Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Article in English | MEDLINE | ID: mdl-37702239

ABSTRACT

BACKGROUND: Dedicator for cytokinesis 4 (DOCK4) is a guanine nucleotide exchange factor (GEF) for the small GTPase Rac1. However, the functions of DOCK4 concerning the tumor microenvironment (TME) in colon adenocarcinoma (COAD) remain uncertain. METHODS: The TIMER and GEPIA databases were used to analyze the DOCK4 expression between COAD tissues and adjunct normal tissues. The PrognoScan database was used to assess the prognosis of DOCK4 expression in COAD. The co-expression networks of DOCK4 in COAD were constructed by the LinkedOmics website. Furthermore, the correlation between DOCK4 expression and TME of COAD was explored using TIMER and TISIDB databases. Finally, the clone formation assay was used to further verify the function of DOCK4 in COAD. The Western blotting assay was used to confirm the mechanism related to DOCK4 in COAD. RESULTS: The DOCK4 expression was different significantly in COAD tissues and paracancerous tissues. The DOCK4 was found to play a poor role in the prognosis of patients with COAD. The DOCK4 was found to participate in the TME by promoting immune evasion of COAD. The reduction of DOCK4 expression inhibited the clone formation and Ras-associated protein 1A (Rap1A) expression of HCT116 cells. CONCLUSIONS: DOCK4 potentially plays an important role in the regulation of TME in COAD. DOCK4 facilitates the development through the Rap1A pathway, thus becoming a novel prognostic biomarker in COAD.

2.
Comput Math Methods Med ; 2022: 8234769, 2022.
Article in English | MEDLINE | ID: mdl-35915770

ABSTRACT

Objective: To explore the application value of the nutrition support team in chemotherapy period of colon cancer based on the internet multidisciplinary treatment mode. Methods: For the method of retrospective study, 90 patients with colon cancer admitted to our hospital from August 2018 to August 2020 were selected as the study subjects. They were equally divided into the experimental group (n = 45) and the control group (n = 45) according to the order of initials and the method of parity group. The control group was given conventional nutrition support, and the experimental group was given the nutrition support under the internet multidisciplinary treatment mode. The serum tumor marker levels (CEA and CA19-9), immune function indexes, nutrition indicators, and the incidence of adverse reactions were compared between the two groups before and after intervention. Results: The serum tumor marker levels in the experimental group after intervention were significantly lower than those in the control group (P < 0.001). The immune function indexes in the experimental group after intervention were significantly better than those in the control group (P < 0.001). The nutrition indicators in the experimental group after intervention were significantly better than those in the control group (P < 0.001). The incidence of gastrointestinal adverse reactions above grade 2 in the experimental group was significantly lower than that in the control group (P < 0.05). There were 20 patients with myelosuppression, 2 patients with neurotoxicity, and 1 patient with hand and foot syndrome in the experimental group, while 22 patients with myelosuppression, 4 patients with neurotoxicity, and 2 patients with hand and foot syndrome in the control group, with no significant difference in the incidence of adverse reactions between the two groups (P > 0.05). Conclusion: The nutrition support team under the internet multidisciplinary treatment mode can improve the immune function of chemotherapy patients with colon cancer and enhance their nutritional level, thereby reducing the incidence of adverse reactions and improving the chemotherapy effects.


Subject(s)
Colonic Neoplasms , Enteral Nutrition , Biomarkers, Tumor , Colonic Neoplasms/drug therapy , Humans , Internet , Retrospective Studies
4.
Int J Cancer ; 124(11): 2709-18, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19219913

ABSTRACT

Lymph nodes metastasis of tumor could be a crucial early step in the metastatic process. Induction of tumor lymphangiogenesis by vascular endothelial growth factor-D may play an important role in promoting tumor metastasis to regional lymph nodes and these processes can be inhibited by inactivation of the VEGFR-3 signaling pathway. Honokiol has been reported to possess potent antiangiogenesis and antitumor properties in several cell lines and xenograft tumor models. However, its role in tumor-associated lymphangiogenesis and lymphatic metastasis remains unclear. Here, we established lymph node metastasis models by injecting overexpressing VEGF-D Lewis lung carcinoma cells into C57BL/6 mice to explore the effect of honokiol on tumor-associated lymphangiogenesis and related lymph node metastasis. The underlying mechanisms were systematically investigated in vitro and in vivo. In in vivo study, liposomal honokiol significantly inhibited the tumor-associated lymphangiogenesis and metastasis in Lewis lung carcinoma model. A remarkable delay of tumor growth and prolonged life span were also observed. In in vitro study, honokiol inhibited VEGF-D-induced survival, proliferation and tube-formation of both human umbilical vein endothelial cells (HUVECs) and lymphatic vascular endothelial cells (HLECs). Western blotting analysis showed that liposomal honokiol-inhibited Akt and MAPK phosphorylation in 2 endothelial cells, and downregulated expressions of VEGFR-2 of human vascular endothelial cells and VEGFR-3 of lymphatic endothelial cells. Thus, we identified for the first time that honokiol provided therapeutic benefit not only by direct effects on tumor cells and antiangiogenesis but also by inhibiting lymphangiogenesis and metastasis via the VEGFR-3 pathway. The present findings may be of importance to investigate the molecular mechanisms underlying the spread of cancer via the lymphatics and explore the therapeutical strategy of honokiol on antilymphangiogenesis and antimetastasis.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Biphenyl Compounds/administration & dosage , Lignans/administration & dosage , Lymphangiogenesis/drug effects , Lymphatic Metastasis/prevention & control , Vascular Endothelial Growth Factor D/antagonists & inhibitors , Animals , Apoptosis/drug effects , Biphenyl Compounds/pharmacology , Carcinoma, Lewis Lung/drug therapy , Cell Line , Endothelial Cells/drug effects , Endothelial Cells/physiology , Female , Humans , Lignans/pharmacology , Liposomes , Mice , Mice, Inbred C57BL , Vascular Endothelial Growth Factor D/physiology , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Xenograft Model Antitumor Assays
5.
Anticancer Drugs ; 16(5): 551-7, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15846121

