Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Immunother Cancer ; 11(6)2023 06.
Article in English | MEDLINE | ID: mdl-37295817

ABSTRACT

BACKGROUND: Advanced or metastatic prostate cancer (PCa) is still an incurable malignancy with high lethality and a poor prognosis. Despite the remarkable success of immunotherapy against many types of cancer, most patients with PCa receive minimal benefit from current immunotherapeutic strategies, because PCa is an immune cold tumor with scarce T-cell infiltration in the tumor microenvironment. The aim of this study was to develop an effective immunotherapeutic approach for immune cold PCa tumors. METHODS: The therapeutic efficacy of androgen deprivation therapy (ADT) and zoledronic acid (ZA) plus thymosin α1 (Tα1) therapy was analyzed retrospectively in patients with advanced or metastatic PCa. The effects and mechanisms by which ZA and Tα1 regulated the immune functions of PCa cells and immune cells were evaluated by a PCa allograft mouse model, flow cytometric analysis, immunohistochemical and immunofluorescence staining assays, and PCR, ELISA, and Western blot analyses. RESULTS: In this study, clinical retrospective analysis revealed that ADT combined with ZA plus Tα1 improved the therapeutic outcomes of patients with PCa, which might be associated with an enhanced frequency of T cells. ZA and Tα1 treatment synergistically inhibited the growth of androgen-independent PCa allograft tumors, with increased infiltration of tumor-specific cytotoxic CD8+ T cells and enhanced tumor inflammation. Functionally, ZA and Tα1 treatment relieved immunosuppression in PCa cells, stimulated pro-inflammatory macrophages, and enhanced the cytotoxic function of T cells. Mechanistically, ZA plus Tα1 therapy blocked the MyD88/NF-κB pathway in PCa cells but activated this signaling in macrophages and T cells, altering the tumor immune landscape to suppress PCa progression. CONCLUSIONS: These findings uncover a previously undefined role for ZA and Tα1 in inhibiting the disease progression of immune cold PCa tumors by enhancing antitumor immunity and pave the way for the application of ZA plus Tα1 therapy as an immunotherapeutic strategy for treating patients with immunologically unresponsive PCa.


Subject(s)
Prostatic Neoplasms , Humans , Male , Animals , Mice , Zoledronic Acid/pharmacology , Zoledronic Acid/therapeutic use , Thymalfasin/pharmacology , Thymalfasin/therapeutic use , Prostatic Neoplasms/pathology , Retrospective Studies , T-Lymphocytes, Cytotoxic/metabolism , CD8-Positive T-Lymphocytes/metabolism , Androgen Antagonists/therapeutic use , Androgens/therapeutic use , Inflammation , Tumor Microenvironment
2.
Free Radic Biol Med ; 204: 301-312, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37217090

ABSTRACT

Colorectal cancer (CRC) is the third most common cause of cancer mortality worldwide. Approximately 40% of CRC patients are KRAS sequence variation, including KRAS G13D mutation (KRASG13D) CRC patients, accounting for approximately 8% of all KRAS mutations in CRC patients and showing little benefit from anti-EGFR therapy. Therefore, there is an urgent need for new and effective anticancer agents in patients with KRASG13D CRC. Here, we identified a natural product, erianin, that directly interacted with purified recombinant human KRASG13D with a Kd of 1.1163 µM, which also significantly improve the thermal stability of KRASG13D. The cell viability assay showed that KRASG13D cells were more sensitive to erianin than KRASWT or KRASG12V cells. In vitro, results showed that erianin suppressed the migration, invasion and epithelial-mesenchymal transition (EMT) of KRASG13D CRC cells. Furthermore, erianin induced ferroptosis, as evidenced by the accumulation of Fe2+ and reactive oxygen species (ROS), lipid peroxidation, and changes in the mitochondrial morphology of KRASG13D CRC cells. Interestingly, we also found that erianin-induced ferroptosis was accompanied by autophagy. Moreover, the occurrence of erianin-induced ferroptosis is reversed by autophagy inhibitors (NH4Cl and Bafilomycin A1) and ATG5 knockdown, suggesting that erianin-induced ferroptosis is autophagy-dependent. In addition, we evaluated the inhibition of tumor growth and metastasis by erianin in vivo using a subcutaneous tumor model and a spleen-liver metastasis model, respectively. Collectively, these data provide novel insights into the anticancer activity of erianin, which is valuable for the further discussion and investigation of the use of erianin in clinical anticancer chemotherapy for KRASG13D CRC.


