Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
Add more filters










Publication year range
1.
Bioconjug Chem ; 29(9): 3104-3112, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30105903

ABSTRACT

Depending on increasing extracellular protein utilization and altering metabolic programs, cancer cells could proliferate and survive without restricion by ingesting human serum albumin (HSA) to serve as nutritional amino acids. Here, we hypothesize that the consumption of albumin by cancer cells could be utilized as an efficient approach to targeted drug delivery. Lidamycin (LDM), an antitumor antibiotic with extremely potent cytotoxicity to cultured cancer cells, consists of an apoprotein (LDP) and an active enediyne chromophore (AE). In the present study, a novel albumin-lidamycin conjugate was prepared by DNA recombination and molecular reconstitution. Results show that the IC50 values of albumin-lidamycin conjugate (HSA-LDP-AE) for a variety of tested cancer cells were at subnanomolar levels. At tolerated doses, the albumin-lidamycin conjugate significantly inhibited the growth of lung carcinoma PG-BE1 xenografts by 97.8%. The therapeutic efficacy of the albumin-lidamycin conjugate was much stronger than that of free lidamycin. Meanwhile, the images of albumin-lidamycin conjugate showed obvious and lasting tumor localization and fluorescence enrichment and there was no detectable signal in nontumor locations. Taken together, albumin-lidamycin conjugate, a new format of lidamycin, could be a promising antitumor therapeutic agent and albumin-integration might be a feasible approach to targeted antitumor drug delivery.


Subject(s)
Albumins/chemistry , Aminoglycosides/chemistry , Aminoglycosides/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Enediynes/chemistry , Enediynes/therapeutic use , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor , Female , Humans , Mice , Optical Imaging , Xenograft Model Antitumor Assays
2.
Acta Pharmacol Sin ; 39(11): 1777-1786, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30013033

ABSTRACT

Defensins play an essential role in innate immunity. In this study, a novel recombinant ß-defensin that targets the epidermal growth factor receptor (EGFR) was designed and prepared. The EGFR-targeting ß-defensin consists of an EGF-derived oligopeptide (Ec), a ß-defensin-1 peptide (hBD1) and a lidamycin-derived apoprotein (LDP), which serves as the "scaffold" for the fusion protein (Ec-LDP-hBD1). Ec-LDP-hBD1 effectively bound to EGFR highly expressed human epidermoid carcinoma A431 cells. The cytotoxicity of Ec-LDP-hBD1 to EGFR highly expressed A431 cells was more potent than that to EGFR low-expressed human lung carcinoma A549 and H460 cells (the IC50 values in A431, A549, and H460 cells were 1.8 ± 0.55, 11.9 ± 0.51, and 5.19 ± 1.21 µmol/L, respectively); in addition, the cytotoxicity of Ec-LDP-hBD1 was much stronger than that of Ec-LDP and hBD1. Moreover, Ec-LDP-hBD1 suppressed cancer cell proliferation and induced mitochondria-mediated apoptosis. Its in vivo anticancer action was evaluated in athymic mice with A431 and H460 xenografts. The mice were administered Ec-LDP-hBD1 (5, 10 mg/kg, i.v.) two times with a weekly interval. Administration of Ec-LDP-hBD1 markedly inhibited the tumor growth without significant body weight changes. The in vivo imaging further revealed that Ec-LDP-hBD1 had a tumor-specific distribution with a clear image of localization. The results demonstrate that the novel recombinant EGFR-targeting ß-defensin Ec-LDP-hBD1 displays both selectivity and enhanced cytotoxicity against relevant cancer cells by inducing mitochondria-mediated apoptosis and exhibits high therapeutic efficacy against the EGFR-expressed carcinoma xenograft. This novel format of ß-defensin, which induces mitochondrial-mediated apoptosis, may play an active role in EGFR-targeting cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Mitochondria/metabolism , Recombinant Fusion Proteins/therapeutic use , beta-Defensins/therapeutic use , Aminoglycosides/metabolism , Aminoglycosides/therapeutic use , Animals , Antineoplastic Agents/metabolism , Apoproteins/metabolism , Apoproteins/therapeutic use , Cell Line, Tumor , Enediynes/metabolism , Enediynes/therapeutic use , ErbB Receptors/metabolism , Female , Humans , Mice, Nude , Mitochondria/pathology , Protein Binding , Recombinant Fusion Proteins/metabolism , Xenograft Model Antitumor Assays , beta-Defensins/metabolism
3.
Pharmacol Res ; 126: 66-76, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28392461

