Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Int J Surg ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38704642

ABSTRACT

OBJECTIVES: The absence of non-invasive biomarkers for the early diagnosis of colorectal cancer (CRC) has contributed to poor prognosis. Extracellular vesicles (EVs) have emerged as promising candidates for cancer monitoring using liquid biopsy. However, the complexity of EVs isolation procedures and absence of clear targets for detecting serum-derived EVs have hindered the clinical application of EVs in early CRC diagnosis. METHODS: In the discovery phase, we conducted a comprehensive 4D-DIA proteomic analysis of serum-derived EVs samples from 37 individuals, performing an initial screening of EVs surface proteins. In the technical validation phase, we developed an extraction-free CRC-EVArray microarray to assess the expression of these potential EVs surface proteins in a multicenter study comprising 404 individuals. In the application phase, we evaluated the diagnostic efficacy of the CRC-EVArray model based on machine-learning algorithms. RESULTS: Through 4D-DIA proteomic analysis, we identified 7 potential EVs surface proteins showing significantly differential expression in CRC patients compared to healthy controls. Utilizing our developed high-throughput CRC-EVArray microarray, we further confirmed the differential expression of 3 EVs surface proteins, FIBG, PDGF-ß and TGF-ß, in a large sample population. Moreover, we established an optimal CRC-EVArray model using the NNET algorithm, demonstrating superior diagnostic efficacy with an AUC of 0.882 in the train set and 0.937 in the test set. Additionally, we predicted the functions and potential origins of these EVs-derived proteins through a series of multi-omics approaches. CONCLUSIONS: Our systematic exploration of surface protein expression profiles on serum-derived EVs has identified FIBG, PDGF-ß, and TGF-ß as novel diagnostic biomarkers for CRC. And the development of CRC-EVArray diagnostic model based on these findings provided an effective tool for the large-scale CRC screening, thus facilitating its translation into clinical practice.

2.
Mol Med ; 28(1): 151, 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36503493

ABSTRACT

BACKGROUND: Endometriosis (EMS) is a "tumour-like" gynaecological disease with distant metastasis, and studies have shown that EMS can induce distant metastasis through vascular vessels, but the driving factors and their mechanism are not clear. METHODS: We used an EMS animal model and gene knockout technique to explore the role of EMS-induced angiogenesis in EMS metastasis in vivo and in vitro and clarify the role and molecular mechanism of oxLDL in promoting EMS-induced angiogenesis. RESULTS: We found that microvascular density (MVD) in metastasized ectopic endometrium and eutopic endometrial tissue was higher than that in normal endometrial tissue, and plasma oxLDL was positively correlated with the distant metastasis of EMS. Furthermore, we clarified that oxLDL enhanced the MVD of endometrial tissue by increasing VEGF-A expression and secretion in endometrial cells. Finally, we illustrated the mechanism by which oxLDL promotes VEGF-A expression through the AKT-HIF-1α signalling pathway. CONCLUSION: OxLDL is a risk factor promoting distant EMS metastasis by increasing VEGF-A expression and secretion through AKT-HIF-1α signalling. This finding may provide theoretical support and therapeutic targets for the clinical prevention and treatment of EMS.


Subject(s)
Endometriosis , Vascular Endothelial Growth Factor A , Humans , Female , Animals , Vascular Endothelial Growth Factor A/metabolism , Endometrium/metabolism , Lipoproteins, LDL/metabolism
3.
Development ; 148(11)2021 06 01.
Article in English | MEDLINE | ID: mdl-34125190