Subject(s)
Colorectal Neoplasms , Ferroptosis , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Ferroptosis/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Mutation , Autophagy
3.
Front Bioeng Biotechnol ; 11: 1168827, 2023.
Article in English | MEDLINE | ID: mdl-37034255

ABSTRACT

Introduction: Radiation therapy has Q6long been a routine and effective treatment for non-small cell lung cancer (NSCLC), but the radioresistance and side effects have limited its application. In recent years, the superiority showed by trace element selenium in tumor radiotherapy sensitization has received wide attention. However, different forms of selenium compounds exhibit different chemical properties and their mechanisms of action on tumors may be different. Methods: Human non-small cell lung cancer SPC-A1 cells were studied. Drug toxicity was detected by MTT assay. The selenium content absorbed in vitro at different time points was detected by ICP-MS. Colony formation were conducted to observe the radiosensitization effect of different selenium compounds on SPC-A1 cells, and to compare the proliferation ability of SPC-A1 cells treated by radiation alone and radiation combined with different selenium compounds. Cell migration was detected by cell scratch assay. The changes of cell cycle and apoptosis were detected by flow cytometry. DCFH-DA fluorescent probe was used to detect the effects of different selenium compounds combined with X-ray on ROS production. Results: In this study, these four representative selenium compounds all have a certain ability to enhance the ability of radiotherapy to inhibit tumor cell proliferation and migration, and the mechanism may be related to blocking cell cycle in G2/M phase, activating the caspase cascade and reducing intracellular ROS levels to induce tumor cell apoptosis. Among them, -2-valent organic selenium has the most obvious effect, mainly inhibits cell migration, and induces early apoptosis by activating a large number of caspase-3, and arrest the cell cycle in S phase and G2/M phase. 0-valent selenium nanoparticles mainly arrest the cell cycle in G2/M phase. +4-valent inorganic selenium exerts its antitumor effects primarily by inhibiting tumor cell migration and inducing early apoptosis of tumor cells. Discussion: In this paper, the antitumor effects of four different forms of selenium compounds combined with X-rays on SPC-A1 cells were investigated, and their inhibitory effects on the proliferation and migration of cancer cells and their mechanisms were examined. We found that the radiosensitizing effect of selenium on NSCLC was closely related to its selenium form through the study of the sensitizing effect of different kinds of selenium compounds on radiotherapy.

4.
Life Sci ; 324: 121715, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37100377

ABSTRACT

AIMS: We aimed to evaluate the effect of periplocin on inhibiting hepatocellular carcinoma (HCC) and further determine its mechanisms. MAIN METHODS: Cytotoxic activity of periplocin against HCC cells was tested by CCK-8 and colony formation assays. The antitumor effects of periplocin were evaluated in human HCC SK-HEP-1 xenograft and murine HCC Hepa 1-6 allograft mouse models. Flow cytometry was used to measure cell cycle distribution, apopotosis, and the number of myeloid-derived suppressor cells (MDSCs). Hoechst 33258 dye was applied to observe the nuclear morphology. Network pharmacology was performed to predict possible signaling pathways. Drug affinity responsive target stability assay (DARTS) was used to evaluate AKT binding of periplocin. Western blotting, immunohistochemistry, and immunofluorescence were used to examine the protein expression levels. KEY FINDING: Periplocin inhibited cell viability with IC50 values from 50 nM to 300 nM in human HCC cells. Periplocin disrupted cell cycle distribution and promoted cell apoptosis. Moreover, AKT was predicted as the target of periplocin by network pharmacology, which was confirmed by that AKT/NF-κB signaling was inhibited in periplocin-treated HCC cells. Periplocin also inhibited the expression of CXCL1 and CXCL3, leading to decreased accumulation of MDSCs in HCC tumors. SIGNIFICANCE: These findings reveal the function of periplocin in inhibiting HCC progression by G2/M arrest, apoptosis and suppression of MDSCs accumulation through blockade of the AKT/NF-κB pathway. Our study further suggests that periplocin has the potential to be developed as an effective therapeutic agent for HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Myeloid-Derived Suppressor Cells , Humans , Mice , Animals , Carcinoma, Hepatocellular/pathology , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Liver Neoplasms/pathology , Myeloid-Derived Suppressor Cells/metabolism , Cell Proliferation , Apoptosis , Cell Line, Tumor
5.
Drug Des Devel Ther ; 17: 1007-1024, 2023.
Article in English | MEDLINE | ID: mdl-37025160