ABSTRACT

Overexpression of EGFR and MMP-2 plays an essential role in the initiation and progression of non-small-cell lung carcinoma (NSCLC). In this study, a novel format of EGFR/MMP-2 bi-targeted fusion protein Ec-LDP-TIMP2 and its enediyne-integrated analogue Ec-LDP(AE)-TIMP2 have been prepared by genetic engineering and molecular reconstitution. The Ec-LDP(AE)-TIMP2 comprises endogenous inhibitor of matrix metalloproteinase 2 (TIMP2), EGF-derived oligopeptide (Ec), lidamycin apoprotein (LDP), and the extremely potent cytotoxic enediyne (AE). By tissue microarray, Ec-LDP-TIMP2 showed high binding intensity and selectivity to human NSCLC specimens as compared with the matched non-cancerous tissues. By in vivo imaging, Ec-LDP-TIMP2 displayed prominent tumor-specific distribution in human NSCLC H460 xenograft. Particularly, Ec-LDP(AE)-TIMP2 inhibited tumor growth of H460 xenograft in athymic mice more striking. At doses of 0.2 and 0.4mg/kg, Ec-LDP(AE)-TIMP2 suppressed tumor growth by 74% and 89%, respectively. No histopathological changes were found in various organs of treated animals, suggesting that the effective dosage was tolerated. In summary, the ligand-based and enediyne-integrated fusion protein displaying extremely potent cytotoxicity might be highly effective for NSCLC therapy and useful as a carrier for drug delivery.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Matrix Metalloproteinase 2/metabolism , Recombinant Fusion Proteins/pharmacology , Tissue Inhibitor of Metalloproteinase-2/pharmacology , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Female , Humans , Ligands , Lung Neoplasms/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Mice , Mice, Inbred BALB C , Mice, Nude , Oligopeptides/pharmacology , Xenograft Model Antitumor Assays/methods
4.
Huan Jing Ke Xue ; 38(6): 2419-2428, 2017 Jun 08.
Article in Chinese | MEDLINE | ID: mdl-29965361

ABSTRACT

As an effective technology for wastewater treatment, bio-filter has been widely used. Nevertheless, there is still a lack of systematic report on purification efficiency and influencing factors of combined bio-filters. To this end, a novel combined system that consisted of aerated vertical-flow filter (AVF) followed by baffled horizontal-flow filter (BHF) was designed. After setting a series of hydraulic loading rates (131, 94 and 60 mm·d-1) and diversion ratios (8:2、6:4、4:6), we comprehensively assessed the impact of running condition adjustment on treatment performance by multiple statistical analyses. The results showed that, the average removal rates of organic matter, ammonia nitrogen and dissolved nitrogen in AVF were all above 80%, while the average removal rates of ammonia nitrogen, total nitrogen and dissolved nitrogen in BHF were all below 40%. Different running conditions had a significant (P<0.05) impact on treatment performance. Meanwhile, there were significant differences in purification efficiency between the two different kinds of filters. Oxidative degradation was one of the main ways to remove organic matter in the two kinds of filters. There were obvious nitrification and denitrification processes within the two kinds of filters. Nitrification followed by denitrification was the main way to remove total nitrogen since ammonium occupied the most portion of total nitrogen in the synthetic wastewater. Meanwhile, the intensity of nitrification and denitrification in AVF was obviously higher than that in BHF. Phosphorus removal was mainly controlled by hydraulic loading rate, temperature, dissolved oxygen, organic matter, etc. This might indicate that microbial absorption was one of the main ways to remove phosphorus for the two filters. Compared to the sole AVF, the removal of total organic matter and total phosphorus in the combined system was increased by 4.4% and 23.2%, respectively, but the removal of total nitrogen was reduced by 12.1%. Reducing the diversion ratio was helpful to improve the denitrification intensity in BHF. However, due to the introduction of excessive ammonia from the raw wastewater, as well as the limited nitrification capacity in BHF, the removal rate of total nitrogen for the combined system was decreased. Therefore, according to the composition of treated raw wastewater, the control of appropriate diversion ratio, residence time and redox conditions inside the filter bed was the key to enhance the overall performance of the combined system.


Subject(s)
Aquaculture , Denitrification , Nitrification , Waste Disposal, Fluid , Water Purification , Ammonia/isolation & purification , Bioreactors , Filtration , Nitrogen/isolation & purification , Wastewater
5.
Oncotarget ; 7(36): 58418-58434, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27517152

ABSTRACT

K-Ras mutant pancreatic cancer cells display intensive macropinocytosis, indicating that this process may be exploited in the design of anticancer targeted therapies. In this study, we constructed a macropinocytosis-oriented recombinantly tailored defensin (DF-HSA) which consists of human ß-defensin-2 (DF) and human serum albumin (HSA). The macropinocytosis intensity and cytotoxicity of DF-HSA were investigated in K-Ras mutant MIA PaCa-2 cells and wild-type BxPC-3 cells. As found, the DF-HSA uptake in MIA PaCa-2 cells was much higher than that in wild-type BxPC-3 cells. Correspondingly, the cytotoxicity of DF-HSA to MIA PaCa-2 cells was more potent than that to BxPC-3 cells. In addition, the cytotoxicity of DF-HSA was much stronger than that of ß-defensin HBD2. DF-HSA suppressed cancer cell proliferation and induced mitochondrial pathway apoptosis. Notably, DF-HSA significantly inhibited the growth of human pancreatic carcinoma MIA PaCa-2 xenograft in athymic mice at well tolerated dose. By in vivo imaging, DF-HSA displayed a prominent accumulation in the tumor. The study indicates that the recombinantly tailored ß-defensin can intensively enter into the K-Ras mutant pancreatic cancer cells through macropinocytosis-mediated process and exert potent therapeutic efficacy against the pancreatic carcinoma xenograft. The novel format of ß-defensin may play an active role in macropinocytosis-mediated targeting therapy.