ABSTRACT

Loss-of-function mutations in multiple morphological abnormalities of the sperm flagella (MMAF)-associated genes lead to decreased sperm motility and impaired male fertility. As an MMAF gene, the function of fibrous sheath-interacting protein 2 (FSIP2) remains largely unknown. In this work, we identified a homozygous truncating mutation of FSIP2 in an infertile patient. Accordingly, we constructed a knock-in (KI) mouse model with this mutation. In parallel, we established an Fsip2 overexpression (OE) mouse model. Remarkably, KI mice presented with the typical MMAF phenotype, whereas OE mice showed no gross anomaly except for sperm tails with increased length. Single-cell RNA sequencing of the testes uncovered altered expression of genes related to sperm flagellum, acrosomal vesicle and spermatid development. We confirmed the expression of Fsip2 at the acrosome and the physical interaction of this gene with Acrv1, an acrosomal marker. Proteomic analysis of the testes revealed changes in proteins sited at the fibrous sheath, mitochondrial sheath and acrosomal vesicle. We also pinpointed the crucial motifs of Fsip2 that are evolutionarily conserved in species with internal fertilization. Thus, this work reveals the dosage-dependent roles of Fsip2 in sperm tail and acrosome formation.


Subject(s)
Acrosome/metabolism , Carrier Proteins/metabolism , Seminal Plasma Proteins/metabolism , Sperm Tail/metabolism , Animals , Fertilization , Homozygote , Male , Membrane Proteins , Mice , Mutation , Phenotype , Proteomics , Sequence Analysis, RNA , Sperm Motility , Spermatogenesis , Testis
4.
Cell Death Dis ; 12(4): 295, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33731707

ABSTRACT

Nasopharyngeal carcinoma (NPC) is one of the most malignant tumors in southern China and Asia, and lymph node metastasis is an important cause for treatment failure. Lymphangiogenesis is a crucial step in lymphatic metastasis of NPC, while little is known about lymphangiogenesis in NPC. Similar to angiogenesis, lymphangitic neovascularization is a process of balance between pro-lymphangiogenesis and anti-lymphangiogenesis factors, but there are few studies on endogenous lymphangiogenesis inhibitors. Pigment epithelium-derived factor (PEDF) is a well-known effective endogenous angiogenesis inhibitor. However, the relationship between PEDF and lymphangiogenesis remains unknown. Our present study reveals that PEDF is lowly expressed in human NPC tissues with poor prognosis and is negatively correlated with lymphatic vessel density (LVD). Consistently, PEDF inhibits lymphangiogenesis and lymphatic metastasis of NPC in vivo experiments. Mechanistically, PEDF inhibits the proliferation, migration, and tube formation of lymphatic endothelial cells and promotes cell apoptosis. On the other hand, PEDF reduces the expression and secretion of vascular endothelial growth factor C (VEGF-C) of NPC cells through the nuclear factor-κB (NF-κB) signaling pathway. Our findings indicate that PEDF plays a vital role in lymphatic metastasis by targeting both lymphatic endothelial cells and NPC cells, and PEDF may represent a novel therapeutic target for NPC.


Subject(s)
Eye Proteins/therapeutic use , Lymphatic Metastasis/drug therapy , Nasopharyngeal Carcinoma/drug therapy , Nerve Growth Factors/therapeutic use , Protease Inhibitors/therapeutic use , Serpins/therapeutic use , Animals , Eye Proteins/pharmacology , Humans , Mice , Nerve Growth Factors/pharmacology , Protease Inhibitors/pharmacology , Serpins/pharmacology , Transfection
5.
J Cell Mol Med ; 25(8): 3963-3975, 2021 04.
Article in English | MEDLINE | ID: mdl-33621408