ABSTRACT

Background: Baicalin is an important active flavonoid isolated from the roots of Scutellaria baicalensis (S. baicalensis), a well-known traditional Chinese herb used in treating inflammatory bowel disease (IBD). The objectives of this study were to assess the potential benefit of baicalin in experimental colitis, as well as to investigate metabolic biomarkers of experimental colitis in conjunction with network pharmacology. Methods: Using a widely utilized network pharmacology technique, baicalin's targets and pathways were predicted. Simultaneously, experimental colitis was induced by intrarectal administration of TNBS. Histopathology examinations were performed to confirm pathological changes. Plasma samples were examined by using an untargeted metabolomics technique based on ultra-high performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS) to screen differential metabolites and associated metabolic pathways. Additionally, network pharmacology and integrated analysis of metabolomics were used to identify the primary targets. Results: Through network pharmacology research, tumor necrosis factor (TNF), interleukin 6 (IL6), serine/threonine-protein kinase (AKT1), and other 7 proteins were found to be the main targets of baicalin against IBD. The untargeted metabolomics results showed that 47 metabolites in glycerophospholipids and sphingolipid metabolism were involved as key pathways in the experimental colitis model group. 19 metabolites, including Sphingomyelin (SM d42:2, SM d42:1, SM d34:1), Lysophosphatidic acids (LPA 18:4), 1-Palmitoylglycerophosphocholine, and 17(18)-EpETE were demonstrated as key metabolites for baicalin to exert effects. Moreover, udp-glucose ceramide glucosyltransferase (UGCG), sphingomyelin synthase 1 (SGMS1), and sphingosine kinase (SPHK1) were predicted as sphingolipids-linked targets of baicalin against experimental colitis by integrative analysis. Conclusion: Based on these results, it implies that sphingolipid metabolism and sphingolipid signaling pathway might be acted as therapeutic mechanism for baicalin against experimental colitis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Colitis , Drugs, Chinese Herbal , Inflammatory Bowel Diseases , Humans , Colitis/drug therapy , Drugs, Chinese Herbal/pharmacology , Flavonoids/pharmacology , Flavonoids/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Metabolomics/methods , Network Pharmacology , Sphingolipids , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
7.
Front Pharmacol ; 12: 654611, 2021.
Article in English | MEDLINE | ID: mdl-33967797

ABSTRACT

Alzheimer's disease (AD) is one of the most common neurodegenerative diseases and accumulating evidences suggest a key role of amyloid-ß (Aß) peptide in the pathogenesis of AD. According to the amyloid cascade hypothesis, the imbalance of producing and clearing Aß is the beginning of neurodegeneration and dementia. Consequently, immunotherapy becomes popular through using antibodies against Aß. However, many studies of monoclonal antibodies were stopped because adverse effects appeared or there were no evident benefits observed. Some antibody fragments have many advantages over monoclonal antibodies, such as small sizes, lack of the crystallizable fraction (Fc) and so on. There are three main antibody fragments, including single chain variable fragments (scFvs), Fab fragments and single-domain antibody fragments. Nanoparticles can facilitate the entry of drug molecules across the blood-brain barrier, making them become excellent carriers. Various kinds of nanoparticles have been applied in the treatment of AD. The combination of nanoparticles and antibody fragments against amyloid-ß can be used in the diagnosis and treatment of Alzheimer's disease. In this review, we summarize the progress of antibody fragments against amyloid-ß in AD, focusing on the combined application with nanoparticles in the diagnosis and treatment of AD.

8.
Mater Sci Eng C Mater Biol Appl ; 121: 111872, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33579497

ABSTRACT

In this study, electrospun scaffolds were fabricated by blending poly(l-lactide-co-ε-caprolactone) (PLCL) and silk fibroin (SF) with different ratios, and further the feasibility of electrospun PLCL/SF scaffolds were evaluated for application of tissue engineered heart valve (TEHV). Scanning electron microscopy (SEM) results showed that the surface of PLCL/SF electrospun scaffolds was smooth and uniform while the mechanical properties were appropriate as valve prosthesis. In vitro cytocompatibility evaluation results demonstrated that all of the PLCL/SF electrospun scaffolds were cytocompatible and valvular interstitial cells (VICs) cultured on PLCL/SF scaffolds of 80/20 & 70/30 ratios exhibited the best cytocompatibility. The in vitro osteogenic differentiation of VICs including alkaline phosphatase (ALP) activity and quantitative polymerase chain reaction (qPCR) assays indicated that PLCL/SF scaffolds of 80/20 & 90/10 ratios behaved better anti-calcification ability. In the in vivo calcification evaluation model of rat subdermal implantation, PLCL/SF scaffolds of 80/20 & 90/10 ratios presented better anti-calcification ability, which was consistent with the in vitro results. Moreover, PLCL/SF scaffolds of 80/20 & 70/30 ratios showed significantly enhanced cell infiltration and M2 macrophage with higher CD206+/CD68+ ratio. Collectively, our data demonstrated that electrospun scaffolds with the PLCL/SF ratio of 80/20 hold great potential as TEHV materials.


