Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Neuroscience ; 549: 84-91, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38460904

ABSTRACT

We aimed to evaluate the role of the spinal lymphatic system in spinal cord injury and whether it has an impact on recovery after spinal cord injury. Flow cytometry was used to evaluate the changes in the number of microvesicles after spinal cord injury. Evans blue extravasation was used to evaluate the function of the lymphatic system. Evans blue extravasation and immunofluorescence were used to evaluate the permeability of blood spinal cord barrier. The spinal cord edema was evaluated by dry and wet weight.Terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay was used to evaluate apoptosis after spinal cord injury. Nuclear factor-kappa B pathway was detected by Western blot. Behavioral tests were used to evaluate limb function. Microvesicles released after spinal cord injury can enter the thoracic duct and then enter the blood through the lymph around the spine. After ligation of the thoracic duct, it can aggravate the neuropathological manifestations and limb function after spinal cord injury. The potential mechanism may involve nuclear factor-kappa B pathway.


Subject(s)
Recovery of Function , Spinal Cord Injuries , Spinal Cord , Spinal Cord Injuries/physiopathology , Spinal Cord Injuries/pathology , Spinal Cord Injuries/metabolism , Animals , Recovery of Function/physiology , Spinal Cord/metabolism , Spinal Cord/pathology , Spinal Cord/physiopathology , NF-kappa B/metabolism , Male , Apoptosis/physiology , Rats, Sprague-Dawley , Disease Models, Animal , Lymphatic System/physiopathology , Lymphatic System/pathology , Edema/pathology , Thoracic Duct/physiopathology , Female , Cell-Derived Microparticles/metabolism
2.
J Hematol Oncol ; 13(1): 142, 2020 Oct 22.
Article in English | MEDLINE | ID: mdl-33092612

ABSTRACT

An amendment to this paper has been published and can be accessed via the original article.

3.
Oncol Lett ; 20(4): 61, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32863894

ABSTRACT

The epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) contribute to an increased response rate, compared with chemotherapy, in patients with inhibitor-sensitive EGFR mutations. The present study evaluated the association between the maximum standardized uptake value (SUVmax) of 18F-fluorodeoxyglucose positron emission tomography-computed tomography (FDG PET/CT), as well as serum carcinoembryonic antigen (CEA) levels and EGFR mutations prior to treatment, in patients with non-small cell lung cancer (NSCLC). Patients with histologically confirmed NSCLC (n=167), who underwent an 18F-FDG PET/CT scan, EGFR mutation analysis and a serum CEA test participated in the present study. Multivariate logistic regression analysis was used to analyze predictors of EGFR mutations. Receiver-operating characteristic (ROC) curve analysis was performed to determine the efficient cut-off value. Survival rate analysis was evaluated according to SUVmax and EGFR mutation status. A decreased SUVmax and an increased CEA level was observed in patients with EGFR-mutations, compared with patients with wild-type primary lesions and metastatic lymph nodes. The exon 19 EGFR mutation was associated with increased SUVmax, compared with the exon 21 L858R mutation. The ROC analysis indicated that an 18F-FDG PET/CT uptake SUVmax >11.5 may be a predictor of the wild-type EGFR genotype and increased CEA levels (CEA >9.4 ng/ml) were associated with EGFR mutations. Furthermore, patients with no smoking history, low SUVmax of the primary tumor, metastatic lymph nodes and a high CEA level were significantly associated with EGFR mutation status. The results of the present study indicated that patients with advanced NSCLC, particularly Chinese patients, with decreased SUVmax and increased CEA levels are associated with EGFR mutations, which may serve as predictors for the EGFR-TKI therapeutic response.