ABSTRACT

Abnormal lipid metabolism is the sign of tumour cells. Previous researches have revealed that the lipolytic pathway may contribute to the progression of colorectal cancer (CRC). However, adipose triglyceride lipase (ATGL) role in CRC cells remains unclear. Here, we find that elevated ATGL positively correlates with CRC clinical stages and negatively associates with overall survival. Overexpression of ATGL significantly promotes CRC cell proliferation, while knockdown of ATGL inhibits the proliferation and promotes the apoptosis of CRC cells in vitro. Moreover, in vivo experiments, ATGL promotes the growth of CRC cells. Mechanistically, ATGL enhances the carcinogenic function of CRC cells via promoting sphingolipid metabolism and CoA biosynthesis pathway-related gene levels by degrading triglycerides, which provides adequate nutrition for the progression of CRC. Our researches clarify for the first time that ATGL is a novel oncogene in CRC and may provide an important prognostic factor and therapeutic target for CRC.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Lipase/metabolism , Lipolysis , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Female , Humans , Lipase/genetics , Lipid Metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Life Sci ; 269: 119098, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33476628

ABSTRACT

AIMS: Far upstream element-binding protein 1 (FUBP1) has been shown to involve in the tumorigenesis and tumor progression of various cancers. However, the expression and function of FUBP1 in cervical carcinoma remains unknown. MAIN METHODS: Transcriptional expression of FUBP1 was initially evaluated using the Oncomine database, followed by evaluation of FUBP1 protein levels using immunohistochemistry in 119 cervical carcinoma patient tissues. In vitro experiments were performed to assess the tumorigenic role of FUBP1. Besides, Gene Set Enrichment Analysis, EnrichmentMap analysis, and protein-protein interaction (PPI) networks were used to evaluate the potential mechanisms of FUBP1 in promoting cervical cancer progression. KEY FUNDINGS: In this research, we found both FUBP1 mRNA transcription and protein expression levels increased significantly in cervical carcinoma tissues compared with adjacent normal cervical tissues. Furthermore, elevated FUBP1 expression was positively correlated with age, T classification, N classification, tumor recurrence, Ki67 expression, and poor prognosis in cervical carcinoma patients. Besides, elevated FUBP1 expression acted as an independent unfavorable predictor for overall survival and disease-free survival in cervical carcinoma. Overexpression of FUBP1 significantly promoted cervical carcinoma cell proliferation and inhibits cell apoptosis in vitro, while knockdown of FUBP1 showed the opposite effect. Mechanistically, bioinformatics analysis revealed that FUBP1 promoted the biological function of cervical carcinoma cells via enhancing DNA repair signal pathways. Our results demonstrate for the first time that FUBP1 is a novel prognostic factor and therapeutic target for cervical carcinoma.


Subject(s)
Biomarkers, Tumor/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , RNA-Binding Proteins/metabolism , Uterine Cervical Neoplasms/pathology , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , DNA-Binding Proteins/genetics , Female , Humans , Middle Aged , Prognosis , RNA-Binding Proteins/genetics , Survival Rate , Tumor Cells, Cultured , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
7.
Int J Gynaecol Obstet ; 153(3): 476-482, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33119129

ABSTRACT

OBJECTIVE: To evaluate the predictive value of the prewash and postwash total progressively motile sperm counts (TPMSC) on clinical pregnancy after intrauterine insemination (IUI) . METHODS: A review of medical records of women undergoing 2064 IUI cycles in a university hospital between April 2010 and October 2018. RESULTS: No pregnancies were seen when the prewash TPMSC was less than 9 million or the postwash TPMSC was less than 2 million. There was no correlation between TPMSC and clinical pregnancies. Further analysis was made in pregnancies after prewash TPMSC of 3-10 million and more than 10 million, and after postwash TPMSC of 10-100 million and more than 100 million. No correlation was found between TPMSC and clinical pregnancy in different strata of female age, infertility duration, type of infertility, type of IUI treatment, and the number of treatment cycles. A negative relationship was found for unstimulated IUI treatment and the number of treatment cycles. The prewash and postwash TPMSC showed no predictive value. CONCLUSION: Prewash and postwash TPMSC have poor predictive value for clinical pregnancy in IUI. Cycles with a prewash TPMSC of 10-100 million or postwash TPMSC of 3-10 million could achieve acceptable pregnancy rates with IUI.