Subject(s)
Aortic Valve Stenosis , Calcinosis , Fibroins , Polyesters , Animals , Aortic Valve , Caproates , Cells, Cultured , Dioxanes , Lactones , Osteogenesis , Rats , Tissue Engineering , Tissue Scaffolds
9.
Mol Pharm ; 18(3): 928-939, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33427470

ABSTRACT

Photodynamic therapy (PDT) is promising for clinical cancer therapy; however, the efficacy was limited as an individual treatment regimen. Here, an approach synergistically combining PDT and nitric oxide (NO) gas therapy along with destruction of the tumor extracellular matrix (ECM) was presented to eliminate cancer. Specifically, the NO donor l-arginine (l-Arg) and the photosensitizer indocyanine green (ICG) were co-encapsulated in poly(lactic-glycolic acid) (PLGA) nanoparticles and then loaded into the poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone) (PCL-PEG-PCL) hydrogel to develop an injectable, thermosensitive dual drug delivery system (PLGA@ICG@l-Arg/Gel). Significantly, reactive oxygen species (ROS) produced by PLGA@ICG@l-Arg/Gel under near-infrared (NIR) light irradiation could not only result in the apoptosis of cancer cells but also oxidize l-Arg to generate NO, which could suppress the proliferation of cancer cells. Moreover, ROS could further oxidize NO to generate peroxynitrite anions (ONOO-). ONOO- could activate matrix metalloproteinases (MMPs), which notably degraded collagen in ECM so as to damage the tumor microenvironment. PLGA@ICG@l-Arg/Gel significantly increased the antitumor efficacy against highly malignant 4T1 tumors in mice. Taken together, PLGA@ICG@l-Arg/Gel is a multifunctional platform that provides a novel strategy for cancer treatment with cascade amplification of the ROS oxidation effect, which holds great potential in clinical translation.


Subject(s)
Arginine/chemistry , Collagen/metabolism , Hydrogels/administration & dosage , Indocyanine Green/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Nitric Oxide/metabolism , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Delivery Systems/methods , Female , Hydrogels/chemistry , Mice , Mice, Inbred BALB C , Neoplasms/metabolism , Photochemotherapy/methods , Photosensitizing Agents/chemistry , Phototherapy/methods , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tumor Microenvironment/drug effects
10.
ACS Appl Bio Mater ; 3(4): 2275-2283, 2020 Apr 20.
Article in English | MEDLINE | ID: mdl-35025279

ABSTRACT

Glutaraldehyde (GA) was conventionally used to crosslink bovine pericardium to prepare bioprosthetic heart valves (BHVs), which usually fail within 10 years because of valve deterioration and calcification. To overcome the high cytotoxicity and severe calcification of GA-crosslinked BHVs, a quaternary ammonium salt of epoxy chitosan (epoxy group-modified 3-chlorine-2-hydroxypropyl trimethyl chitosan, abbreviated as "eHTCC") was developed to modify the acellular bovine pericardium to substitute GA and improve its anti-calcification and biocompatible properties. Mechanical test, enzymatic stability test, blood compatibility assay, and cytocompatibility assay were used to investigate its mechanical property and biocompatibility. The anti-calcification effect of the eHTCC-modified bovine pericardium (eHTCC-BP) was assessed by in vitro assay and rat subcutaneous implantation assay. The results showed that eHTCC-BP could improve the mechanical properties and anti-enzymolysis ability of BP, as well as retain the original three-dimensional structure, compared with the uncrosslinked-BP group. Moreover, the in vivo calcification level of the eHTCC-BP group was much lower than that of the GA-BP group, which was 5.1% (2 weeks), 2.3% (4 weeks), and 0.8% (8 weeks) of the GA-BP group. In summary, this study demonstrated that eHTCC could be a potential crosslinking agent for the extracellular matrix for its favorable crosslinking effects, anti-enzymolysis, anti-calcification, and biocompatibility.

