (MDR) breast cancer cells (CD44+), thereby significantly enhancing the intracellular accumulation of the photosensitizer Ce6. CAT endows Ce6@sAuNRs with self-supporting oxygen production, which enables them to efficiently generate singlet oxygen (1O2) under 660 nm laser irradiation and enhances the photodynamic effect against hypoxic breast cancer cells. The results highlight the prospect of this novel multi-functional nanoplatform integrating active biological macromolecules (THA and CAT) into photosensitizer/photothermal gold nanocomposites in overcoming the limitations of hypoxic MDR breast cancer cell treatment.


Subject(s)
Breast Neoplasms , Photochemotherapy , Porphyrins , Catalase , Gold/pharmacology , Hyaluronic Acid/pharmacology , Oxygen , Photochemotherapy/methods , Photosensitizing Agents , Porphyrins/pharmacology , Breast Neoplasms/drug therapy , Humans , Hyaluronan Receptors , Nanotubes , MCF-7 Cells
2.
Carbohydr Polym ; 281: 119035, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35074103

ABSTRACT

Chitosan (CS) electrospun nanofiber (ENF) membranes were modified with fucoidan (Fu) and CuS NPs through polyelectrolyte complexation and genipin (GP)-involved cross-linking reaction. The formation of Fu/CS complex and cross-linking of CS with GP increased the acid resistance and reduced the swelling rate of CS ENF, while the covalent conjugation of CuS NPs provided CS ENF with durable Fenton-like catalytic activity. The CuS@ENF composite (ENFC) effectively adsorbed H2O2 and near-infrared (NIR) light, enabling it to kill bacteria by photothermal and photocatalytic bactericidal effects. Fu and copper ions were able to release from the ENFC in a pH-dependent manner, and promoted the alkaline phosphatase activity of osteoblast cells and capillary tube formation of endothelial cells. This study provides a new approach to modify CS ENF with antibacterial and osteoblast differentiation activities, which may be available for bone infection prevention and tissue regeneration.


Subject(s)
Chitosan , Nanofibers , Anti-Bacterial Agents/pharmacology , Chitosan/pharmacology , Copper , Endothelial Cells , Hydrogen Peroxide , Polysaccharides , Tissue Engineering
3.
Cells ; 10(7)2021 07 03.
Article in English | MEDLINE | ID: mdl-34359854

ABSTRACT

Integrin αvß3, a cell surface receptor, participates in signaling transduction pathways in cancer cell proliferation and metastasis. Several ligands bind to integrin αvß3 to regulate proliferation and metastasis in cancer cells. Crosstalk between the integrin and other signal transduction pathways also plays an important role in modulating cancer proliferation. Carcinoembryonic antigen cell adhesion molecule 6 (CEACAM6) activates the downstream integrin FAK to stimulate biological activities including cancer proliferation and metastasis. Blockage of signals related to integrin αvß3 was shown to be a promising target for cancer therapies. 3,3',5,5'-tetraiodothyroacetic acid (tetrac) completely binds to the integrin with the thyroid hormone to suppress cancer proliferation. The (E)-stilbene analog, resveratrol, also binds to integrin αvß3 to inhibit cancer growth. Recently, nanotechnologies have been used in the biomedical field for detection and therapeutic purposes. In the current review, we show and evaluate the potentiation of the nanomaterial carrier RGD peptide, derivatives of PLGA-tetrac (NDAT), and nanoresveratrol targeting integrin αvß3 in cancer therapies.


Subject(s)
Integrin alphaVbeta3/metabolism , Nanomedicine , Neoplasms/therapy , Animals , Humans , Molecular Targeted Therapy , Nanoparticles/chemistry , Signal Transduction
4.
Cells ; 9(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32756527