ABSTRACT

Endostatin is an important endogenous inhibitor of neovascularization, which has been widely used in anti-angiogenesis therapy for cancer. To fully explore the potential of endostatin, we evaluated the anti-tumor efficacy of the combination of recombinant human endostatin adenovirus and low-dose gemcitabine in nude mice. We injected recombinant human endostatin adenovirus intratumorally plus a low dose of gemcitabine i.p. routinely. The combination treatment produced no side-effects, and resulted in marked suppression in tumor formation and growth of established human lung carcinoma xenografts in nude mice, with decreased microvessel density and increased apoptosis percentage. Our data support the idea of synergistic anti-tumor properties of endostatin plus low-dose chemotherapy against human lung cancer in vivo.


Subject(s)
Antineoplastic Agents/therapeutic use , Deoxycytidine/analogs & derivatives , Endostatins/therapeutic use , Genetic Therapy , Neoplasms, Experimental/drug therapy , Neovascularization, Pathologic/prevention & control , Adenoviridae/genetics , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Deoxycytidine/administration & dosage , Deoxycytidine/therapeutic use , Drug Synergism , Endostatins/administration & dosage , Endostatins/genetics , Female , Genetic Vectors , Humans , Injections, Intralesional , Injections, Intraperitoneal , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms, Experimental/blood supply , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Xenograft Model Antitumor Assays , Gemcitabine
6.
Int J Cancer ; 112(1): 143-9, 2004 Oct 20.
Article in English | MEDLINE | ID: mdl-15305386

ABSTRACT

Vesicular stomatitis virus (VSV) has been shown to replicate rapidly in vitro and kill selectively a variety of tumor cell lines. The present study was designed to determine whether gemcitabine potentiates the antitumor activity of VSV in vitro and in vivo. A549 human lung adenocarcinoma cells and LLC Lewis lung carcinoma cells were treated with VSV (0.1-10 plaque-forming units per cell) plus gemcitabine (20 nM to 20 microM). Mice bearing A549 or LLC were treated with VSV (5 x 10(4) to 1 x 10(8) plaque-forming units) daily for 5 days plus gemcitabine (5-125 mg/kg/day) once every 3 days for 4 times. Induction of apoptosis and effects on growth inhibition were assessed. The lung cancer cells treated with VSV plus gemcitabine displayed the apparently increased apoptotic cells compared with treatment with VSV or gemcitabine alone. The combined treatment with VSV plus gemcitabine induced the apparent antitumor activity with complete regression of the established lung cancer in both A549 and LLC lung cancer models and augmented the induction of apoptosis in lung cancer cells in vivo as well. This study suggests that the combined treatment with VSV plus gemcitabine may augment the induction of apoptosis in lung cancer cells in vitro and in vivo, and that the augmented antitumor activity in vivo may result from the increased induction of apoptosis in lung cancer cells. The present findings may be of importance to the further exploration of the potential application of this combined approach in the treatment of lung cancer.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Apoptosis , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Lung Neoplasms/prevention & control , Vesicular stomatitis Indiana virus/physiology , Adenocarcinoma/prevention & control , Adenocarcinoma/virology , Animals , Carcinoma, Lewis Lung/prevention & control , Carcinoma, Lewis Lung/virology , Combined Modality Therapy , Humans , In Vitro Techniques , Lung Neoplasms/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Tumor Cells, Cultured , Vesicular stomatitis Indiana virus/isolation & purification , Gemcitabine
7.
Immunol Invest ; 31(1): 51-69, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11990463

ABSTRACT

The breaking of immune tolerance against angiogenesis-associated molecules should be a useful approach for cancer therapy by active immunity. We used chicken integrin beta3 as a model antigen to explore the feasibility of immunogene therapy of the tumors with a vaccine based on a single xenogeneic homologous gene, targeting the molecules associated with angiogenesis. To test this concept we constructed a plasmid DNA encoding the ligand-binding domain of chicken integrin beta3 (P-BD-C) and control vectors. We found that immunogene therapy of tumors with a vaccine based on the ligand-binding domain of chicken integrin beta3 (P-BD-C) was effective in both protective and therapeutic anti-tumor immunity in several tumor models in mice. Autoantibodies against integrin beta3 in sera of mice immunized with the ligand-binding domain of chicken integrin beta3 could be found by Western blot analysis and ELISA assay. The purified immunoglobulins were effective in the inhibition of endothelial cell proliferation in vitro, and in anti-tumor activity as well as in the inhibition of angiogenesis by adoptive transfer in vivo. The anti-tumor activity and the production of integrin beta3-specific autoantibodies (manifested by significantly elevated Ig G1 and Ig G2b) could be abrogated by the depletion of CD4+ T lymphocytes. These observations may provide a vaccine strategy for cancer therapy through the induction of the autoimmunity against the molecules associated with tumor growth in a cross-reaction with a single xenogeneic homologous gene and may be of importance in the further exploration of the applications of other xenogeneic homologous genes identified in human and other animal genome sequence projects in cancer therapy.


Subject(s)
Cancer Vaccines/immunology , Integrin beta3/genetics , Vaccines, DNA/immunology , Animals , Autoantibodies/immunology , CD4-Positive T-Lymphocytes/immunology , COS Cells , Chick Embryo , Chlorocebus aethiops , Integrin beta3/immunology , Ligands , Mice , Neovascularization, Pathologic/immunology , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL
...