Subject(s)
Insemination, Artificial , Pregnancy Rate , Sperm Count , Sperm Motility , Adult , Female , Humans , Male , Pregnancy , Retrospective Studies , Specimen Handling
8.
Am J Cancer Res ; 10(11): 3666-3685, 2020.
Article in English | MEDLINE | ID: mdl-33294260

ABSTRACT

Colorectal cancer (CRC) is one of the top three most deadly cancers despite using chemotherapy based on oxaliplatin or irinotecan combined with targeted therapy. Chiauranib has recently been identified to be a promising anticancer candidate with impressive efficacy and safety. However, the role and molecular mechanisms of Chiauranib in the treatment of CRC remain to be elucidated. Our study shows that Chiauranib inhibits cell proliferation and induces apoptosis in KRAS wild-type CRC cells in a dose- and time-dependent manner, but not mutation ones. Meanwhile, Chiauranib increases ROS production in KRAS wild-type CRC cells. Moreover, Chiauranib selectively suppresses KRAS wild-type CRC cells growth in vivo. Mechanistically, Chiauranib inhibits KRAS wild-type CRC cells by triggering ROS production via activating the p53 signaling pathway. Further, KRAS mutation CRC cells are resistant to Chiauranib by increasing Nrf2 to stably elevate the basal antioxidant program and thereby lower intracellular ROS induced by Chiauranib. Our findings provide the rationale for further clinical evaluation of Chiauranib as a therapeutic agent in treating KRAS wild-type CRC.

9.
Dev Biol ; 454(2): 118-127, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31255637

ABSTRACT

Sperm fibrous sheath, a unique cytoskeletal structure, is implicated in various sperm physiological functions, such as sperm maturation, motility and capacitation. AKAP4 has been described to be required for structural and functional integrity of the fibrous sheath. We generated Akap4-knockout mice line using CRISPR-Cas9 system. Cytomorphology and motility of sperm and testes were studied, confirming loss of Akap4 led to abnormal sperm morphology, motility and infertility. The proteomic components of testes were studied and Akap4 was found to be significantly decreased in the Akap4-knockout mice. Testis single-cell RNA sequencing and analysis revealed three genes with significant change in the general cell population, i.e., Akap4, Haspin, and Ccdc38. The single-cell RNA expression profiles also showed that the major difference between Akap4-knockout and wild-type testes existed in the elongating cell cluster, where in the Akap4-knockout testes, a subgroup of elongating cells with marker genes involved in cell adhesion and migration were increased, while a subgroup of elongating cells marked by mitochondrial sheath genes were decreased. Our results revealed the complex and well-coordinated procedures of spermatogenesis, and substantiated Akap4's indispensable roles in the integrity of sperm flagellum and the step-wise maturation of spermatozoa.


Subject(s)
A Kinase Anchor Proteins/metabolism , Spermatogenesis/genetics , A Kinase Anchor Proteins/genetics , Animals , Female , Infertility, Male/genetics , Infertility, Male/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteomics/methods , RNA/metabolism , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Sperm Motility/genetics , Sperm Motility/physiology , Sperm Tail/metabolism , Spermatogenesis/physiology , Spermatozoa/metabolism , Testis/metabolism
10.
Int J Oncol ; 54(2): 572-584, 2019 02.
Article in English | MEDLINE | ID: mdl-30483757