4.
Curr Med Sci ; 39(6): 965-971, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31845228

ABSTRACT

Shenqi Fuzheng injection (SFI) has been confirmed to be able to alleviate brain injury in mice. This study examined the brain-protective effect of SFI on patients after cranial radiation. Lung cancer patients with brain metastasis were randomly assigned to two groups. The SFI group received cranial radiation in combination with SFI. The control group received cranial radiation alone. The changes in cognitive function were evaluated pre- and post-radiation against the Mini-Mental State Exam (MMSE), Montreal Cognitive Assessement (MoCA), Zung Self-Rating Depression Scale (SDS) and Zung Self-Rating Anxiety Scale (SAS). The changes in inflammatory factors, such as TGF-ß1, TNF-α and IL-10, were also detected before, during and after radiation (15Gy/5F). The results showed that 6 months after cranial radiation, the total scores on the MMSE and MoCA scales of the patients decreased, especially memory ability. The control group experienced a more evident decline, the memory ability being the greatest. TGF-ß1 and TNF-a increased shortly after radiation and decreased one month later, and the change was more conspicuous in SFI group than in control group. IL-10 increased after radiation and stayed at a high level one month later in both groups, the level being higher in the SFI group than in the control group. Our study indicated that cognitive functions, especially memory ability, were impaired after cranial radiation. SFI could alleviate radiation-induced brain injury by regulating inflammatory factors.


Subject(s)
Brain Injuries/drug therapy , Brain Neoplasms/radiotherapy , Brain Neoplasms/secondary , Drugs, Chinese Herbal/administration & dosage , Lung Neoplasms/radiotherapy , Radiation Injuries/prevention & control , Adult , Aged , Animals , Brain Injuries/etiology , Brain Injuries/metabolism , Brain Neoplasms/metabolism , Cognition/drug effects , Cytokines/metabolism , Down-Regulation , Drugs, Chinese Herbal/pharmacology , Female , Humans , Injections , Lung Neoplasms/metabolism , Male , Middle Aged , Radiation Injuries/metabolism , Random Allocation , Treatment Outcome
5.
Brain Behav Immun ; 81: 111-121, 2019 10.
Article in English | MEDLINE | ID: mdl-31176001

ABSTRACT

PURPOSE: Elevated catecholamines in the tumor microenvironment often correlate with tumor development. However, the mechanisms by which catecholamines modulate lung cancer growth are still poorly understood. This study is aimed at examining the functions and mechanisms of catecholamine-induced macrophage polarization in angiogenesis and tumor development. EXPERIMENTAL DESIGN: We established in vitro and in vivo models to investigate the relationship between catecholamines and macrophages in lung cancer. Flow cytometry, cytokine detection, tube formation assay, immunofluorescence, and western blot analysis were performed, and animal models were also used to explore the underlying mechanism of catecholamine-induced macrophage polarization and host immunological response. RESULTS: Catecholamines were shown to be secreted into tumor under the control of the sympathetic nerve system to maintain the pro-tumoral microenvironment. In vivo, the chemical depletion of the natural catecholamine stock with 6OHDA could reduce the release of catecholamines within tumor tissues, restrain the function of alternatively activated M2 macrophage, attenuate tumor neovascularization, and inhibit tumor growth. In vitro, catecholamine treatment triggered the M2 polarization of macrophages, enhanced the expression of VEGF, promoted tumor angiogenesis, and these catecholamine-stimulated effects could be reversed by the adrenergic receptor antagonist propranolol. In addition to regulating tumor-associated macrophages (TAM) recruitment, decreasing catecholamine levels could also shift the immunosuppressive microenvironment by decreasing myeloid-derived suppressor cells' (MDSCs) recruitment and facilitating dendritic cells' (DCs) activation, potentially resulting in a positive antitumor immune response. CONCLUSION: Our study demonstrates the potential of adrenergic stress and catecholamine-driven adrenergic signaling of TAMs to regulate the immune status of a tumor microenvironment and provides promising targets for anticancer therapies.


