Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Exp Bot ; 73(10): 3205-3220, 2022 05 23.
Article in English | MEDLINE | ID: mdl-34758079

ABSTRACT

Whether photosynthesis has improved with increasing yield in major crops remains controversial. Research in this area has often neglected to account for differences in light intensity experienced by cultivars released in different years. Light intensity is expected to be positively associated with photosynthetic capacity and the resistance of the photosynthetic apparatus to high light but negatively associated with light-utilization efficiency under low light. Here, we analyzed the light environment, photosynthetic activity, and protein components of leaves of 26 winter wheat cultivars released during the past 60 years in China. Over time, light levels on flag leaves significantly decreased due to architectural changes, but photosynthetic rates under high or low light and the resistance of the photosynthetic apparatus to high light remained steady, contrary to expectations. We propose that the difference between the actual and expected trends is due to breeding. Specifically, breeding has optimized photosynthetic performance under high light rather than low light. Moreover, breeding selectivity altered the stoichiometry of several proteins related to dynamic photosynthesis, canopy light distribution, and photoprotection. These results indicate that breeding has significantly altered the photosynthetic mechanism in wheat and its response to the light environment. These changes likely have helped increase wheat yields.


Subject(s)
Plant Breeding , Triticum , Light , Photosynthesis/physiology , Plant Leaves/physiology , Triticum/metabolism
2.
BMC Plant Biol ; 20(1): 339, 2020 Jul 17.
Article in English | MEDLINE | ID: mdl-32680459

ABSTRACT

BACKGROUND: Plants are always exposed to dynamic light. The photosynthetic light use efficiency of leaves is lower in dynamic light than in uniform irradiance. Research on the influence of environmental factors on dynamic photosynthesis is very limited. Nitrogen is critical for plants, especially for photosynthesis. Low nitrogen (LN) decreases ribulose-1,5-bisphosphate carboxylase/oxygenase (Rubisco) and thus limits photosynthesis. The decrease in Rubisco also delays photosynthetic induction in LN leaves; therefore, we hypothesized that the difference of photosynthetic CO2 fixation between uniform and dynamic light will be greater in LN leaves compared to leaves with sufficient nitrogen supply. RESULTS: To test this hypothesis, soybean plants were grown under low or high nitrogen (HN), and the photosynthetic gas exchange, enzyme activity and protein amount in leaves were measured under uniform and dynamic light. Unexpectedly, dynamic light caused less photosynthetic suppression, rather than more, in LN leaves than in HN leaves. The underlying mechanism was also clarified. Short low-light (LL) intervals did not affect Rubisco activity but clearly deactivated fructose-1,6-bisphosphatase (FBPase) and sedoheptulose-1,7-bisphosphatase (SBPase), indicating that photosynthetic induction after a LL interval depends on the reactivation of FBPase and SBPase rather than Rubisco. In LN leaves, the amount of Rubisco decreased more than FBPase and SBPase, so FBPase and SBPase were present in relative excess. A lower fraction of FBPase and SBPase needs to be activated in LN leaves for photosynthesis recovery during the high-light phase of dynamic light. Therefore, photosynthetic recovery is faster in LN leaves than in HN leaves, which relieves the photosynthetic suppression caused by dynamic light in LN leaves. CONCLUSIONS: Contrary to our expectations, dynamic light caused less photosynthetic suppression, rather than more, in LN leaves than in HN leaves of soybean. This is the first report of a stress condition alleviating the photosynthetic suppression caused by dynamic light.


Subject(s)
Glycine max/physiology , Nitrogen/deficiency , Photosynthesis/radiation effects , Light , Nitrogen/physiology , Photosynthesis/drug effects , Plant Leaves/drug effects , Plant Leaves/physiology , Plant Leaves/radiation effects , Ribulose-Bisphosphate Carboxylase/drug effects , Ribulose-Bisphosphate Carboxylase/radiation effects , Glycine max/drug effects , Glycine max/radiation effects , Stress, Physiological
3.
Theranostics ; 8(7): 2044-2060, 2018.
Article in English | MEDLINE | ID: mdl-29556372