ABSTRACT

Gastric cancer is one of the most malignant tumor types, and its metastasis is a notable cause of mortality. Among the methods of tumor metastasis, lymphatic metastasis is the predominant one in gastric cancer. A previous study reported that the plasma oxidized low­density lipoprotein (oxLDL) is the risk factor associated with the development of tumors in patients with abnormal lipid metabolism, but the influence of plasma oxLDL in the lymphatic metastasis of gastric cancer remains unclear. In the present study, the concentration of plasma oxLDL from patients with gastric cancer was detected with an ELISA kit, and the lymphatic vessel density in gastric cancer tissues was determined by D2­40 staining. The correlation analysis of oxLDL concentration and lymphatic vessel density demonstrated that plasma oxLDL was positively correlated with lymphatic metastasis in patients with gastric cancer. Subsequently, the popliteal lymph node metastasis animal experiment with nude mice confirmed that oxLDL could promote the lymphatic metastasis of gastric cancer. Following this, the western blotting and ELISA data demonstrated that oxLDL promoted the expression and secretion of vascular endothelia growth factor (VEGF)­C in gastric cancer cell lines. Finally, blocking the lectin­like oxLDL­1 (LOX­1) receptor, a specific receptor for oxLDL, and the nuclear factor (NF)­κB signaling pathway following oxLDL (50 µg/ml) treatment in HGC­27 cells revealed that oxLDL could activate the NF­κB signaling pathway mediated by LOX­1, with subsequent upregulation of VEGF­C expression, and secretion in and from gastric cancer cells, and finally that it could promote the lymphatic metastasis of gastric cancer. These data indicate the association between the plasma oxLDL and the lymphatic metastasis of gastric cancer, and indicate that oxLDL elimination may be a potential therapeutic target for the prevention and intervention of early lymph node metastasis in gastric cancer.


Subject(s)
Lipoproteins, LDL/genetics , Lymphangiogenesis/genetics , Scavenger Receptors, Class E/genetics , Stomach Neoplasms/genetics , Vascular Endothelial Growth Factor C/genetics , Aged , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Lipoproteins, LDL/blood , Lymphatic Metastasis , Male , Mice , Middle Aged , NF-kappa B/genetics , Risk Factors , Signal Transduction/genetics , Stomach Neoplasms/blood , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcriptional Activation/genetics , Vascular Endothelial Growth Factor C/blood , Xenograft Model Antitumor Assays
11.
Gastric Cancer ; 21(4): 617-631, 2018 07.
Article in English | MEDLINE | ID: mdl-29243194

ABSTRACT

BACKGROUND: Tumor-induced lymphangiogenesis and lymphatic metastasis are predominant during the metastasis of many types of cancers. However, the endogenous inhibitors that counterbalance the lymphangiogenesis and lymphatic metastasis of tumors have not been well evaluated. Kallistatin has been recognized as an endogenous angiogenesis inhibitor. METHODS AND RESULTS: Our recent study showed for the first time that the lymphatic vessel density (LVD) was reduced in lung and stomach sections from kallistatin-overexpressing transgenic mice. Kallistatin expresses anti-lymphangiogenic activity by inhibiting the proliferation, migration, and tube formation of human lymphatic endothelial cells (hLECs). Therefore, the present study focuses on the relationships of changes in kallistatin expression with the lymphangiogenesis and lymphatic metastasis of gastric cancer and its underlying mechanisms. Our results revealed that the expression of kallistatin in cancer tissues, metastatic lymph nodes, and plasma of gastric cancer patients was significantly downregulated and that the plasma level of kallistatin was negatively associated with the phase of lymph node metastasis. Furthermore, treatment with kallistatin recombinant protein decreased LVD and lymph node metastases in the implanted gastric xenograft tumors of nude mice. Mechanically, kallistatin suppressed the lymphangiogenesis and lymphatic metastasis by downregulating VEGF-C expression and secretion through the LRP6/IKK/IÒ¡B/NF-Ò¡B signaling pathway in gastric cancer cells. CONCLUSIONS: These findings demonstrated that kallistatin functions as an endogenous lymphangiogenesis inhibitor and has an important part in the lymphatic metastasis of gastric cancer.