11.
Biomater Sci ; 7(3): 914-925, 2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30511718

ABSTRACT

Small diameter vascular grafts have been promising substitutes for bypass surgery to treat cardiovascular disease. However, no ideal product is available in the clinic. In order to design improved, next generation vascular grafts, it is essential to understand the cellular and molecular mechanisms underlying tissue regeneration after vascular graft implantation. Two diverse microenvironments, circulating blood and the surrounding tissue, are involved in the regeneration process after vascular graft implantation in situ. However, their regenerative functions are not completely understood. To elucidate their roles in regeneration, we used electrospinning to fabricate four types of tubular scaffolds with a structure consisting of a microfiber layer (fiber diameter ∼ 6 µm) and a nanofiber layer (fiber diameter < 1 µm): microfiber scaffold, nanofiber scaffold, outer microfiber bilayer scaffold and inner microfiber bilayer scaffold. In the outer microfiber scaffold, cells from the surrounding tissue were allowed into the scaffold but not cells from the circulating blood while it was opposite in the inner microfiber scaffold. The processes of endothelium formation, smooth muscle cell regeneration, neo-tissue formation and vascularization of these scaffolds were analyzed with a rat left common carotid artery replacement model. Our data showed that smooth muscle cells' regeneration and vascularization were different among the four types of scaffolds. The thickest neo-tissue and α-SMA+ cell layers were detected in the microfiber scaffold group while the thinnest in the nanofiber scaffold group, and thicker neo-tissue and α-SMA+ cell layers were found in the outer microfiber bilayer scaffold group compared to the inner microfiber bilayer scaffold group. In addition, vascularization in the outer microfiber bilayer scaffold group and microfiber group was dramatically better than the inner microfiber bilayer scaffold group and the nanofiber group. Furthermore, we demonstrated that the regenerated SMCs were associated with the CD206+ macrophages in the graft wall. In all, the microfiber scaffold showed the best neo-tissue regeneration in vivo. These results indicate that the surrounding tissue contributes more to vascular regeneration than circulating blood. This finding gives a significant design clue that modulating the vascular surrounding tissue will be an alternative strategy for designing advanced and feasible small diameter vascular grafts.


Subject(s)
Myocytes, Smooth Muscle/physiology , Regeneration , Tissue Scaffolds/chemistry , Actins/metabolism , Animals , Blood Vessel Prosthesis , Carotid Arteries/surgery , Cell Movement , Endothelial Cells/cytology , Endothelial Cells/physiology , Macrophages/cytology , Macrophages/immunology , Macrophages/metabolism , Microscopy, Fluorescence , Myocytes, Smooth Muscle/cytology , Nanofibers/chemistry , Polyesters/chemistry , Rats , Tissue Engineering
12.
ACS Biomater Sci Eng ; 5(3): 1452-1461, 2019 Mar 11.
Article in English | MEDLINE | ID: mdl-33405620

ABSTRACT

In valvular replacement surgery, especially in the construction of bioprosthetic valves with decellularized pericardial xenograft, glutaraldehyde (GA) is routinely utilized as the golden standard reagent to fix bovine or porcine pericardial tissues. However, the apparent defects of GA, including cytotoxicity and calcification, increase the probability of leaflet failure and motivate the exploration for alternatives. Thus, the aim of this study is to develop nonglutaraldehyde combined-cross-linking reagents composed of alginate-EDC/NHS (Alg) or oxidized alginate-EDC/NHS (Alg-CHO) as substitute for GA, which is confirmed to be less toxic and more biocompatible. Evaluations of the fixed acellular bovine pericardial tissues included mechanical performance, thermodynamics/enzymatic/in vivo stability tests, blood compatibility assay, cytocompatibility assay, in vitro anticalcification, and in vivo anticalcification assay by subcutaneous implantation in juvenile Wistar rats. The data revealed that the tissues fixed with the combined cross-linking reagents were superior to GA control and commercially available Sino product in terms of better in vitro hemocompatibility and cytocompatibility, lower calcification levels, better thermodynamics stability, and better regenerative capacity in subcutaneous implants, while the mechanical strength and in vivo stability were comparable. Considering all above performances, it indicated that both Alg and Alg-CHO are appropriate to replace GA as the cross-linkers for biological tissue, particularly as a nonglutaraldehyde fixation for off the shelf decellularized bovine pericardial tissue in the anticalcification cardiac valve applications. Nevertheless, studies on the long-term durability and calcification-resistance capacity in large animal model are further needed.