Subject(s)
Catecholamines/metabolism , Lung Neoplasms/metabolism , Neovascularization, Pathologic/physiopathology , Animals , Catecholamines/physiology , Cell Line, Tumor , Cell Movement/immunology , Cytokines/metabolism , Humans , Macrophage Activation/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred BALB C , Signal Transduction , Tumor Microenvironment
6.
Int J Cancer ; 144(9): 2227-2238, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30374974

ABSTRACT

Vasculogenic mimicry (VM) is a special vascular pattern in malignant tumors, which is composed of highly aggressive tumor cells. This tumor cell-mediated blood supply pattern is closely associated with a poor prognosis in cancer patients. The interaction of axon guidance factor Sema4D and its high affinity receptor plexinB1 could activate small GTPase RhoA and its downstream ROCKs; this process has an active role in the migration of endothelial cells and tumor angiogenesis. Here, we have begun to uncover the role of this pathway in VM formation in non-small cell lung cancer (NSCLC). First, we confirmed this special form of vasculature in NSCLC tissues and found the existence of VM channels in tumor tissues was correlated with Sema4D expression. Further, we found that inhibition of Sema4D in the human NSCLC cells H1299 and HCC827 reduces VM formation both in vitro and in vivo. Moreover, we demonstrated that downregulating the expression of plexinB1 by siRNA expressing vectors and inhibiting the RhoA/ROCK signaling pathway using fasudil can reduce VM formation of H1299 and HCC827 cells. Finally, we found that suppression of Sema4D leads to less stress fibers and depleted the motility of H1299 and HCC827 cells. Collectively, our study implicates Sema4D plays an important role in the process of VM formation in NSCLC through activating the RhoA/ROCK pathway and regulating tumor cell plasticity and migration. Modulation of the Sema4D/plexinB1 and downstream RhoA/ROCK pathway may prevent the tumor blood supply through the VM pattern, which may eventually halt growth and metastasis of NSCLC.


Subject(s)
Antigens, CD/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Neovascularization, Pathologic/pathology , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Semaphorins/metabolism , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , A549 Cells , Animals , Cell Line, Tumor , Cell Movement/physiology , Endothelial Cells/physiology , Enzyme Activation , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Nerve Tissue Proteins/genetics , Protein Kinase Inhibitors/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Receptors, Cell Surface/genetics , Semaphorins/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
8.
J Hematol Oncol ; 10(1): 125, 2017 06 19.
Article in English | MEDLINE | ID: mdl-28629431

ABSTRACT

BACKGROUND: Brain metastasis (BM) is associated with poor prognosis in patients with non-small cell lung cancer (NSCLC). Recent studies demonstrated that microRNA-330-3p (miR-330-3p) was involved in NSCLC brain metastasis (BM). However, the exact parts played by miR-330-3p in BM of NSCLC remain unknown. Discovery and development of biomarkers and elucidation of the mechanism underlying BM in NSCLC is critical for effective prophylactic interventions. Here, we evaluated the expression and biological effects of miR-330-3p in NSCLC cells and explored the underlying mechanism of miR-330-3p in promoting cell migration and invasion in NSCLC. METHODS: Stable over-expression and knockdown of miR-330-3p in NSCLC cells was constructed with lentivirus. Expression levels of miR-330-3p in NSCLC cells were quantified by quantitive real-time PCR (qRT-PCR). The effects of miR-330-3p on NSCLC cells were investigated using assays of cell viability, migration, invasion, cell cycle, apoptosis, western blotting, immunohistochemical, and immunofluorescence staining. A xenograft nude mouse model and in situ brain metastasis model were used to observe tumor growth and brain metastasis. The potential target of miR-330-3p in NSCLC cells was explored using the luciferase reporter assay, qRT-PCR, and western blotting. The miR-330-3p targets were identified using bioinformatics analysis and verified by luciferase reporter assay. The correlation between GRIA3 and DNA methyltransferase (DNMT) 1 and DNMT3A was tested by RT-PCR, western blotting, and co-immunoprecipitation (IP). RESULTS: miR-330-3p was significantly up-regulated in NSCLC cell lines. MTT assay, transwell migration, and invasion assays showed that miR-330-3p promoted the growth, migration, and invasion of NSCLC cells in vitro and induced tumor growth and metastasis in vivo. Luciferase reporter assays showed that GRIA3 was a target of miR-330-3p. qRT-PCR and western blotting exhibited that miR-330-3p promoted the growth, invasion, and migration of NSCLC cells by activating mitogen-activated protein kinase (MAPK)/extracellular-regulated protein kinases (ERK) signaling pathway. Furthermore, miR-330-3p up-regulated the total DNA methylation in NSCLC cells, and co-IP-demonstrated GRIA3 was directly related with DNMT1 and DNMT3A. CONCLUSIONS: miR-330-3p promoted the progression of NSCLC and might be a potential target for the further research of NSCLC brain metastasis.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , MAP Kinase Signaling System , MicroRNAs/genetics , Receptors, AMPA/genetics , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA Methylation , Enzyme Activation , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Up-Regulation
9.
J Huazhong Univ Sci Technolog Med Sci ; 36(6): 916-922, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27924504