Subject(s)
Lymphangiogenesis/physiology , Serpins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Vascular Endothelial Growth Factor C/metabolism , Aged , Animals , Cell Line, Tumor , Down-Regulation , Female , Humans , Low Density Lipoprotein Receptor-Related Protein-6/genetics , Lymphatic Metastasis/pathology , Male , Mice, Inbred BALB C , Middle Aged , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Serpins/blood , Serpins/genetics , Serpins/pharmacology , Stomach Neoplasms/drug therapy
12.
Int J Oncol ; 50(6): 2000-2010, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28440474

ABSTRACT

Kallistatin has been recognized as an endogenous angiogenic inhibitor. However, its effects on lymphatic endothelial cells and lymphangiogenesis remain poorly understood. Lymphangiogenesis is involved in tumor metastasis via the lymphatic vasculature in various types of tumors. The aim of this study was to investigate the effects of kallistatin on lymphangiogenesis and the mechanism of action involved. Treatment with kallistatin recombinant protein or overexpression of kallistatin inhibited the proliferation, migration and tube formation of human lymphatic endothelial cells (hLECs), and induced apoptosis of hLECs. Furthermore, our results showed that the lymphatic vessel density (LVD) was reduced in lung and stomach sections from kallistatin-overexpressing transgenic mice. Treatment with kallistatin recombinant protein decreased the LVD in the implanted gastric xenograft tumors of nude mice. To the best of our knowledge, the present study is the first to demonstrate that kallistatin possesses anti-lymphangiogenic activity in vitro and in vivo. Moreover, kallistatin inhibited proliferation and migration of hLECs by reducing the phosphorylation of ERK and Akt, respectively. These findings suggested that kallistatin may be a promising agent that could be used to suppress cancer metastasis by inhibiting both angiogenesis and lymphangiogenesis.


Subject(s)
Lymphangiogenesis/genetics , Neovascularization, Pathologic/genetics , Recombinant Proteins/genetics , Serpins/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Lymphatic Metastasis/genetics , Mice , Mice, Transgenic , Neovascularization, Pathologic/pathology , Recombinant Proteins/administration & dosage , Recombinant Proteins/drug effects , Serpins/administration & dosage , Xenograft Model Antitumor Assays
13.
Oncotarget ; 8(3): 4582-4594, 2017 Jan 17.
Article in English | MEDLINE | ID: mdl-27999189

ABSTRACT

Müller cells are retinal glial cells and exhibit a fibroblast-like phenotype and ability to migrate in diabetic retinopathy (DR). However, expression of mesenchymal markers, which promote fibrosis in various organs, has not been characterized in the diabetic retina. We examined changes in the expression of these markers in Müller cells exposed to high glucose and in animal models of diabetic retinopathy. High glucose conditions increased mesenchymal maker expression and migration in Müller cells. Snail, N-cadherin, Vimentin, ß-catenin, and α-smooth muscle actin (α-SMA) levels were all dramatically increased in retinas from humans with diabetic retinopathy (DR) and from DR mouse models. In addition, Snail overexpression increased the expression of connective tissue growth factor (CTGF) and fibronectin, while Snail knockdown attenuated high glucose-induced increases in fibronectin and CTGF expression. These results demonstrate for the first time that mesenchymal markers are upregulated in retinas from a diabetic mouse model, and that Snail and N-cadherin levels are also increased in Müller cells exposed to high glucose. This suggests mesenchymal proteins may play a crucial role in the development of DR.


Subject(s)
Biomarkers/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/metabolism , Diabetic Retinopathy/metabolism , Ependymoglial Cells/cytology , Glucose/pharmacology , Actins/metabolism , Animals , Cadherins/metabolism , Cell Line , Cell Movement , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Humans , Mice , Snail Family Transcription Factors/metabolism , Streptozocin , Up-Regulation , Vimentin/metabolism , beta Catenin/metabolism
14.
Int J Mol Med ; 38(6): 1815-1822, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27748912