13.
J Pharmacol Sci ; 137(3): 305-312, 2018 Jul.
Article in English | MEDLINE | ID: mdl-30119963

ABSTRACT

The aberrant expression of cancerous inhibitor of protein phosphatase 2A (CIP2A) indicates poor prognosis and promotes EMT and metastasis. EMT, a crucial cellular process that occurs during cancer progression and metastasis, has been reported to promote drug resistance in several previous studies. Consequently, ongoing research has been focused on exploring therapeutic options for preventing EMT to delay or reverse drug resistance. Polyphyllin I (PPI) is a natural component extracted from Paris polyphylla that displays anti-cancer properties. In the present study, we investigated whether PPI can be used in the cisplatin (DDP)-resistant human gastric cancer cell line SGC7901/DDP. The results demonstrated that PPI treatment significantly inhibited cell proliferation, invasion and EMT. TGF-ß1 is known to promote EMT-induced metastasis in numerous tumor types. PPI inhibited the invasion of TGFß1-induced SGC7901/DDP cells in vitro. PPI also increased the mRNA and protein expression levels of E-cadherin but decreased the expression levels of vimentin. Further examination of the mechanism revealed that the CIP2A/PP2A/Akt pathway is partially involved in this regulation of EMT-related biomarkers and invasion. Furthermore, xenograft tests also confirmed the antitumor effects of PPI in vivo. We propose that PPI could be developed as a candidate drug for treating cancer invasion and migration.


Subject(s)
Antineoplastic Agents, Phytogenic , Autoantigens/genetics , Autoantigens/metabolism , Cell Proliferation/drug effects , Cisplatin/pharmacology , Diosgenin/analogs & derivatives , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Metastasis/genetics , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Depression, Chemical , Diosgenin/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression/drug effects , Gene Expression/genetics , Humans , Intracellular Signaling Peptides and Proteins , Neoplasm Invasiveness/prevention & control , Transforming Growth Factor beta1/physiology , Vimentin/genetics , Vimentin/metabolism
14.
Int J Oncol ; 53(3): 1279-1288, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29956727

ABSTRACT

Polyphyllin I (PPI) is a natural compound extracted from the rhizomes of Paris polyphylla and has been used to treat fevers and headaches in China. In the present study, the antitumor activity of PPI in prostate cancer (PC) cells was evaluated. At low doses, PPI decreased proliferation, invasion and epithelial-mesenchymal transition (EMT) in PC cells. PPI decreased the expression of matrix metalloproteinase 7 (MMP7), an enzyme that is critical for tumor metastasis. PPI also decreased the expression of Snail and vimentin, which are EMT-associated factors. Additionally, PPI suppressed AP-1 transcriptional activity and AP-1 binding to the MMP7 and vimentin promoters. The results demonstrated that PPI downregulated the phosphorylation of extracellular signaling­related kinase (ERK), which is upstream modulator of AP-1. The results of the present study demonstrated that PPI may inhibit the cancerous inhibitor of protein phosphatase 2A (CIP2A)/protein phosphatase 2A (PP2A)/ERK axis, downregulate the expression of MMP7, vimentin, and Snail, and suppress tumor invasion and EMT. A PC xenograft mouse model was employed and the results revealed that PPI may decrease tumor growth and weight. Additionally, PPI may inhibit proliferating cell nuclear antigen expression and CIP2A/PP2A/ERK signaling pathway in PPI-treated tumors. Therefore, the results of the present study suggest that PPI may suppress the growth, invasion and EMT of PC cells via inhibition of CIP2A/PP2A/ERK signaling axis. As a result, PPI may be a novel target for the treatment of PC.


Subject(s)
Antineoplastic Agents/pharmacology , Diosgenin/analogs & derivatives , Epithelial-Mesenchymal Transition/drug effects , Prostatic Neoplasms/drug therapy , Signal Transduction/drug effects , Animals , Antineoplastic Agents/therapeutic use , Autoantigens/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Diosgenin/pharmacology , Diosgenin/therapeutic use , Down-Regulation , Drug Evaluation, Preclinical , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Male , Melanthiaceae/chemistry , Membrane Proteins/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness/prevention & control , Phosphorylation/drug effects , Prostatic Neoplasms/pathology , Protein Phosphatase 2/metabolism , Xenograft Model Antitumor Assays
15.
ACS Appl Mater Interfaces ; 10(24): 20315-20325, 2018 Jun 20.
Article in English | MEDLINE | ID: mdl-29808993