ABSTRACT

To better understand the outcomes of small cell lung cancer (SCLC), we examined the clinical features and prognostic factors of SCLC in this study. A total of 148 patients who were diagnosed as having SCLC between January 2009 and December 2013 in Cancer Center of Union Hospital, Wuhan, China, were enrolled and their clinical features and prognostic factors were retrospectively analyzed. Log-rank test and Cox regression model were employed for analysis of prognostic factors. The 1- and 2-year overall survival (OS) rates were 59.7% and 25.7%, respectively, for limited disease (LD) patients whose median survival time (MST) was 16 months. The 1- and 2-year OS rates were 29.5% and 5.3%, respectively, for extensive disease (ED) patients whose MST was 10 months. The univariate analysis and multivariate analysis revealed that age, tumor stage, serum CEA and Ki-67 antigen were significantly correlated to the outcomes of SCLC, and they were significant prognostic factors for SCLC.


Subject(s)
Lung Neoplasms/pathology , Small Cell Lung Carcinoma/pathology , Adult , Aged , Aged, 80 and over , Female , Humans , Ki-67 Antigen/blood , Lung Neoplasms/blood , Lung Neoplasms/epidemiology , Male , Middle Aged , Neoplasm Staging , Small Cell Lung Carcinoma/blood , Small Cell Lung Carcinoma/epidemiology , Survival Analysis
10.
J Huazhong Univ Sci Technolog Med Sci ; 36(1): 88-94, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26838746

ABSTRACT

In patients with advanced cancer, cancer-induced bone pain (CIBP) is a severe and common problem that is difficult to manage and explain. As c-Jun N-terminal kinase (JNK) and chemokine (C-X-C motif) ligand 1 (CXCL1) have been shown to participate in several chronic pain processes, we investigated the role of JNK and CXCL1 in CIBP and the relationship between them. A rat bone cancer pain model was established by intramedullary injection of Walker 256 rat gland mammary carcinoma cells into the left tibia of Sprague-Dawley rats. As a result, intramedullary injection of Walker 256 carcinoma cells induced significant bone destruction and persistent pain. Both phosphorylated JNK1 (pJNK1) and pJNK2 showed time-dependent increases in the ipsilateral spinal cord from day 7 to day 18 after tumor injection. Inhibition of JNK activation by intrathecal administration of SP600125, a selective pJNK inhibitor, attenuated mechanical allodynia and heat hyperalgesia caused by tumor inoculation. Tumor cell inoculation also induced robust CXCL1 upregulation in the ipsilateral spinal cord on day 18 after tumor injection. Inhibition of CXCL1 by intrathecal administration of CXCL1 neutralizing antibody showed a stable analgesic effect. Intrathecal administration of SP600125 reduced CXCL1 increase in the spinal cord, whereas inhibition of CXCL1 in the spinal cord showed no influence on JNK activation. Taken together, these results suggested that JNK activation in spinal cord contributed to the maintenance of CIBP, which may act through modulation of CXCL1. Inhibition of the pJNK/CXCL1 pathway may provide a new choice for treatment of CIBP.