ABSTRACT

The property of drug-resistance may attenuate clinical therapy in cancer cells, such as chemoresistance to gefitinib in colon cancer cells. In previous studies, overexpression of PD-L1 causes proliferation and metastasis in cancer cells; therefore, the PD-L1 pathway allows tumor cells to exert an adaptive resistance mechanism in vivo. Nano-diamino-tetrac (NDAT) has been shown to enhance the anti-proliferative effect induced by first-line chemotherapy in various types of cancer, including colorectal cancer (CRC). In this work, we attempted to explore whether NDAT could enhance the anti-proliferative effect of gefitinib in CRC and clarified the mechanism of their interaction. The MTT assay was utilized to detect a reduction in cell proliferation in four primary culture tumor cells treated with gefitinib or NDAT. The gene expression of PD-L1 and other tumor growth-related molecules were quantified by quantitative polymerase chain reaction (qPCR). Furthermore, the identification of PI3K and PD-L1 in treated CRC cells were detected by western blotting analysis. PD-L1 presentation in HCT116 xenograft tumors was characterized by specialized immunohistochemistry (IHC) and the hematoxylin and eosin stain (H&E stain). The correlations between the change in PD-L1 expression and tumorigenic characteristics were also analyzed. (3) The PD-L1 was highly expressed in Colo_160224 rather than in the other three primary CRC cells and HCT-116 cells. Moreover, the PD-L1 expression was decreased by gefitinib (1 µM and 10 µM) in two cells (Colo_150624 and 160426), but 10 µM gefitinib stimulated PD-L1 expression in gefitinib-resistant primary CRC Colo_160224 cells. Inactivated PI3K reduced PD-L1 expression and proliferation in CRC Colo_160224 cells. Gefitinib didn't inhibit PD-L1 expression and PI3K activation in gefitinib-resistant Colo_160224 cells. However, NDAT inhibited PI3K activation as well as PD-L1 accumulation in gefitinib-resistant Colo_160224 cells. The combined treatment of NDAT and gefitinib inhibited pPI3K and PD-L1 expression and cell proliferation. Additionally, NDAT reduced PD-L1 accumulation and tumor growth in the HCT116 (K-RAS mutant) xenograft experiment. (4) Gefitinib might suppress PD-L1 expression but did not inhibit proliferation through PI3K in gefitinib-resistant primary CRC cells. However, NDAT not only down-regulated PD-L1 expression via blocking PI3K activation but also inhibited cell proliferation in gefitinib-resistant CRCs.


Subject(s)
B7-H1 Antigen/genetics , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Gefitinib/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Polyglactin 910/pharmacology , Thyroxine/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/metabolism , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Gefitinib/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Mice , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Polyglactin 910/therapeutic use , Thyroxine/pharmacology , Thyroxine/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Mar Drugs ; 18(7)2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32630719

ABSTRACT

BACKGROUND: Heteronemin, a marine sesterterpenoid-type natural product, possesses an antiproliferative effect in cancer cells. In addition, heteronemin has been shown to inhibit p53 expression. Our laboratory has demonstrated that the thyroid hormone deaminated analogue, tetrac, activates p53 and induces antiproliferation in colorectal cancer. However, such drug mechanisms are still to be studied in oral cancer cells. METHODS: We investigated the antiproliferative effects by Cell Counting Kit-8 and flow cytometry. The signal transduction pathway was measured by Western blotting analyses. Quantitative PCR was used to evaluate gene expression regulated by heteronemin, 3,3',5,5'-tetraiodothyroacetic acid (tetrac), or their combined treatment in oral cancer cells. RESULTS: Heteronemin inhibited not only expression of proliferative genes and Homo Sapiens Thrombospondin 1 (THBS-1) but also cell proliferation in both OEC-M1 and SCC-25 cells. Remarkably, heteronemin increased TGF-ß1 expression in SCC-25 cells. Tetrac suppressed expression of THBS-1 but not p53 expression in both cancer cell lines. Furthermore, the synergistic effect of tetrac and heteronemin inhibited ERK1/2 activation and heteronemin also blocked STAT3 signaling. Combined treatment increased p53 protein and p53 activation accumulation although heteronemin inhibited p53 expression in both cancer cell lines. The combined treatment induced antiproliferation synergistically more than a single agent. CONCLUSIONS: Both heteronemin and tetrac inhibited ERK1/2 activation and increased p53 phosphorylation. They also inhibited THBS-1 expression. Moreover, tetrac suppressed TGF-ß expression combined with heteronemin to further enhance antiproliferation and anti-metastasis in oral cancer cells.


Subject(s)
Carcinoma/drug therapy , Cell Proliferation/drug effects , Gingival Neoplasms/drug therapy , Terpenes/pharmacology , Thyroxine/analogs & derivatives , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Terpenes/administration & dosage , Thyroxine/administration & dosage , Thyroxine/pharmacology
6.
ACS Comb Sci ; 21(4): 269-275, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30775908

ABSTRACT

Base-controlled regioselective synthesis of 2-imino thiazolines and 2-thioxoimidazolin-4-ones was achieved to use a one-pot reaction between chiral amino esters, isothiocyanates, and α-bromoketones/alkyl halides. This three-component coupling reaction in acetonitrile provides 2-imino thiazolines, whereas the formation of 2-thioxoimidazolin-4-ones was observed under basic conditions at ambient temperature. The corresponding products were obtained in good to excellent yield with broad substrate scope. Isolation of thiourea and thiohydantoin intermediates disclosed the course of the reaction mechanism.


Subject(s)
Imidazolidines/chemical synthesis , Thiazoles/chemical synthesis , Esters/chemistry , Imidazolidines/chemistry , Imines/chemistry , Isothiocyanates/chemistry , Ketones/chemistry , Molecular Structure , Oxidation-Reduction , Stereoisomerism , Thiazoles/chemistry , Thiohydantoins/chemistry , Thiourea/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...