Subject(s)
Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pinocytosis , Proto-Oncogene Proteins p21(ras)/genetics , Serum Albumin, Human/metabolism , beta-Defensins/metabolism , A549 Cells , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Female , Genes, ras , Genetic Vectors , Humans , Membrane Potential, Mitochondrial , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Neoplasm Transplantation , Reactive Oxygen Species/metabolism , Recombinant Proteins/metabolism
6.
Oncotarget ; 6(28): 26322-34, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26314845

ABSTRACT

Recent studies have shown that MMP-14 is highly expressed in a panel of human solid tumors and poses as a potential molecular target for anticancer drugs. Currently, major strategies for targeted therapeutics have mainly focused on the use of antibody or ligand-based agents. For seeking an alternative approach, it is of interest to employ endogenous proteins as drug delivery carriers. Considering the facts that TIMP2, the tissue inhibitor of metalloproteinase 2, shows specific interaction with MMP-14 and that Lidamycin (LDM), an extremely potent cytotoxic antitumor antibiotic, consists of an apoprotein (LDP) and a highly active enediyne (AE); we designed and prepared a TIMP2-based and enediyne-integrated fusion protein LDP(AE)-TIMP2 by DNA recombination and molecular reconstitution consecutively. Furthermore, the MMP-14 binding attributes of the active fusion protein were determined and its therapeutic efficacy against human esophageal carcinoma KYSE150 xenograft and human fibrosarcoma HT1080 xenograft models in nude mice was investigated. It is suggested that TIMP2, the endogenous and MMP-14 binding protein, might serve as a guided carrier for targeted therapeutics.


Subject(s)
Aminoglycosides/pharmacology , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Enediynes/pharmacology , Esophageal Neoplasms/drug therapy , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Protein Engineering , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Aminoglycosides/biosynthesis , Aminoglycosides/genetics , Angiogenesis Inhibitors/pharmacology , Animals , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Design , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/pathology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Mice, Nude , Molecular Targeted Therapy , Neovascularization, Physiologic/drug effects , Protein Binding , Recombinant Fusion Proteins/pharmacology , Time Factors , Tissue Inhibitor of Metalloproteinase-2/biosynthesis , Tissue Inhibitor of Metalloproteinase-2/genetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
7.
Acta Pharmacol Sin ; 36(4): 473-82, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25832428

ABSTRACT

AIM: (-)-Epigallocatechin-3-gallate (EGCG) is one of the most abundant polyphenols in green tea with strong antioxidant activity and various therapeutic effects. In this study, we investigated the anti-fibrotic effects of EGCG and underlying mechanisms in bile duct-ligated (BDL) rats and a liver fibrosis model in vitro. METHODS: BDL rats were treated with EGCG (25 mg·kg(-1)·d(-1), po) for 14 d, and then the serum, bile and liver samples were collected. Liver fibrosis was assessed by serum, urine and bile biochemistry analyses and morphological studies of liver tissues. TGF-ß1-stimulated human hepatic stellate LX-2 cells were used as a liver fibrosis model in vitro. The expression of liver fibrogenic genes and signaling proteins in the PI3K/Akt/Smad pathway was examined using Western blotting and/or real-time PCR. RESULTS: In BDL rats, EGCG treatment significantly ameliorates liver necrosis, inflammation and fibrosis, and suppressed expression of the genes associated with liver inflammation and fibrogenesis, including TNF-α, IL-1ß, TGF-ß1, MMP-9, α-SMA, and COL1A1. In LX-2 cells, application of EGCG (10, 25 µmol/L) dose-dependently suppressed TGF-ß1-stimulated expression of COL1A1, MMP-2, MMP-9, TGF-ß1, TIMP1, and α-SMA. Furthermore, EGCG significantly suppressed the phosphorylation of Smad2/3 and Akt in the livers of BDL rats and in TGF-ß1-stimulated LX-2 cells. Application of LY294002, a specific inhibitor of PI3K, produced similar effects as EGCG did in TGF-ß1-stimulated LX-2 cells, but co-application of EGCG and LY294002 did not produce additive effects. CONCLUSION: EGCG exerts anti-fibrotic effects in BDL rats and TGF-ß1-stimulated LX-2 cells in vitro via inhibiting the PI3K/Akt/Smad pathway.


Subject(s)
Catechin/analogs & derivatives , Cholestasis/complications , Liver Cirrhosis/complications , Liver Cirrhosis/drug therapy , Liver/drug effects , Liver/pathology , Signal Transduction/drug effects , Animals , Bile Ducts/drug effects , Bile Ducts/metabolism , Bile Ducts/pathology , Catechin/therapeutic use , Cell Line , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Smad Proteins/metabolism
8.
Cancer Immunol Immunother ; 63(12): 1261-72, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25164878