ABSTRACT

Rationale: Cardenolides have potential as anticancer drugs. 3'-epi-12ß-hydroxyfroside (HyFS) is a new cardenolide structure isolated by our research group, but its molecular mechanisms remain poorly understood. This study investigates the relationship between its antitumor activities and autophagy in lung cancer cells. Methods: Cell growth and proliferation were detected by MTT, lactate dehydrogenase (LDH) release, 5-ethynyl-20-deoxyuridine (EDU) and colony formation assays. Cell apoptosis was detected by flow cytometry. Autophagic and signal proteins were detected by Western blotting. Markers of autophagy and autophagy flux were also detected by immunofluorescence, transmission electron microscopy and acridine orange staining. Real time RT-PCR was used to analyze the gene expression of Hsp90. Hsp90 ubiquitination was detected by coimmunoprecipitation. The antitumore activities of HyFS were observed in nude mice. Results: HyFS treatment inhibited cell proliferation and induced autophagy in A549 and H460 lung cancer cells, but stronger inhibition of cell proliferation and induction of cell apoptosis were shown when HyFS-mediated autophagy was blocked. The Hsp90/Akt/mTOR axis was found to be involved in the activation of HyFS-mediated autophagy. Evidence of direct interaction between Hsp90 and Akt was observed. HyFS treatment resulted in decreased levels of heat shock protein 90 (Hsp90) and phosphorylated Akt, overexpression of Hsp90 increased activation of autophagy, and inhibition of Hsp90 expression decreased autophagy. In addition, ubiquitin-mediated degradation of Hsp90 and subsequent dephosphorylation of its client protein Akt were also found in HyFS-treated lung cancer cells. Moreover, combination treatment with HyFS and chloroquine showed remarkably increased tumor inhibition in both A549- and H460-bearing mice. Conclusion: Our results demonstrate that HyFS induced cytoprotective autophagy through ubiquitin-mediated degradation of Hsp90, which further blocked the Akt/mTOR pathway in lung cancer cells. Thus, a combination of a HyFS-like cardenolide and an autophagic inhibitor is a potential alternative approach for the treatment of lung cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cardenolides/pharmacology , Lung Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Blotting, Western , Cardenolides/administration & dosage , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Formazans/analysis , Gene Expression Profiling , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice, Inbred BALB C , Mice, Nude , Microscopy, Electron, Transmission , Neoplasm Transplantation , Oncogene Protein v-akt/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Staining and Labeling , TOR Serine-Threonine Kinases/metabolism , Tetrazolium Salts/analysis , Treatment Outcome
4.
Sci Rep ; 8(1): 4253, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523804

ABSTRACT

Bisphenol A (BPA), a widely distributed pollutant, suppresses photosynthesis in leaves. In previous studies on higher plants, the plants were treated by BPA through irrigation to root. This method cannot distinguish whether the BPA directly suppresses photosynthesis in leaves, or indirectly influences photosynthesis through affecting the function of root. Here, only the leaves but not the roots of cucumber were infiltrated with BPA solution. The photosystem II and I (PSII, PSI) were insensitive to BPA under darkness. BPA aggravated the PSII but not the PSI photoinhibition under light. BPA also inhibited CO2 assimilation, and the effect of BPA on PSII photoinhibition disappeared when the CO2 assimilation was blocked. The H2O2 accumulated in BPA-treated leaves under light. And the BPA-caused PSII photoinhibition was prevented under low (2%) O2. We also proved that the BPA-caused PSII photoinhibition depend on the turnover of D1 protein. In conclusion, this study proved that BPA could directly suppress photosynthesis in leaves, however, BPA does not damage PSII directly, but inhibits CO2 assimilation and over-reduces the electron transport chain under light, which increases the production of reactive oxygen species (H2O2), the over-accumulated ROS inhibits the turnover of D1 protein and consequently aggravates PSII photoinhibition.


Subject(s)
Air Pollutants/pharmacology , Benzhydryl Compounds/pharmacology , Cucumis sativus/drug effects , Phenols/pharmacology , Photosynthesis/drug effects , Photosystem II Protein Complex/metabolism , Carbon Dioxide/metabolism , Cucumis sativus/metabolism , Hydrogen Peroxide/metabolism , Plant Leaves/drug effects , Plant Leaves/metabolism
5.
Cell Prolif ; 51(4): e12451, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29484762

ABSTRACT

OBJECTIVES: Coroglaucigenin (CGN), a natural product isolated from Calotropis gigantean by our research group, has been identified as a potential anti-cancer agent. However, the molecular mechanisms involved remain poorly understood. MATERIALS AND METHODS: Cell viability and cell proliferation were detected by MTT and BrdU assays. Flow cytometry, SA-ß-gal assay, western blotting and immunofluorescence were performed to determine CGN-induced apoptosis, senescence and autophagy. Western blotting, siRNA transfection and coimmunoprecipitation were carried out to investigate the mechanisms of CGN-induced senescence and autophagy. The anti-tumour activities of combination therapy with CGN and chloroquine were observed in mice tumour models. RESULTS: We demonstrated that CGN inhibits the proliferation of colorectal cancer cells both in vitro and in vivo. We showed that the inhibition of cell proliferation by CGN is independent of apoptosis, but is associated with cell-cycle arrest and senescence in colorectal cancer cells. Notably, CGN induces protective autophagy that attenuates CGN-mediated cell proliferation. Functional studies revealed that CGN disrupts the association of Hsp90 with both CDK4 and Akt, leading to CDK4 degradation and Akt dephosphorylation, eventually resulting in senescence and autophagy, respectively. Combination therapy with CGN and chloroquine resulted in enhanced anti-tumour effects in vivo. CONCLUSIONS: Our results demonstrate that CGN induces senescence and autophagy in colorectal cancer cells and indicate that combining it with an autophagy inhibitor may be a novel strategy suitable for CGN-mediated anti-cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Biological Products/pharmacology , Cardenolides/pharmacology , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Animals , Calotropis/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Chloroquine/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Drug Therapy, Combination , HSP90 Heat-Shock Proteins/metabolism , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism
6.
Oncotarget ; 8(32): 52783-52791, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28881770