ABSTRACT

It has been reported that epithelial-mesenchymal transition (EMT) mediates multiple physiological and pathological processes. However, the occurrence and the pathogenic role of high glucose-induced EMT in retinal pigment epithelial cells (RPE cells) is unknown. The aim of this study was to examine the effects of high glucose on EMT in RPE cells. Cultured RPE cells were exposed to 25 mM D-glucose. A vector encoding the Snail gene and siRNA targeting Snail (Snail siRNA) were transfected into the cells to induce the overexpression or silencing of Snail, respectively. AKT and extracellular signal-regulated kinase (ERK) inhibitors were used to block the activation of AKT and ERK, respectively. The levels of EMT markers, fibrogenic factors, phosphorylated ERK and phosphorylated AKT were determined by western blot analysis and immunofluorescence staining. Cell migration was evaluated by wound healing assay. Our results revealed that high glucose elevated the expression of the key EMT transcriptional factor, Snail, and that of other mesenchymal makers, and promoted cell migration. Moreover, the overexpression of Snail elevated the levels of fibronectin and connective tissue growth factor (CTGF), whereas the silencing of Snail decreased the expression of fibronectin and CTGF induced by high glucose in the cells. Mechanistically, the AKT inhibitor (AKT inhibitor IV) and ERK inhibitor (U0126) significantly decreased the expression of Snail, as well as the levels of fibronectin and CTGF which were induced by high glucose. On the whole, and to the best of our knowedge, the present study is the first to demonstrate the upregulation of mesenchymal markers in RPE cells induced by high glucose, and suggest that mesenchymal transition may be involved in the pathological processes of retinal diseases.


Subject(s)
Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition , Glucose/metabolism , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Biomarkers , Cell Line , Cell Movement/drug effects , Cytokines/genetics , Cytokines/metabolism , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibrosis , Gene Expression Regulation/drug effects , Glucose/pharmacology , Humans , Proto-Oncogene Proteins c-akt/metabolism , Retinal Pigment Epithelium/drug effects , Signal Transduction/drug effects , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
15.
World J Gastroenterol ; 22(17): 4345-53, 2016 May 07.
Article in English | MEDLINE | ID: mdl-27158203

ABSTRACT

AIM: To investigate the anti-apoptotic capability of the hepatitis B virus (HBV) in the HepG2 hepatoma cell line and the underlying mechanisms. METHODS: Cell viability and apoptosis were measured by MTT assay and flow cytometry, respectively. Targeted knockdown of manganese superoxide dismutase (MnSOD), AMP-activated protein kinase (AMPK) and hepatitis B virus X protein (HBx) genes as well as AMPK agonist AICAR and antagonist compound C were employed to determine the correlations of expression of these genes. RESULTS: HBV markedly protected the hepatoma cells from growth suppression and cell death in the condition of serum deprivation. A decrease of superoxide anion production accompanied with an increase of MnSOD expression and activity was found in HepG2.215 cells. Moreover, AMPK activation contributed to the up-regulation of MnSOD. HBx protein was identified to induce the expression of AMPK and MnSOD. CONCLUSION: Our results suggest that HBV suppresses mitochondrial superoxide level and exerts an anti-apoptotic effect by activating AMPK/MnSOD signaling pathway, which may provide a novel pharmacological strategy to prevent HCC.


Subject(s)
AMP-Activated Protein Kinases/physiology , Apoptosis , Carcinoma, Hepatocellular/etiology , Hepatitis B virus/pathogenicity , Liver Neoplasms/etiology , Signal Transduction/physiology , Superoxide Dismutase/physiology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , Hep G2 Cells , Humans , Liver Neoplasms/pathology , Liver Neoplasms/virology , Reactive Oxygen Species/metabolism
16.
J Biol Chem ; 289(47): 32628-38, 2014 Nov 21.
Article in English | MEDLINE | ID: mdl-25296756