ABSTRACT

Tuftsin (TF) is an immunomodulator tetrapeptide (Thr-Lys-Pro-Arg) that binds to the receptor neuropilin-1 (Nrp1) on the surface of cells. Many reports have described anti-tumor activity of tuftsin to relate with nonspecific activation of the host immune system. Lidamycin (LDM) that displays extremely potent cytotoxicity to cancer cells is composed of an apoprotein (LDP) and an enediyne chromophore (AE). In addition, Ec is an EGFR-targeting oligopeptide. In the present study, LDP was used as protein scaffold and the specific carrier for the highly potent AE. Genetically engineered fusion proteins LDP-TF and Ec-LDP-TF were prepared; then, the enediyne-energized fusion protein Ec-LDM-TF was generated by integration of AE into Ec-LDP-TF. The tuftsin-based fusion proteins LDP-TF and Ec-LDP-TF significantly enhanced the phagocytotic activity of macrophages as compared with LDP (P < 0.05). Ec-LDP-TF effectively bound to tumor cells and macrophages; furthermore, it markedly suppressed the growth of human epidermoid carcinoma A431 xenograft in athymic mice by 84.2 % (P < 0.05) with up-regulated expression of TNF-α and IFN-γ. Ec-LDM-TF further augmented the therapeutic efficacy, inhibiting the growth of A431 xenograft by 90.9 % (P < 0.05); notably, the Ec-LDM-TF caused marked down-regulation of CD47 in A431 cells. Moreover, the best therapeutic effect was recorded in the group of animals treated with the combination of Ec-LDP-TF with Ec-LDM-TF. The results suggest that tuftsin-based, enediyne-energized, and EGFR-targeting fusion proteins exert highly antitumor efficacy with CD47 modulation. Tuftsin-based fusion proteins are potentially useful for treatment of EGFR- and CD47-overexpressing cancers.


Subject(s)
CD47 Antigen/immunology , Enediynes/pharmacology , ErbB Receptors/immunology , Immunotoxins/pharmacology , Recombinant Fusion Proteins/pharmacology , Tuftsin/pharmacology , Animals , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/therapy , Cell Communication , Cell Line, Tumor , Down-Regulation , Enediynes/chemistry , Female , Humans , Immunotoxins/chemistry , Immunotoxins/immunology , Mice , Mice, Inbred BALB C , Murine pneumonia virus , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/immunology , Tuftsin/chemistry , Tuftsin/immunology , Xenograft Model Antitumor Assays
9.
J Neurooncol ; 119(1): 91-100, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24842385

ABSTRACT

The present work evaluated the synergistic efficacy of an enediyne antibiotic lidamycin (LDM) plus temozolomide (TMZ) against glioma in vitro and in vivo. LDM plus TMZ inhibited the proliferations of rat glioma C6 cells and human glioma U87 cells more efficiently than the single usage of LDM or TMZ. In addition, LDM also potentiated the apoptosis inductions by TMZ in rat C6 cells and human U87 cells. Meanwhile, the results of TdT-mediated dUTP Nick End Labeling assay for subcutaneous U87 tumor sections indicated an enhanced apoptosis induction in vivo by LDM plus TMZ, which confirmed the high potency of the combination for glioma therapy. As determined by Western blot, apoptosis signal pathways in C6 cells and U87 cells were markedly affected by the synergistic alteration of P53, bax, procaspase 3, and bcd-2 expression. In both subcutaneous U87 xenograft and C6 intracerebral orthotopic implant model, TMZ-induced glioma growth suppression was dramatically potentiated by LDM. As shown, the combination therapy efficiently reduced the tumor volumes and tumor weights of the human glioma U87 xenograft. Kaplan-Meier assay revealed that LDM plus TMZ dramatically prolonged the life span of C6 intracerebral tumor-bearing rats with decreased tumor size. This study indicates that the combination of LDM with TMZ might be a promising strategy for glioma therapy.


Subject(s)
Aminoglycosides/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Enediynes/therapeutic use , Glioma/drug therapy , Aminoglycosides/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Synergism , Enediynes/pharmacology , Glioma/pathology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Rats , Signal Transduction/drug effects , Temozolomide
10.
Yao Xue Xue Bao ; 49(2): 198-203, 2014 Feb.
Article in Chinese | MEDLINE | ID: mdl-24761609

ABSTRACT

This study aimed to investigate the synergistic effect of lidamycin (LDM) and rituximab on human B cell lymphoma Ramos cells. Cell proliferation was measured using MTS assay, cell apoptosis was analyzed by Annexin V-FITC/PI assay, the expression of apoptosis related proteins was analyzed by Western blotting, and the in vivo lymphoma inhibition was verified using BALB/c mice inoculated via tail vein using Ramos cells which stably expressed pEGFP-N1 plasmid. The results showed that, after the pretreatment with rituximab for 48 h, rituximab and LDM showed significantly synergistic effects on cell proliferation. Cells in combined treatment group had a higher apoptosis rate than that in LDM treatment group. Compared with the LDM treatment group, the expression of apoptosis-related proteins such as Cleaved caspase-3, Cleaved caspase-7, Cleaved caspase-9 and Cleaved PARP in combined treatment groups increased, and expression of cIAP-2 and Bcl-2 decreased. The result of in vivo experiment showed that, in the combined treatment group, the survival time of BALB/c mice was significantly longer than the mice in control group and LDM treatment group, and the degree of tumor accumulation and metastasis to lymph nodes and spleen was lower.