ABSTRACT

Colorectal cancer is the most common cancer. It has high morbidity and mortality worldwide, and more effective treatment strategies need to be developed. Toxicarioside O (TCO), a natural product derived from Antiaris toxicaria, has been shown to be a potential anticancer agent. However, the molecular mechanisms involved remain poorly understood. In this study, our results demonstrated that TCO can induce both apoptosis and autophagy in colorectal cancer cells. Moreover, TCO-induced autophagy was due to the increase of the expression and activity of the enzyme sirtuin-1 (SIRT1), and subsequent inhibition of the Akt/mTOR pathway. Inhibition of SIRT1 activity by its inhibitor, EX-527, attenuated TCO-induced autophagy. Of interest, inhibition of autophagy by chloroguine, an autophagy inhibitor, enhanced TCO-induced apoptotic cell death, suggesting that autophagy plays a protective role in TCO-induced apoptosis. Together, these findings suggest that combination of TCO and autophagy inhibitor may be a novel strategy suitable for potentiating the anticancer activity of TCO for treatment of colorectal cancer.

7.
Int J Cancer ; 134(8): 1981-90, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24150860

ABSTRACT

Various angiogenesis-related self-molecules have been considered to be therapeutic targets. However, the direct use of self-molecules as vaccines is not recommended because of the inherent ability of the host to develop immune tolerance. Antigen 43 (Ag43) is a surface protein found in E. coli and contains an α and a ß subunits, which contains multiple T epitopes in α subunit. Here we construct a novel Ag43 surface display system (Ag43 system) to express Ag43 chimeric proteins to disrupt immune tolerance against self-molecules. The Ag43 system was constructed from an Escherichia coli strain Tan109, derived from JM109, in which the Ag43 gene was deleted and a recombinant plasmid (pETAg43') expressing a partial Ag43 gene was introduced. The extracellular domain of angiogenesis-related endoglin gene was then subcloned into plasmid pETAg43', resulting in a recombinant plasmid pETAg43'/END(e) which was then used to transform Tan109 for protein expression. We found that Ag43 and endoglin chimeric protein (Ag43'/END(e) ) was expressed on the bacterial surface. The chimeric protein could be separated from the bacterial surface by heating to 60°C and yet retain activity. We used Ag43'/END(e) as a protein vaccine and found that it could disrupt immune tolerance against endoglin by inducing significant antitumor activities and inhibit angiogenesis in several tumor models without significant side effects. These data suggest that Ag43'/END(e) chimeric protein is a potential model vaccine for active tumor immunotherapy, and that Ag43 system could be an effective tool for novel vaccine preparation to break immune tolerance to other angiogenesis-related self-molecules for cancer therapy.


Subject(s)
Adhesins, Escherichia coli/immunology , Cancer Vaccines/immunology , Carcinoma, Lewis Lung/therapy , Immune Tolerance/immunology , Intracellular Signaling Peptides and Proteins/immunology , Neovascularization, Pathologic/therapy , Adhesins, Escherichia coli/genetics , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Antigens, Surface/genetics , Antigens, Surface/immunology , Carcinoma, Lewis Lung/immunology , Endoglin , Epitopes, T-Lymphocyte , Escherichia coli/genetics , Escherichia coli/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
8.
Oncol Rep ; 30(1): 478-84, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23615686

ABSTRACT

Cytochalasin D (CytD) targets actin, a ubiquitous protein in eukaryotic cells. Previous studies have focused mainly on the antitumor effects of CytD. We previously found CytD to promote lung metastasis in B16 melanoma cells, which we had not anticipated, and, therefore, in the present study we investigated the possible underlying mechanisms. B16 melanoma cells were co-cultured with CytD and other agents and used to establish a lung metastatic model. In this B16 melanoma metastatic model, significantly increased lung metastasis and lung weight were found in CytD-treated mice, which was almost completely suppressed by tissue factor (TF) RNA interference expressed via lentivirus. The results of northern and western blot, and real-time RT-PCR analysis showed that the expression of TF was significantly upregulated in B16 cells treated with CytD but was significantly inhibited by TF RNA interference. In addition, upregulation and phosphorylation of mitogen-activated protein kinase p38 were also found in the metastatic lung tissues treated with CytD and in the B16 cells co-cultured with CytD and factor VIIa (FVIIa), but not in cells cultured with CytD, dimethyl sulfoxide or FVIIa alone. These results indicate that CytD stimulates the expression of TF in B16 melanoma cells, activating both coagulation-dependent and -independent pathways via binding to FVIIa, eventually promoting lung metastasis. TF interference is a potential approach to the prevention of B16 melanoma metastasis.