Subject(s)
Bone Neoplasms/metabolism , Cancer Pain/metabolism , Chemokine CXCL1/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Spinal Cord/metabolism , Animals , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/therapeutic use , Bone Neoplasms/complications , Cancer Pain/drug therapy , Cancer Pain/etiology , Cell Line, Tumor , Chemokine CXCL1/immunology , Female , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Rats , Rats, Sprague-Dawley
11.
Mol Cancer Ther ; 14(7): 1582-90, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25934709

ABSTRACT

The aim of this study was to investigate the biologic role of the Rho kinase inhibitor fasudil in the vasculogenic mimicry (VM) of B16 mouse melanoma cells. It was previously reported that RhoA plays a critical role in angiogenesis by coordinating endothelial cell cytoskeleton remodeling and promoting endothelial cell motility. Although RhoA has been implicated in the regulation of angiogenesis, little has been described regarding its control of these tumor cell-lined channels. In this study, we established an in vitro model of VM using 3-dimensional cell culturing of mouse B16 melanoma cells and studied VM in vivo by transplanting B16 cells into C57/BL mice. Next, we explored the effect of RhoA and Rho-associated, coiled-coil containing protein kinase (ROCK) on VM formation using the Rho kinase inhibitor fasudil. We provide direct evidence that fasudil leads to reduced vascular-like channels in Matrigel. Additional experiments suggested that fasudil prevents both initial cellular architecture changes and cell migration in vitro. Finally, we provide in-depth evidence for the underlying mechanisms of fasudil-induced VM destruction using the Rho-GTPase agonist lysophosphatidic acid. In vivo studies revealed that fasudil reduced B16 melanoma cell xenograft tumor growth without causing significant toxicity in mice. Fasudil-treated tumors also displayed fewer VM channels. These results suggest that fasudil may be an emerging therapeutic option for targeting cancer VM.


Subject(s)
1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Melanoma, Experimental/drug therapy , Neovascularization, Pathologic/prevention & control , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Lysophospholipids/pharmacology , Male , Melanoma, Experimental/blood supply , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Microscopy, Confocal , Neoplasm Transplantation/methods , Protein Kinase Inhibitors/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/drug effects , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
12.
Int J Radiat Biol ; 89(9): 724-31, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23682582

ABSTRACT

PURPOSE: Sorafenib, an oral multikinase inhibitor, is the first agent that has demonstrated an improved overall survival benefit in advanced hepatocellular carcinoma, setting a new standard for first-line treatment. However, the association between radiosensitivity and sorafenib remains unclear. The purpose of this study was to investigate whether sorafenib could enhance radiosensitivity and the possible mechanisms of sorafenib-mediated radiosensitization in human hepatocellular carcinoma cell line SMMC-7721. EXPERIMENTAL DESIGN: The cell viability of human hepatocellular carcinoma cell line SMMC-7721 was determined by the 3-(4, 5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays. The radiosensitization of sorafenib in SMMC-7721 cells was evaluated by clonogenic assays, and immunofluorescence for DNA double-strand break detection, and Western blotting for protein detection. RESULTS: The MTT results clearly showed that sorafenib affected cell viability in human hepatocellular cell line SMMC-7721. Sorafenib administered 1 h before the radiation treatment resulted in radiosensitization with a sensitivity enhancement ratio (SER) of 1.65 as shown by clonogenic assays. Furthermore, sorafenib pretreatment led to decreased phosphorylation levels of extracellular signal-regulated kinase (ERK) and protein kinase B (AKT) in SMMC-7721 cells. Interestingly, pretreatment of mitogen-activated protein kinase kinases/extracellular signal-regulated kinase (MEK/ERK) signaling inhibitor U0126 had a similar effect as that of sorafenib pretreatment in hepatocellular carcinoma cells, whereas pretreatment of phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) signaling inhibitor LY294002 in the same cells had no effect on radiosensitization. The group treated with sorafenib and radiation was more sensitive to irradiation alone as demonstrated by the results of the DNA double-strand break detection. CONCLUSIONS: The combination of sorafenib and radiation affected cell viability more effectively than radiation alone. Sorafenib significantly enhanced the sensitivity of SMMC-7721 cells to radiation showing significantly reduced repair of DNA double-strand breaks. The MEK/ERK signaling pathway may be a pathway responsible for the radiosensitivity enhancement of sorafenib. Our data provided experimental support for the possible combination of sorafenib with radiation for the treatment of hepatocellular carcinoma and other cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , MAP Kinase Signaling System/radiation effects , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Radiation-Sensitizing Agents/pharmacology , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Repair/drug effects , Extracellular Signal-Regulated MAP Kinases , Humans , Liver Neoplasms/pathology , Mitogen-Activated Protein Kinase Kinases , Niacinamide/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Sorafenib
13.
Zhonghua Zhong Liu Za Zhi ; 33(4): 251-5, 2011 Apr.
Article in Chinese | MEDLINE | ID: mdl-21575493