ABSTRACT

The combination of an antigen and adjuvant has synergistic effects on an immune response. Coadministration of an antigen and a granulocyte-macrophage colony-stimulating factor (GM-CSF) hydrogel delivery system will afford a novel strategy for enhancement of an immune response because of the dual role of the hydrogel as a vaccine carrier with a sustained release and a platform for recruiting dendritic cells (DCs). Herein, an injectable poly(caprolactone)-poly(ethylene glycol)-poly(caprolactone) thermosensitive hydrogel coencapsulating GM-CSF and ovalbumin nanoparticles was developed to enhance antigen uptake efficiency. The GM-CSF released from the hydrogel ensured accumulation of DCs; this effect improved the antigen uptake efficiency with the targeted delivery to antigen-presenting cells. Furthermore, the dual delivery system induced a stronger immune effect, including higher CD8+ T proportion, interferon γ secretion, and a greater cytotoxic T lymphocyte response, which may benefit from the recruitment of DCs, increasing antigen residence time, and the controllable antigen release owing to the combined effect of the hydrogel and nanoparticles. Meanwhile, the real-time antigen delivery process in vivo was revealed by a noninvasive fluorescence imaging method. All of the results indicated that the visible dual delivery system may have a greater potential for the efficient and trackable vaccine delivery.


Subject(s)
Nanoparticles , Animals , Dendritic Cells , Granulocyte-Macrophage Colony-Stimulating Factor , Hydrogels , Ovalbumin
16.
Nanomedicine ; 14(4): 1087-1098, 2018 06.
Article in English | MEDLINE | ID: mdl-29474923

ABSTRACT

Accurate and efficient antigen delivery is crucial for inducing a strong and long-term immune response. A visible protein nanovaccine made from antigen could provide a novel and promising technology for secure and efficient delivery of the antigen with imaging visualization. In this study, a functional nanovaccine based on genipin crosslinked ovalbumin (OVA) fluorescent nanoparticles with chitosan (CS-OVA-NPs) was developed. The nanovaccine can carry abundant antigens by self-crosslinking without additional carriers. The fluorescence imaging technique was applied to monitor and reveal the process of antigen delivery in vivo based on the fluorescence of genipin with a non-invasive and real-time manner. This functional OVA nanovaccine can enhance the uptake of OVA in Dendritic Cells (DCs) and further promote DCs to maturate in vitro. In vivo study further indicated CS-OVA-NPs could trigger antigen-specific immune responses, which demonstrated that this fluorescent nanovaccine provided a novel design approach for accurate and efficient vaccine delivery.


Subject(s)
Iridoids/chemistry , Nanoparticles/chemistry , Ovalbumin/chemistry , Vaccines/chemistry , Animals , Cell Survival , Cells, Cultured , Chitosan/chemistry , Dendritic Cells/cytology , Dendritic Cells/metabolism , Drug Delivery Systems/methods , Male , Mice , Mice, Inbred C57BL , Ovalbumin/metabolism
17.
Oncol Rep ; 38(3): 1733-1741, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28677734

ABSTRACT

SE translocation (SET) oncoprotein, an inhibitor of protein phosphatase 2A, is abnormally expressed in many cancers. In this study, SET was aberrantly upregulated in gastric cancer (GC) compared with control tissues. Clinicopathological analysis showed that SET expression was significantly correlated with pathological grade (p=0.002), lymph node stage (p=0.014), and invasive depth (p=0.022). Kaplan-Meier analysis indicated that patients with high SET expression showed poorer overall survival rates than those with low SET expression. Moreover, SET knockdown downregulated GC cell proliferation, colony formation, tumorigenesis, and metastasis. The biological effect of SET on proliferation and invasion was mediated by inhibition of protein phosphatase 2, which in turn, activated Akt. Taken together, our results suggested that SET overexpression is associated with GC progression, and it might be a potential diagnostic marker for GC, thereby a possible target for GC drug development.


Subject(s)
Histone Chaperones/genetics , Oncogene Proteins/genetics , Stomach Neoplasms/genetics , Transcription Factors/genetics , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , DNA-Binding Proteins , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis/genetics , Lymphatic Metastasis/pathology , Male , Mice , Mice, Nude , Middle Aged , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Prognosis , Protein Phosphatase 2/genetics , Stomach Neoplasms/pathology , Survival Rate , Translocation, Genetic/genetics , Up-Regulation/genetics
18.
Nanomedicine ; 13(7): 2271-2280, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28712919