Subject(s)
Cytochalasin D/pharmacology , Lung Neoplasms/secondary , Melanoma, Experimental/metabolism , Thromboplastin/metabolism , Animals , Cell Line, Tumor , Factor VIIa/metabolism , Female , Gene Expression Regulation, Neoplastic , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Nucleic Acid Synthesis Inhibitors/pharmacology , Phosphorylation , RNA Interference , RNA, Small Interfering , Thromboplastin/biosynthesis , Thromboplastin/genetics , Up-Regulation , p38 Mitogen-Activated Protein Kinases/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
9.
PLoS One ; 7(11): e50351, 2012.
Article in English | MEDLINE | ID: mdl-23209720

ABSTRACT

Toxicarioside A is a cardenolide isolated mainly from plants and animals. Emerging evidence demonstrate that cardenolides not only have cardiac effects but also anticancer effects. In this study, we used in vivo models to investigate the antitumor activities of toxicarioside A and the potential mechanisms behind them. Murine colorectal carcinoma (CT26) and Lewis lung carcinoma (LL/2) models were established in syngeneic BALB/c and C57BL/6 mice, respectively. We found that the optimum effective dose of toxicarioside A treatment significantly suppressed tumor growth and angiogenesis in CT and LL/2 tumor models in vivo. Northern and Western blot analysis showed significant inhibition of endoglin expression in toxicarioside A-treated human umbilical vein endothelial cells (HUVECs) in vitro and tumor tissues in vivo. Toxicarioside A treatment significantly inhibited cell proliferation, migration and invasion, but did not cause significant cell apoptosis and affected other membrane protein (such as CD31 and MHC I) expression. In addition, TGF-ß expression was also significantly inhibited in CT26 and LL/2 tumor cells treated with toxicarioside A. Western blot analysis indicated that Smad1 and phosphorylated Smad1 but not Smad2/3 and phosphorylated Smad2/3 were attenuated in HUVECs treated with toxicarioside A. Smad1 and Smad2/3 signaling remained unchanged in CT26 and LL/2 tumor cells treated with toxicarioside A. Endoglin knockout by small interfering RNA against endoglin induced alternations in Smad1 and Smad2/3 signaling in HUVECs. Our results indicate that toxicarioside A suppresses tumor growth through inhibition of endoglin-related tumor angiogenesis, which involves in the endoglin/TGF-ß signal pathway.


Subject(s)
Cardiac Glycosides/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Neovascularization, Pathologic , Transforming Growth Factor beta/metabolism , Alginates/chemistry , Animals , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Endoglin , Endothelial Cells/cytology , Female , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microcirculation , Models, Chemical , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplasms/pathology , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction
10.
World J Gastroenterol ; 18(14): 1602-9, 2012 Apr 14.
Article in English | MEDLINE | ID: mdl-22529688

ABSTRACT

AIM: To investigate the inhibitory role of toxicarioside A on the gastric cancer cell line human gastric cancer cell line (SGC-7901) and determine the underlying molecular mechanism. METHODS: After SGC-7901 cells were treated with toxicarioside A at various concentrations (0.5, 1.5, 4.5, 9.0 µg/mL) for 24 h or 48 h, cell viability was determined by 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide assay, and the motility and invasion of tumor cells were assessed by the Transwell chamber assay. Immunofluorescence staining, reverse transcription polymerase chain reaction and Western blotting were performed to detect the expression of basic fibroblast growth factor (bFGF) and fibroblast growth factor receptor-1 (FGFR1), and nuclear factor-kappa B (NF-κB) activation was examined by electrophoretic mobility shift assay. RESULTS: The results showed that toxicarioside A was capable of reducing cell viability, inhibiting cell growth, and suppressing cell migration and invasion activities in a time- and dose-dependent manner in SGC-7901 cells. Further analysis revealed that not only the expression of bFGF and its high-affinity receptor FGFR1 but also the NF-κB-DNA binding activity were effectively blocked by toxicarioside A in a dose-dependent manner compared with the control group (P < 0.05 or P < 0.01). Interestingly, application of the NF-κB specific inhibitor, pyrrolidinedithiocarbamate (PDTC), to SGC-7901 cells significantly potentized the toxicarioside A-induced down-regulation of bFGF compared with the control group (P < 0.05). CONCLUSION: These findings suggest that toxicarioside A has an anti-gastric cancer activity and this effect may be achieved partly through down-regulation of NF-κB and bFGF/FGFR1 signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Cardiac Glycosides/pharmacology , Fibroblast Growth Factor 2/metabolism , NF-kappa B/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/drug therapy , Cardenolides/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Fibroblast Growth Factor 2/drug effects , Humans , NF-kappa B/drug effects , Neoplasm Invasiveness , Proline/analogs & derivatives , Proline/pharmacology , Stomach Neoplasms/pathology , Thiocarbamates/pharmacology
11.
Asian Pac J Trop Med ; 5(3): 169-74, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22305779