ABSTRACT

Human plasminogen kringle 5 (K5) is known to display its potent anti-angiogenesis effect through inducing endothelial cell (EC) apoptosis, and the voltage-dependent anion channel 1 (VDAC1) has been identified as a receptor of K5. However, the exact role and underlying mechanisms of VDAC1 in K5-induced EC apoptosis remain elusive. In the current study, we showed that K5 increased the protein level of VDAC1, which initiated the mitochondrial apoptosis pathway of ECs. Our findings also showed that K5 inhibited the ubiquitin-dependent degradation of VDAC1 by promoting the phosphorylation of VDAC1, possibly at Ser-12 and Thr-107. The phosphorylated VDAC1 was attenuated by the AKT agonist, glycogen synthase kinase (GSK) 3ß inhibitor, and siRNA, suggesting that K5 increased VDAC1 phosphorylation via the AKT-GSK3ß pathway. Furthermore, K5 promoted cell surface translocation of VDAC1, and binding between K5 and VDAC1 was observed on the plasma membrane. HKI protein blocked the impact of K5 on the AKT-GSK3ß pathway by competitively inhibiting the interaction of K5 and cell surface VDAC1. Moreover, K5-induced EC apoptosis was suppressed by VDAC1 antibody. These data show for the first time that K5-induced EC apoptosis is mediated by the positive feedback loop of "VDAC1-AKT-GSK3ß-VDAC1," which may provide new perspectives on the mechanisms of K5-induced apoptosis.


Subject(s)
Apoptosis/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Peptide Fragments/pharmacology , Plasminogen/pharmacology , Voltage-Dependent Anion Channel 1/metabolism , Apoptosis/genetics , Blotting, Western , Caspases/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Feedback, Physiological/drug effects , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , HEK293 Cells , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Peptide Fragments/genetics , Phosphorylation/drug effects , Plasminogen/genetics , Protein Binding , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics , Ubiquitin/metabolism , Voltage-Dependent Anion Channel 1/genetics
17.
J Biol Chem ; 289(44): 30785-30799, 2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25225287

ABSTRACT

Pigment epithelium-derived factor (PEDF), a potent antiangiogenesis agent, has recently attracted attention for targeting tumor cells in several types of tumors. However, less is known about the apoptosis-inducing effect of PEDF on human lung cancer cells and the underlying molecular events. Here we report that PEDF has a growth-suppressive and proapoptotic effect on lung cancer xenografts. Accordingly, in vitro, PEDF apparently induced apoptosis in A549 and Calu-3 cells, predominantly via the Fas-L/Fas death signaling pathway. Interestingly, A549 and Calu-3 cells are insensitive to the Fas-L/Fas apoptosis pathway because of the low level of cell surface Fas. Our results revealed that, in addition to the enhancement of Fas-L expression, PEDF increased the sensitivity of A549 and Calu-3 cells to Fas-L-mediated apoptosis by triggering the translocation of Fas protein to the plasma membrane in a p53- and FAP-1-dependent manner. Similarly, the up-regulation of Fas-L by PEDF was also mediated by p53. Furthermore, peroxisome proliferator-activated receptor γ was determined to be the upstream regulator of p53. Together, these findings uncover a novel mechanism of tumor cell apoptosis induced by PEDF and provide a potential therapeutic strategy for tumors that are insensitive to Fas-L/Fas-dependent apoptosis because of a low level of cell surface Fas.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Eye Proteins/pharmacology , Fas Ligand Protein/genetics , Nerve Growth Factors/pharmacology , Serpins/pharmacology , Tumor Suppressor Protein p53/physiology , fas Receptor/metabolism , Animals , Antineoplastic Agents/therapeutic use , Caspase 8/metabolism , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Eye Proteins/physiology , Eye Proteins/therapeutic use , Fas Ligand Protein/metabolism , Humans , Lung Neoplasms , Male , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/prevention & control , Nerve Growth Factors/physiology , Nerve Growth Factors/therapeutic use , PPAR gamma/metabolism , Protein Transport , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , Serpins/physiology , Serpins/therapeutic use , Up-Regulation , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...