ABSTRACT

Chemotherapy and photothermal therapy can be efficiently integrated to achieve enhanced antitumor efficacy by using carbon nanotubes (CNTs) which are super in delivering drug and converting near infrared radiation (NIR) into heat. We previously developed an innovative TAT-chitosan functionalized MWCNTs (MWCNTs/TC) based drug delivery system for doxorubicin (DOX) and preliminarily investigated its release profile and antitumor effect. In the present study, the application potential of MWCNTs/DOX/TC in chemo-photothermal combination therapy was further explored. The in vitro drug release, photothermal effect, cellular uptake and cytotoxicity were assessed. The in vivo anti-tumor effect of MWCNTs/DOX/TC was further evaluated by noninvasive bioluminescence imaging. It was demonstrated that this innovative drug delivery system not only realized a conspicuously sustained release of DOX, but also retained the optical properties of MWCNTs for a high photothermal effect upon NIR irradiation, and exhibited remarkably enhanced anti-tumor efficacy through the synergistic function of chemotherapy and photothermal ablation.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Chitosan/analogs & derivatives , Doxorubicin/therapeutic use , Nanotubes, Carbon/chemistry , Neoplasms/therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Cell Line, Tumor , Chitosan/therapeutic use , Combined Modality Therapy/methods , Doxorubicin/administration & dosage , Drug Delivery Systems , Drug Liberation , Hyperthermia, Induced/methods , Male , Mice, Inbred BALB C , Mice, Nude , Nanotubes, Carbon/ultrastructure , Phototherapy/methods
19.
Oncol Rep ; 38(1): 598-606, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28560452

ABSTRACT

We have shown that a novel STAT3 inhibitor arctigenin (Atn) induces significant cytotoxicity in triple-negative breast cancer (TNBC) cells. This study further delineated molecular mechanisms where by Atn triggered cytotoxicity in TNBC cells. We found Atn can also inhibit metastasis in TNBC cells through cancerous inhibitor of protein phosphatase 2A (CIP2A) pathway. CIP2A is an endogenous inhibitor of protein phosphatase 2A (PP2A), which can increase the migration and invasion of various cancer cells. PP2A is a tumor suppressor, which is functionally defective in various cancers. Atn-induced metastasis inhibition was associated with reactivation of PP2A, downregulation of CIP2A and Akt phosphorylation. Silencing CIP2A enhanced Atn-induced metastasis inhibition and apoptosis in TNBCs. Furthermore, ectopic expression of CIP2A or inhibition of PP2A in TNBC cells abolished the effects of Atn. In conclusion, we found that enhancement of PP2A activity by inhibition of CIP2A, at least in part, promotes the anti-metastasis effect induced by Atn. Our findings disclose the novel therapeutic mechanism of this targeted agent, and suggest the therapeutic potential and feasibility of developing PP2A enhancers as a novel anticancer strategy.


Subject(s)
Antineoplastic Agents/pharmacology , Autoantigens/metabolism , Furans/pharmacology , Lignans/pharmacology , Membrane Proteins/metabolism , Protein Phosphatase 2/metabolism , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Apoptosis , Autoantigens/genetics , Cell Line, Tumor , Down-Regulation , Furans/therapeutic use , Humans , Intracellular Signaling Peptides and Proteins , Lignans/therapeutic use , Membrane Proteins/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/pathology
20.
Drug Deliv ; 24(1): 143-151, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28156171

ABSTRACT

Monitoring drug release and therapeutic efficacy is crucial for developing drug delivery systems. Our preliminary study demonstrated that, as compared with pristine multiwalled carbon nanotubes (MWCNTs), transactivator of transcription (TAT)-chitosan functionalized MWCNTs (MWCNTs-TC) were a more promising candidate for drug delivery in cancer therapy. In the present study, a MWCNTs/TC-based drug delivery system was developed for an anticancer drug, doxorubicin (DOX). The drug loading and in vitro release profiles, cellular uptake and cytotoxicity were assessed. More importantly, the in vivo drug release and antitumor effect of MWCNTs/DOX/TC were evaluated by noninvasive fluorescence and bioluminescence imaging. It was demonstrated that MWCNTs/DOX/TC can be efficiently taken up by BEL-7402 hepatoma cells. The release of DOX from MWCNTs/DOX/TC was faster under lower pH condition, which was beneficial for intrcellular drug release. The in vivo release process of DOX and antitumor effect in animal model were monitored simultaneously by noninvasive fluorescence and luminescence imaging, which demonstrated the application potential of MWCNTs/DOX/TC for cancer therapy.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Drug Liberation/physiology , Nanotubes, Carbon/chemistry , Animals , Cell Line, Tumor , Chitosan/chemistry , Drug Delivery Systems/methods , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL
...