ABSTRACT

OBJECTIVE: To investigate whether cytochalasin D can induce antitumor activities in a tumor model. METHODS: Murine CT26 colorectal carcinoma cells were cultured in vitro and cytochalasin D was used as a cytotoxic agent to detect its capabilities of inhibiting CT26 cell proliferation and inducing cell apoptosis by MTT and a TUNEL-based apoptosis assay. Murine CT26 tumor model was established to observe the tumor growth and survival time. Tumor tissues were used to detect the microvessel density by immunohistochemistry. In addition, alginate encapsulated tumor cell assay was used to quantify the tumor angiogenesis in vivo. RESULTS: Cytochalasin D inhibited CT26 tumor cell proliferation in time and dose dependent manner and induced significant CT26 cell apoptosis, which almost reached the level induced by the positive control nuclease. The optimum effective dose of cytochalasin D for in vivo therapy was about 50 mg/kg. Cytochalasin D in vivo treatment significantly inhibited tumor growth and prolonged the survival times in CT26 tumor-bearing mice. The results of immunohistochemistry analysis and alginate encapsulation assay indicated that the cytochalasin D could effectively inhibited tumor angiogenesis. CONCLUSIONS: Cytochalasin D inhibits CT26 tumor growth potentially through inhibition of cell proliferation, induction of cell apoptosis and suppression of tumor angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/drug therapy , Cytochalasin D/pharmacology , Angiogenesis Inhibitors/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/blood supply , Cytochalasin D/administration & dosage , Dose-Response Relationship, Drug , In Situ Nick-End Labeling , Mice , Microvessels/drug effects , Neovascularization, Pathologic/prevention & control , Tumor Cells, Cultured
12.
Eur J Cancer ; 48(14): 2260-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22257793

ABSTRACT

Cytochalasin D targets actin and is ubiquitous in eukaryotic cells. When cytochalasin D is used as a cytotoxic agent in cancer therapy, it causes significant side effects. To prevent this, cytochalasin D can be encapsulated in polyethylene liposomes. In this study, high-performance liquid chromatography observation of the biodistribution of pegylated liposomal cytochalasin D in tumour-bearing mice showed that liposomal cytochalasin D could be conveniently dissolved in water for i.v. injection and that it specifically accumulated in tumour tissues, more than natural cytochalasin D did. The half-time of liposomal cytochalasin D in the plasma was also significantly longer than that of natural cytochalasin D (4h versus 10 min). MTT 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay showed that liposomal cytochalasin D treatment could cause significant inhibition of cell proliferation in vitro in a manner similar to that of natural cytochalasin D. The antitumour activities of liposomal cytochalasin D were investigated in B16 melanoma, CT26 colorectal carcinoma and H22 hepatoma models, and the results indicated that liposomal cytochalasin D could significantly inhibit tumour growth and prolong survival in a manner similar to that of cisplatin. TUNEL-based apoptosis assays showed that liposomal cytochalasin D induced significant tumour cell apoptosis. Significant inhibition of tumour angiogenesis was observed in mice treated with liposomal cytochalasin D. In addition, no significant side effects were observed in mice treated with liposomal cytochalasin D. Our results show that liposomal cytochalasin D increases solubility and bioavailability, a lower incidence of side effects and improves antitumour effects, indicating its potential as a chemical agent for cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/drug therapy , Colorectal Neoplasms/drug therapy , Cytochalasin D/pharmacology , Liver Neoplasms, Experimental/drug therapy , Melanoma, Experimental/drug therapy , Polyethylene Glycols/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Biological Availability , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry, Pharmaceutical , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytochalasin D/administration & dosage , Cytochalasin D/analogs & derivatives , Cytochalasin D/pharmacokinetics , Cytochalasin D/toxicity , Dose-Response Relationship, Drug , Half-Life , Injections, Intravenous , Liposomes , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/pharmacokinetics , Polyethylene Glycols/toxicity , Solubility , Tissue Distribution , Tumor Burden/drug effects
13.
Asian Pac J Trop Med ; 5(2): 91-7, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22221748