Subject(s)
Aminoglycosides/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Enediynes/pharmacology , Lymphoma, B-Cell/pathology , Rituximab/pharmacology , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Caspase 3/metabolism , Caspase 7/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Drug Synergism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Random Allocation
11.
PLoS One ; 9(3): e92986, 2014.
Article in English | MEDLINE | ID: mdl-24664246

ABSTRACT

Esophageal cancer is one of the most common cancers, and the 5-year survival rate is less than 10% due to lack of effective therapeutic agents. This study was to evaluate antitumor activity of Ec-LDP-Hr-AE, a recently developed bispecific enediyne-energized fusion protein targeting both epidermal growth factor receptor (EGFR) and epidermal growth factor receptor 2 (HER2), on esophageal cancer. The fusion protein Ec-LDP-Hr-AE consists of two oligopeptide ligands and an enediyne antibiotic lidamycin (LDM) for receptor binding and cell killing, respectively. The current study demonstrated that Ec-LDP-Hr had high affinity to bind to esophageal squamous cell carcinoma (ESCC) cells, and enediyne-energized fusion protein Ec-LDP-Hr-AE showed potent cytotoxicity to ESCC cells with differential expression of EGFR and HER2. Ec-LDP-Hr-AE could cause significant G2-M arrest in EC9706 and KYSE150 cells, and it also induced apoptosis in ESCC cells in a dosage-dependent manner. Western blot assays showed that Ec-LDP-Hr-AE promoted caspase-3 and caspase-7 activities as well as PARP cleavage. Moreover, Ec-LDP-Hr-AE inhibited cell proliferation via decreasing phosphorylation of EGFR and HER2, and further exerted inhibition of the activation of their downstream signaling molecules. In vivo, at a tolerated dose, Ec-LDP-Hr-AE inhibited tumor growth by 88% when it was administered to nude mice bearing human ESCC cell KYSE150 xenografts. These results indicated that Ec-LDP-Hr-AE exhibited potent anti-caner efficacy on ESCC, suggesting it could be a promising candidate for targeted therapy of esophageal cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Enediynes/chemistry , ErbB Receptors/antagonists & inhibitors , Esophageal Neoplasms/drug therapy , Receptor, ErbB-2/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Animals , Antineoplastic Agents/chemistry , Caspase 2/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cysteine Endopeptidases/metabolism , ErbB Receptors/metabolism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , G2 Phase Cell Cycle Checkpoints , Humans , M Phase Cell Cycle Checkpoints/drug effects , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Receptor, ErbB-2/metabolism , Recombinant Fusion Proteins/chemistry , Xenograft Model Antitumor Assays
12.
Cancer Biol Ther ; 15(4): 398-408, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24424202

ABSTRACT

Present work mainly evaluated the inhibitory effects of lidamycin (LDM), an enediyne antibiotic, on angiogenesis or glioma-induced angiogenesis in vitro and in vivo, especially its synergistic anti-angiogenesis with temozolomide (TMZ). LDM alone efficiently inhibited proliferations and induced apoptosis of rat brain microvessel endothelial cells (rBMEC). LDM also interrupted the tube formation of rat brain microvessel endothelial cells (rBMEC) and rat aortic ring spreading. The blockade of rBMEC invasion and C6 cell-induced rBMEC migration by LDM was associated with decrease of VEGF secretion in a co-culture system. TMZ dramatically potentiated the effects of LDM on anti-proliferation, apoptosis induction, and synergistically inhibited angiogenesis events. As determined by western blot and ELISA, the interaction of tumor cells and the rBMEC was markedly interrupted by LDM plus TMZ with synergistic regulations of VEGF induced angiogenesis signal pathway, tumor cell invasion/migration, and apoptosis signal pathway. Immunofluorohistochemistry of CD31 and VEGF showed that LDM plus TMZ resulted in synergistic decrease of microvessel density (MVD) and VEGF expression in human glioma U87 cell subcutaneous xenograft. This study indicates that the high efficacy of LDM and the synergistic effects of LDM plus TMZ against glioma are mediated, at least in part, by the potentiated anti-angiogenesis.


Subject(s)
Aminoglycosides/pharmacology , Angiogenesis Inhibitors/pharmacology , Anti-Bacterial Agents/pharmacology , Brain Neoplasms/blood supply , Dacarbazine/analogs & derivatives , Enediynes/pharmacology , Glioma/blood supply , Neovascularization, Pathologic/metabolism , Aminoglycosides/therapeutic use , Angiogenesis Inhibitors/therapeutic use , Animals , Anti-Bacterial Agents/therapeutic use , Apoptosis/drug effects , Brain/blood supply , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Line, Tumor/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , Drug Synergism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enediynes/therapeutic use , Glioma/drug therapy , Glioma/pathology , Heterografts , Humans , Male , Mice, Nude , Rats , Temozolomide , Vascular Endothelial Growth Factor A/metabolism
13.
BMC Cancer ; 13: 479, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24128285