ABSTRACT

OBJECTIVE: To determine the effect of exogenous GM3 on proliferation, apoptosis and VEGF expression in human lung adenocarcinoma cell line A549 cells. METHODS: A549 cells were treated with GM3 at different concentrations for 48 hours. MTT assay was used to detect the cell proliferation and flow cytometry was applied to analyze cell apoptosis. RT-PCR was used to detect the expression level of VEGF mRNA and confocal laser scanning microscopy was applied to observe the localization and fluorescence intensity of VEGF. RESULTS: Comparing with the control, being treated with higher than 10 µmol/L GM3 significantly inhibited A549 cell proliferation (P < 0.05), and the suppressive effect could be enhanced following increasing doses. The IC(50) was 412 µmol/L. Comparing with the control, being treated with higher than 40 µmol/L GM3 significantly promoted the apoptotic rate of A549 cells (P < 0.05). Comparing with the control, being treated with higher than 40 µmol/L GM3 significantly decreased the VEGF mRNA level of A549 cells (P < 0.05), and the fluorescence intensity of VEGF distinctly weakened. CONCLUSIONS: Exogenous ganglioside GM3 can inhibit the proliferation, promote apoptosis, and down-regulate the VEGF expression level in A549 cells. This may be considered as two mechanisms of GM3 for its anti-tumor effect by modulating cell apoptosis and angiogenesis.


Subject(s)
Adenocarcinoma/pathology , Apoptosis/drug effects , Cell Proliferation/drug effects , G(M3) Ganglioside/pharmacology , Lung Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma of Lung , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Dose-Response Relationship, Drug , Down-Regulation , G(M3) Ganglioside/administration & dosage , Humans , Inhibitory Concentration 50 , Lung Neoplasms/metabolism , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/genetics
14.
Tumour Biol ; 32(4): 677-87, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21442281

ABSTRACT

Recombinant human endostatin (rh-endostatin), a potential antiangiogenic agent, is used in non-small cell lung carcinoma treatment and represses vascular endothelial cell growth factor (VEGF) levels in tumor cell. However, precise affection of rh-endostatin on the proangiogenic VEGF isoforms (VEGF(165)) or antiangiogenic VEGF isoforms (VEGF(165)b) is not clear. We therefore tested the hypothesis that rh-endostatin could alter expression of these isoforms to regulate tumor growth. A549 cells were exposed to rh-endostatin, and the expression of VEGF(165) and VEGF(165)b was detected. The role of SP1 as a regulator of isoform expression was investigated. We then examined the anticancer and antiangiogenic efficacy of rh-endostatin in combination with exogenous VEGF(165)b against A549 cells, EA.HY 926 cells and xenograft model of human lung cancer. rh-Endostatin reduced VEGF(165) and induced VEGF(165)b as well as inhibited SP1 in A549 cells. SP1 inhibitor (betulinic acid) also developed those changes. VEGF(165)b-rh-endostatin combination was highly synergistic and inhibited growth, survival, and migration of A549 cells, VEGF-mediated VEGFR2 phosphorylation in EA.HY 926 cells, and tumor growth in xenograft model of human lung cancer. rh-Endostatin downregulates proangiogenic vascular endothelial growth factor A (VEGFA) isoform and upregulates antiangiogenic VEGFA isoform, possibly through inhibition of SP1. Furthermore, VEGF(165)b sensitizes A549 to rh-endostatin treatment and enhances the anticancer effect of rh-endostatin.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Endostatins/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Animals , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunohistochemistry , Mice , Mice, Nude , Neovascularization, Pathologic/metabolism , Protein Isoforms/biosynthesis , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation , Xenograft Model Antitumor Assays
15.
Med Oncol ; 28(3): 673-8, 2011 Sep.
Article in English | MEDLINE | ID: mdl-20354813