ABSTRACT

OBJECTIVE: To investigate possible mechanism of toxicarioside A in HS-5 bone stromal cells. METHODS: HS-5 bone stromal cells were cultured in media supplemented with various concentrations of toxicarioside A or control DMSO (not treatment). Endoglin and TGF-ß were detected by Northern and Western blot analysis and quantified in a standard method. Downstream molecules of endoglin and TGF-ß (Smad1, Smad2 and their active phosphorylated counterparts, pSmad1 and pSmad2) were also detected and quantified by Western blot analysis. In addition, cell proliferation assay and small interfering RNA (siRNA) against endoglin were used to certificate the function of endolgin in the HS-5 cells. RESULTS: Compared with the not treated (0 µg/mL) or DMSO treated control HS-5 cells, HS-5 cells treated with toxicarioside A were found significant attenuation of endolgin and TGF-ß expression. Significant inhibition of cell proliferation was also found in the HS-5 cells treated with toxicarioside A. ALK1-related Smad1 and ALK5-related Smad2 were decreased in HS-5 cells treated with toxicarioside A. In addition, phosphorylated Smad1 (pSmad1) and Smad2 (pSmad2) were also found attenuation in toxicarioside A-treated HS-5 cells. RNA interference showed that blockage of endoglin by siRNA also decreased Smad1 and Smad2 expression in HS-5 cells. CONCLUSIONS: Our results indicate that toxicarioside A can influence bone marrow stromal HS-5's function and inhibit HS-5 cell proliferation by alteration of endoglin-related ALK1 (Smad1) and ALK5 (Smad2) signaling.


Subject(s)
Antiaris , Bone Marrow Cells/drug effects , Cardenolides/pharmacology , Receptors, Cell Surface/antagonists & inhibitors , Smad1 Protein/metabolism , Smad2 Protein/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Antigens, CD/metabolism , Blotting, Northern , Blotting, Western , Bone Marrow Cells/metabolism , Cell Line , Cell Proliferation , Endoglin , Humans , Male , Receptors, Cell Surface/metabolism , Signal Transduction/drug effects , Smad1 Protein/drug effects , Smad2 Protein/drug effects , Stromal Cells/drug effects , Transforming Growth Factor beta/metabolism
14.
J Zhejiang Univ Sci B ; 9(8): 602-9, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18763309

ABSTRACT

Induction of tumor vasculature occlusion by targeting a thrombogen to newly formed blood vessels in tumor tissues represents an intriguing approach to the eradication of primary solid tumors. In the current study, we construct and express a fusion protein containing vascular endothelial growth factor (VEGF) and tissue factor (TF) to explore whether this fusion protein has the capability of inhibiting tumor growth in a colon carcinoma model. The murine cDNA of VEGF A and TF were amplified by reverse transcriptase polymerase chain reaction (RT-PCR), and then cloned into prokaryotic expression plasmid pQE30 with a linker. The expression product recombinant VEGF-TF (rVEGF-TF) was purified and proved to have comparable enzyme activity to a commercial TF and the capability of specific binding to tumor vessels. Significant decrease of tumor growth was found in the mice administered with rVEGF-TF on Day 6 after initiated rVEGF-TF treatment (P<0.05), and the tumor masses in 2 of 10 mice were almost disappeared on Day 14 after the first treatment. In addition, valid thrombogenesis and tumor necrosis were observed in the tumor tissues injected with rVEGF-TF. Our results demonstrate that occlusion of tumor vasculature with rVEGF-TF is potentially an effective approach for cancer therapy.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Recombinant Fusion Proteins/therapeutic use , Thromboplastin/therapeutic use , Thrombosis/drug therapy , Vascular Endothelial Growth Factor A/therapeutic use , Animals , Cell Line, Tumor , Cloning, Molecular , Colonic Neoplasms/blood supply , Disease Models, Animal , Disease Progression , Gene Expression , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Plasmids/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Thromboplastin/genetics , Thromboplastin/isolation & purification , Thromboplastin/metabolism , Thrombosis/pathology , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/isolation & purification , Vascular Endothelial Growth Factor A/metabolism
15.
Immunol Invest ; 36(4): 423-40, 2007.
Article in English | MEDLINE | ID: mdl-17691024

ABSTRACT

The immunization approaches with DNA vaccine priming and subsequent protein or peptide boosting has been widely tested in various models of infectious diseases. However, these approaches are seldom reported in the areas of cancer immunotherapy. In this study we combined endoglin plasmid DNA and recombinant protein as vaccines and used them to prime and boost, simultaneously, as a vaccine strategy. Our results showed that combination of endoglin DNA and protein vaccines could enhance both protective and therapeutic anti-tumor efficacy in both colon carcinoma and Lewis lung carcinoma models. Significant inhibition of tumor angiogenesis was found in the tumor tissues. The titers of autoantibodies against murine endoglin were significantly increased and the antibody levels lasted longer in the mice with combined endoglin DNA and recombinant protein vaccination. CTL response against endoglin-positive HUVECs, but not against endoglin-negative tumor cells was found in the mice combined DNA with protein vaccination. In addition, combination of endoglin DNA and recombinant protein vaccination significantly induced IFN-gamma secreting cells. These observations suggested that a combination of endoglin DNA and recombinant protein immunization as a vaccine strategy was superior to those using endoglin DNA or recombinant protein alone as vaccines.