ABSTRACT

BACKGROUND: Endostatin (ES) inhibits endothelial cell proliferation, migration, invasion, and tube formation. It also shows antiangiogenesis and antitumor activities in several animal models. Endostatin specifically targets tumor vasculature to block tumor growth. Lidamycin (LDM), which consists of an active enediyne chromophore (AE) and a non-covalently bound apo-protein (LDP), is a member of chromoprotein family of antitumor antibiotics with extremely potent cytotoxicity to cancer cells. Therefore, we reasoned that endostatin-lidamycin (ES-LDM) fusion proteins upon energizing with enediyne chromophore may obtain the combined capability targeting tumor vasculature and tumor cell by respective ES and LDM moiety. METHODS: In this study, we designed and obtained two new endostatin-based fusion proteins, endostatin-LDP (ES-LDP) and LDP-endostatin (LDP-ES). In vitro, the antiangiogenic effect of fusion proteins was determined by the wound healing assay and tube formation assay and the cytotoxicity of their enediyne-energized analogs was evaluated by CCK-8 assay. Tissue microarray was used to analyze the binding affinity of LDP, ES or ES-LDP with specimens of human lung tissue and lung tumor. The in vivo efficacy of the fusion proteins was evaluated with human lung carcinoma PG-BE1 xenograft and the experimental metastasis model of 4T1-luc breast cancer. RESULTS: ES-LDP and LDP-ES disrupted the formation of endothelial tube structures and inhibited endothelial cell migration. Evidently, ES-LDP accumulated in the tumor and suppressed tumor growth and metastasis. ES-LDP and ES show higher binding capability than LDP to lung carcinoma; in addition, ES-LDP and ES share similar binding capability. Furthermore, the enediyne-energized fusion protein ES-LDP-AE demonstrated significant efficacy against lung carcinoma xenograft in athymic mice. CONCLUSIONS: The ES-based fusion protein therapy provides some fundamental information for further drug development. Targeting both tumor vasculature and tumor cells by endostatin-based fusion proteins and their enediyne-energized analogs probably provides a promising modality in cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Endostatins/pharmacology , Lung Neoplasms/drug therapy , Recombinant Fusion Proteins/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Endostatins/pharmacokinetics , Endostatins/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/physiology , Female , Humans , Lung/metabolism , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Tissue Array Analysis , Tissue Distribution , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
14.
Cancer Chemother Pharmacol ; 72(4): 777-88, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23975242

ABSTRACT

PURPOSE: Capecitabine (CAP), a prodrug, needs to be converted to 5-fluorouracil by several key enzymes, including thymidine phosphorylase (TP). To improve the therapeutic index, potentiation of antitumor activity of CAP is required. In this study, we explored whether lidamycin (LDM), an enediyne anticancer antibiotic, can induce synergistic antitumor effects in combination with CAP in murine breast cancer in vitro and in vivo. METHODS: Using MTT, cell migration and invasion, siRNA knockdown, and Western blot assays, the in vitro synergistic effects of LDM plus CAP on 4T1(LUC) cells were evaluated, and the mechanism of this synergy was explored. For in vivo model of orthotopic implantation model of 4T1(LUC) cells, optical molecular imaging system was utilized to evaluate the growth of primary tumor and metastasis. To further understand the mechanism of action of the LDM/CAP combination, immunohistochemistry analysis was carried out to detect thymidine phosphorylase induction and ERK signaling. RESULTS: As determined by MTT and transwell assay, LDM enhanced the inhibitory effects of CAP on cancer cell proliferation, migration, and invasion. Western blot showed that this synergistic effect was attributed to the up-regulated expression of TP induced by LDM. Knocking down TP impaired the synergistic anti-proliferative effect of LDM and CAP. Furthermore, our data suggested that LDM-induced up-regulation of TP both in vitro and in vivo is associated with ERK activation, because the inhibition of ERK activity by ERK inhibitor U0126 abrogated LDM-induced TP up-regulation. In animal models, LDM plus CAP potently inhibited primary tumor growth as well as lung metastasis compared with control or single-agent-treated group. CONCLUSIONS: LDM can potentiate the antitumor effects of CAP on breast cancer line. The synergistic effects suggest that the combination of LDM and CAP is an innovative antitumor strategy for breast cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Lung Neoplasms/prevention & control , Mammary Neoplasms, Experimental/drug therapy , Thymidine Phosphorylase/genetics , Up-Regulation/drug effects , Aminoglycosides/administration & dosage , Animals , Blotting, Western , Capecitabine , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Drug Synergism , Enediynes/administration & dosage , Enzyme Induction/drug effects , Female , Fluorouracil/administration & dosage , Fluorouracil/analogs & derivatives , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Lung Neoplasms/secondary , MAP Kinase Signaling System/drug effects , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Thymidine Phosphorylase/biosynthesis
15.
Oncol Lett ; 5(4): 1183-1188, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23599760