ABSTRACT

Overall outcome of those patients with non-small cell lung cancer (NSCLC) remains poor. Recently, several studies demonstrated that cyclin-dependent kinase-5 (CDK5) activity with its specific activator protein p35 was important for spontaneous metastasis in various types of carcinomas. Our objective was to explore the expression of CDK5 and its prognostic indicator in patients with NSCLC. Immunofluorescent staining was used to detect the expression of CDK5/p35 in the lung tissue of 95 patients with NSCLC and 20 patients with benign pulmonary disease. The correlation between the expression of CDK5/p35 and clinicopathologic features of patients with NSCLC was investigated. The 5-year overall survival of patients with tumors expressing different levels of CDK5/p35 was evaluated by the Kaplan-Meier method. Positive expressions of CDK5/p35 were detected in the tumor cells in 66 samples (69.5%) of the 95 patients with NSCLC. Although no remarkable correlation between CDK5/p35 expression and age at the time of surgery, gender, and histopathologic type, there were significant differences between CDK5/p35 expression and degree of differentiation, pathological stage and lymph node metastasis in patients with NSCLC. In addition, we demonstrated that median survival for patients with and without CDK5/p35 expression was 24 and 58 months, respectively, and 5-year overall survival rate 25.8 and 48.3%, respectively (P<0.05). Patients with lung cancer with a positive CDK5/p35 expression had a poorer prognosis than those with a negative CDK5/p35 expression. Based on our results, CDK5/p35 may represent a biomarker for prognosis in patients with NSCLC.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Non-Small-Cell Lung/metabolism , Cyclin-Dependent Kinase 5/biosynthesis , Lung Neoplasms/metabolism , Adaptor Proteins, Signal Transducing , Adult , Aged , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Cycle Proteins , Female , Fluorescent Antibody Technique , Humans , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , Retrospective Studies
16.
Cancer Biother Radiopharm ; 25(1): 75-80, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20187799

ABSTRACT

Activation of autophagy is a hallmark in tumor cells treated with chemotherapy, but the role of autophagy in acquired resistance of lung adenocarcinoma to cisplatin-based chemotherapy remains to be clarified. Our aim was to address that question by surveying the autophagic activity in parental lung adenocarcinoma cell line A549 and its 8-fold, more resistant subcell line, A549/DDP, which was obtained by treating cisplatin with increasing concentrations. A549/DDP and A549 cells were exposed to serum-free culture medium or ionizing radiation. To measure the stress-induced autophagy, LC3-II, as an autophagosome marker, was measured by immunofluorescence and Western blotting. To determine the effect of 3-MA, a known inhibitor of autophagy, on overcoming acquired cisplatin resistance, the MTT assay and flow cytometry were performed. Western blotting analysis demonstrated that LC3-II was increased in A549/DDP cells, compared with those of parental A549 cells, under stress conditions. Meanwhile, immunofluorescence staining showed that LC3-II protein was located mainly in the cytoplasm of A549/DDP. We also found that 3-MA can enhance the growth inhibition and apoptotic effect of cisplatin in acquired resistant cells (A549/DDP). Collectively, our results provide evidence that the upregulation of autophagy plays a major role in cisplatin resistance of A549/DDP cells.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Autophagy/physiology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cisplatin/pharmacology , Adenocarcinoma/metabolism , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...