Subject(s)
Cancer Vaccines/immunology , Carcinoma, Lewis Lung/immunology , Colorectal Neoplasms/immunology , Intracellular Signaling Peptides and Proteins/immunology , Vaccines, DNA/immunology , Animals , Cancer Vaccines/therapeutic use , Carcinoma, Lewis Lung/blood supply , Carcinoma, Lewis Lung/therapy , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/therapy , Endoglin , Immunization Schedule , Interferon-gamma/immunology , Interferon-gamma/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic , Recombinant Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Vaccines, DNA/therapeutic use
16.
World J Gastroenterol ; 13(17): 2479-83, 2007 May 07.
Article in English | MEDLINE | ID: mdl-17552032

ABSTRACT

AIM: To explore the capability of a monoclonal antibody (mAb) against murine endoglin to inhibit tumor angiogenesis and suppression of hepatoma growth in murine models. METHODS: A monoclonal antibody against murine endoglin was purified by affinity chromatography and passively transfused through tail veins in two murine hepatoma models. Tumor volume and survival time were observed at three-day intervals for 48 d. Microvessels in tumor tissues were detected by immunohistochemistry against CD31, and angiogenesis in vivo was determined by alginate encapsulated assay. In addition, tumor cell apoptosis was detected by TUNEL assay. RESULTS: Passive immunotherapy with anti-endoglin mAb could effectively suppress tumor growth, and prolonged the survival time of hepatoma-bearing mice. Angiogenesis was apparently inhibited within the tumor tissues, and the vascularization of alginate beads was also reduced in the mice passively transfused with anti-endoglin mAb. In addition, increased apoptotic cells were observed within the tumor tissues from the mice passively transfused with anti-endoglin mAb. CONCLUSION: Passive immunotherapy with anti-endoglin mAb effectively inhibits tumor growth via inhibiting tumor angiogenesis and increasing tumor cell apoptosis, which may be highly correlated with the blockage of endoglin-related signal pathway induced by anti-endoglin mAb.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunization, Passive/methods , Intracellular Signaling Peptides and Proteins/immunology , Liver Neoplasms, Experimental/drug therapy , Alginates , Animals , Antibodies, Monoclonal/immunology , Apoptosis/drug effects , Cell Line, Tumor , Disease Models, Animal , Endoglin , Glucuronic Acid , Hexuronic Acids , Liver Neoplasms, Experimental/blood supply , Liver Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Microspheres , Neovascularization, Pathologic/drug therapy , Random Allocation , Signal Transduction/drug effects , Survival Rate
17.
Ai Zheng ; 24(10): 1179-83, 2005 Oct.
Article in Chinese | MEDLINE | ID: mdl-16219129

ABSTRACT

BACKGROUND & OBJECTIVE: Endoglin is a marker of tumor angiogenesis. Increasing evidences proved that passive immunotherapy with anti-endoglin monoclonal antibody can effectively inhibit tumor growth, and xenogeneic homologous DNA vaccine can inhibit cross antitumor immunity. This study was to explore the inhibitory effect of a DNA vaccine encoding the extracellular domain of porcine endoglin (ppEDG) on tumor growth in a mouse colon carcinoma model. METHODS: ppEDG was used as a DNA vaccine to actively immunize the colon carcinoma-bearing mice. Tumor volume and survival rate of the mice were observed in 3-day intervals. Microvessel density (MVD) was detected by immunohistochemistry; antibodies against self-endoglin were detected by Western blot and ELISA; the B cells that secrete auto-antibodies against self-endoglin were detected by ELISPOT assay. RESULTS: Eighteen days after tumor cell inoculation, the tumor volume was significantly smaller in ppEDG-immunized group than in empty plasmid (e-p) group and normal saline (NS) group (P<0.05), and the survival time was significantly longer in ppEDG-immunized group than in the control groups (P<0.001). MVD was significantly lower in ppEDG-immunized group than in e-p group and NS group (19.2+/-4.5 vs. 76.9+/-14.4 and 81.4+/-16.9, P<0.001). The antibodies against self-endoglin were identified in ppEDG-immunized group; the major subtypes were IgG(1) and IgG(2b). The auto-antibody-producing B cells were much more in ppEDG-immunized group than in e-p group and NS group (82.5+/-14.1 vs. 3.6+/-1.3 and 4.7+/-2.0, P<0.001). CONCLUSION: ppEDG DNA vaccine could induce the production of auto-antibodies against self-endoglin, which further inhibit angiogenesis and growth of colon carcinoma.