ABSTRACT

Gelatinases are overexpressed in several types of maligancies and tumor stromal cells. Lidamycin is an enediyne antitumor antibiotic, which is composed of an apoprotein (LDP) and an active chromophore (AE). It is known that the heavy-chain complementarity-determining region-3 (CDR3) domain of scFv is important in antibody affinity. The aim of this study was to prepare the enediyne-energized fusion proteins with a heavy-chain CDR3 domain of anti-gelatinases scFv and lidamycin, and to evaluate their antitumor efficiency. Fusion proteins comprising the CDR3 domain and the lidamycin apoprotein were generated, and ELISA, immunofluorescence and FACS were used to analyze the binding of the fusion protein with antigen gelatinases. The purified fusion proteins were assembled with the lidamycin chromophore, and the antitumor effects were evaluated in vitro and in vivo. It was found that the CDR3-LDP and CDR3-LDP-CDR3 fusion proteins demonstrated high affinity towards antigen gelatinases. Following stimulation of CDR3-LDP with enediyne, the results of MTT showed potent cytotoxicity towards tumor cells; the IC50 values of CDR3-LDP-AE to HepG2 and Bel-7402 tumor cells were 1.05×10-11 and 6.6×10-14 M, respectively. In addition, CDR3-LDP-AE displayed a potent antitumor effect in H22 cell xenografts in mice; the combination of CDR3-LDP (10 mg/kg) and CDR3-LDP-AE (0.25 and 0.5 mg/kg) revealed that the tumor inhibitory rates were 85.2 and 92.7%, respectively (P<0.05 compared with CDR3-LDP-AE). In conclusion, these results suggest that the CDR3-LDP fusion protein and its analog CDR3-LDP-AE may both be promising candidates for tumor targeting therapy.

16.
J Sci Food Agric ; 93(1): 149-56, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-22696075

ABSTRACT

BACKGROUND: Lycium barbarum polysaccharide (LBP), isolated with boiling water from the famous Chinese medicinal herb Lycium barbarum fruits, is one of the most important functional constituents in Lycium barbarum. In this study the effects of LBP on cell proliferation, cell cycle and apoptosis in human cervical carcinoma cells (HeLa cells) were investigated. RESULTS: LBP could inhibit the proliferation of HeLa cells by changing cell cycle distribution and inducing apoptosis. In addition, the loss of mitochondrial transmembrane potential (Δψm ) was observed by flow cytometry and the increase of intracellular Ca(2+) concentration was detected by laser scanning confocal microscope in apoptotic cells. At the same time, the nitric oxide content, nitric oxide synthase and inducible nitric oxide synthase activities were also increased. CONCLUSION: The inhibitory effect of LBP on the proliferation of HeLa cells was caused by inducing apoptosis through the mitochondrial pathway. The results showed that LBP can be developed as a potential chemotherapeutic agent candidate against human cervical cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Drugs, Chinese Herbal/therapeutic use , Lycium/chemistry , Phytotherapy , Uterine Cervical Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Calcium/metabolism , Cell Cycle/drug effects , Cell Proliferation , Drugs, Chinese Herbal/pharmacology , Female , Fruit/chemistry , HeLa Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Uterine Cervical Neoplasms/metabolism
17.
J Environ Biol ; 34(2 Spec No): 409-19, 2013 Apr.
Article in English | MEDLINE | ID: mdl-24620612

ABSTRACT

In this study, two adverse environments: low dissolved oxygen (DO) and high ammonia concentration, were employed to investigate the morphology, interspecies quorum sensing, extracellular polymers (EPS) characterization and microbial communities in the formation of aerobic granular sludge. Results showed that low DO could promote filamentous bacterial outgrowth. Under high ammonia concentration aerobic granular sludge (AGS) could still be cultivated, although it was looser and lighter than the control group. During the early stage of the AGS cultivation process, Al-2 activity reached a peak value in all three reactors, and ultrasonic pre-treatment was not beneficial to the release of Al-2. During AGS formation, the production of polysaccharide exhibited increases from 12.2% to 40.3%, 49.6%, and 29.3%. And PS in R2 was the highest as the result of sludge bulking. PS/PN was 1.5 to approximately 8 in the three reactors. Three-dimensional EEM fuorescence spectroscopy variation indicated the change of protein in EPS, and the highest intensity of Peak T1 was obtained. The location shift of Peak T1 was not obvious, and Peaks A, C, and T2 shifted toward longer wavelengths (red shift) of 5 to approximately 60 nm, or shorter wavelengths (blue shift) of 10 to approximately 25 nm on the emission scale and/or excitation scale in all three reactors. This provided spectral information on the chemical structure changes. Bacteria in R3 had the highest species diversity, and all bacteria in beta-Proteobacteria were identified as genus Thauera, which suggested that simultaneous nitrification and denitrification occurred in R3. The filamentous bacteria in seed sludge and R2 were species-richer. There was a low abundance of filamentous bacteria in R1 and R3, which contributed to the granule structure stability.


Subject(s)
Ammonia/chemistry , Oxygen/chemistry , Sewage , Aerobiosis , Bacteria, Aerobic/physiology , Oxygen/metabolism , Time Factors , Water Pollutants, Chemical/chemistry , Water Pollutants, Chemical/metabolism
18.
Yao Xue Xue Bao ; 48(12): 1771-7, 2013 Dec.
Article in Chinese | MEDLINE | ID: mdl-24689233