Subject(s)
Antigens, CD/genetics , Autoantibodies/blood , Colonic Neoplasms/pathology , Neovascularization, Pathologic , Receptors, Cell Surface/genetics , Vaccines, DNA/immunology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , COS Cells/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Chlorocebus aethiops , Colonic Neoplasms/blood supply , Colonic Neoplasms/immunology , Endoglin , Female , Humans , Immunization , Immunoglobulin G/blood , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Plasmids , Vaccines, DNA/genetics
18.
Hunan Yi Ke Da Xue Xue Bao ; 28(3): 233-6, 2003 Jun.
Article in Chinese | MEDLINE | ID: mdl-14653075

ABSTRACT

OBJECTIVE: To investigate the effect of the expression of phosphatase and tensin homolog deleted on chromosome ten (PTEN) and p27 proteins in non-Hodgkin's lymphoma (NHL). METHODS: The expressions of PTEN and p27 proteins were detected with immunohistochemical technique in 77 cases of NHL. RESULTS: In NHL, the positive rates of PTEN and p27 protein were 90.9% (70/77) and 51.9% (40/77), respectively. The positive strength of PTEN protein was positively correlated to that of p27 protein (r = 0.6841, P < 0.01). The expression intensity of PTEN and p27 proteins in the low grade group was all higher than that in the high grade group. The expression intensity of PTEN and p27 proteins in B NHL showed no significant difference with that of T NHL. CONCLUSION: The decrease of p27 protein expression caused by the deletion and descent of PTEN protein expression cannot effectively play the anti-oncogene role in negative regulation of the cell cycle, which makes the tumor cells more malignant.


Subject(s)
Lymphoma, Non-Hodgkin/metabolism , Microfilament Proteins/metabolism , Muscle Proteins , Adolescent , Adult , Aged , Aged, 80 and over , Child , Female , Genes, Tumor Suppressor , Humans , Lymph Nodes/metabolism , Lymphoma, Non-Hodgkin/genetics , Male , Microfilament Proteins/genetics , Middle Aged
19.
Ai Zheng ; 22(3): 266-9, 2003 Mar.
Article in Chinese | MEDLINE | ID: mdl-12654183

ABSTRACT

BACKGROUND & OBJECTIVE: Signal cascades of mitogen-actived protein kinase(MAPK) and signal transducer and activator of transcription 3 (Stat3) are two main signal transduction pathways which were associated with cell proliferation and malignant transformation.MAPK and Stat3 proteins are activated by phosphorylation. The biological effects which are caused by multiple cytokines produced by Hodgkin's lymphoma(HL) cells are mediated through MAPK and Stat3 signal pathways. This study was designed to investigate significance of MAPK and Stat3 phosphorylation(p-MAPK and p-Stat3) and cyclin D1 protein expression in HL. METHODS: SP immunohistochemistry was used to detect expression of p-MAPK, p-Stat3, and cyclin D1 protein in 45 cases of HL of various types. RESULTS: The expression positive rates of p-MAPK, p-Stat3, and cyclin D1 proteins were 73.3%(33/45),64.4%(29/45), and 68.9%(31/45), respectively. The positive expression levels of p-MAPK and cyclin D1 protein gradually increased(P< 0.05), whereas that of p-stat3 had no significant difference(P >0.05) in four subsets(LR:lymphocyte-rich classical type; NS:nodular sclerosis type; MC:mixed cellularity type; LD:lymphocyte depletion type) of all cases. The expression of p-MAPK was positively related to that of cyclin D1 protein (r(s)=0.7254,P< 0.01), but the expression of p-Stat3 was not related to that of cyclin D1 protein (r(s)=0.2197,P >0.05). CONCLUSION: The data suggest that activation of MAPK may play an important role in genesis and progression of HL, but Stat3 activation is not associated with the progression of HL. MAPK may induce overexpression of cyclin D1 protein and results in persistent proliferation of RS/H cells in genesis and development of HL.


Subject(s)
Cyclin D1/biosynthesis , DNA-Binding Proteins/biosynthesis , Hodgkin Disease/metabolism , Lymphoma/metabolism , Mitogen-Activated Protein Kinases/metabolism , Trans-Activators/biosynthesis , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Immunohistochemistry , MAP Kinase Signaling System , Male , Middle Aged , Phosphorylation , STAT3 Transcription Factor , Signal Transduction , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...