ABSTRACT

In the present study, a new compound named 17-(6-cinnamamido-hexylamino-)-17-demethoxygeldanamycin (CDG) was obtained by introducing the cinnamic acid (CA) group into the 17-site of geldanamycin (GDM). The anti-cancer effects of CDG in vitro and in vivo were evaluated. MTT assay was used to examine the inhibitory effect of CDG on the proliferation of MCF-7, HepG2, H460 and SW1990 cells. Immunofluorescent staining flow cytometry combined with Annexin V-FITC/PI staining were used to detect apoptotic cells. Transwell assay was used to analyze the effect of CDG on cell invasion and migration ability. Western blotting was used to detect the expression levels of RAF-1, EGFR, AKT, CDK4 and HER-2 of MCF-7, HepG2 and H460 cells. The toxicities of CDG and GDM were evaluated in mice. Using the subcutaneously transplanted MCF-7 xenograft in nude mice, inhibitory effect was evaluated in vivo. The results showed that CDG inhibited the proliferation of cancer cells (IC50: 13.6-67.4 microg.mL-1). After exposure to CDG for 48 h, most cells presented typical morphologic changes of apoptosis such as chromatin condensation or shrunken nucleus. The rates of apoptosis of MCF-7, HepG2, H460 and SW1990 cells incubated with 10 microg.mL-1 CDG were 23.16%, 27.55%, 22.21%, 20.47%, respectively. A dose-dependent reduction of migration of four cell lines was found after exposure to CDG. The decreased levels of RAF-1, EGFR, AKT, CDK4 and HER-2 showed that CDG possessed HSP90 inhibitory effect. The result of animal toxicity test on the mice suggested that CDG had lower toxicity than GDM. Meanwhile, CDG inhibited the growth of MCF-7 xenografts of athymic mice.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzoquinones/pharmacology , Lactams, Macrocyclic/pharmacology , Tumor Burden/drug effects , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzoquinones/chemical synthesis , Benzoquinones/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/metabolism , ErbB Receptors/metabolism , Female , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Lactams, Macrocyclic/chemical synthesis , Lactams, Macrocyclic/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Proto-Oncogene Proteins A-raf/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Random Allocation , Receptor, ErbB-2/metabolism , Xenograft Model Antitumor Assays
19.
Oncol Rep ; 28(4): 1193-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22797730

ABSTRACT

Gelatinases play an important role in tumor growth and metastasis, and overexpression of these molecules is strongly correlated with poor prognosis in a variety of malignant tumors. Lidamycin is an enediyne antitumor antibiotic with potent cytotoxicity. We previously reported that a tandem scFv format (dFv-LDP-AE) showed enhanced binding ability with gelatinases compared with the scFv-lidamycin conjugate (Fv-LDP-AE). In this study, the antitumor activities of dFv-LDP-AE on hepatocellular carcinoma (HCC) were evaluated in vitro and in vivo. By SDS-PAGE analysis, it was found that partial fusion protein dFv-LDP existed as dimer; the results of ELISA and immunofluorescence demonstrated that the fusion protein dFv-LDP could efficiently bind to hepatoma cells in vitro. The apparent arrest of cell cycle at G2/M phase and induction of apoptosis at nanomole levels indicated that the dFv-LDP-AE was very potent against HCC. In in vivo experiments, dFv-LDP-AE shown enhanced cytotoxic effects compared to those of LDM. Administration at mouse tolerable dosage level, the inhibition rate of tumor growth was 89.5% of dFv-LDP-AE vs. 73.6% of LDM on transplantable H22 in mice (P<0.05) and, 87.3% of dFv-LDP-AE vs. 63.4% of LDM on hepatoma Bel-7402 in athymic mice (P<0.01). Small animal optical imaging showed that the FITC-labeled dFv-LDP preferentially localized in the tumor site in less than 30 min, which demonstrated remarkable tumor-targeting properties. Taken together with the above findings, the enediyne-energized fusion protein dFv-LDP-AE showed potential application as a new agent for therapeutic appications in HCC.


Subject(s)
Aminoglycosides/pharmacology , Antibiotics, Antineoplastic/pharmacology , Enediynes/pharmacology , Gelatinases/immunology , Single-Chain Antibodies/pharmacology , Aminoglycosides/metabolism , Animals , Antibiotics, Antineoplastic/immunology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Enediynes/metabolism , Gelatinases/metabolism , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Male , Mice , Mice, Nude , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/immunology , Xenograft Model Antitumor Assays
20.
Bioresour Technol ; 102(11): 6421-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21482465

ABSTRACT

To investigate the inducements of increase of cell hydrophobicity from aerobic biofloc (ABF) and granular sludge (AGS), in this study, as the first time the hydrophilic and hydrophobic bacterial communities were analyzed independently. Meanwhile, the effect of extracellular polymers (EPS) on the cell hydrophobicity is also studied. Few Bacteroidetes were detected (1.35% in ABF and 3.84% in AGS) in hydrophilic bacteria, whereas they are abundant in the hydrophobic cells (47.8% and 43% for ABF and AGS, respectively). The main species of Bacteroidetes changed from class Sphingobacteria to Flavobacteria in AGS. On the other hand, EPS is directly responsible to cell hydrophobicity. For AGS, cell hydrophobicity was sharply decreased after EPS extraction. Both quantity and property of the extracellular protein are related to hydrophobicity. Our results showed the variation of cell hydrophobicity was resulted from variations of both bacterial population and EPS.


Subject(s)
Bacteria/cytology , Bacteria/metabolism , Biopolymers/metabolism , Extracellular Space/metabolism , Hydrophobic and Hydrophilic Interactions , Sewage/microbiology , Aerobiosis , Bacteria/genetics , DNA, Ribosomal/genetics , Flocculation , Fluorescence , Gene Library , Molecular Sequence Data , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...