Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Medicine (Baltimore) ; 99(22): e20305, 2020 May 29.
Article in English | MEDLINE | ID: mdl-32481400

ABSTRACT

BACKGROUND: TNF-like cytokine 1A (TL1A) is a subgroup of the tumor necrosis factor superfamily that exerts pleiotropic effects on cell proliferation, inflammation, activation, and differentiation of immune cells. The purpose of the current study is to investigate the clinical significance of TL1A expression in coronary and peripheral blood of patients with acute coronary syndrome (ACS) to determine if TL1A levels can serve as an accurate prognostic indicator. METHODS: A total of 141 patients undergoing coronary angiography were divided into 4 groups: Control (n = 35), Unstable Angina (UA) (n = 35), acute non-ST segment elevation myocardial infarction (NSTEMI) (n = 37), and acute ST segment elevation myocardial infarction (STEMI) (n = 34). The levels of TL1A, MPO, hs-CRP, and IL-10 were detected in coronary and peripheral blood using enzyme linked immunosorbent assay (ELISA), and the MACE incidence rates were compared during 26.3 months of follow-up. RESULTS: TL1A levels were not significantly different between the UA group and control group. In the UA group, TL1A levels were not significantly different between coronary blood and peripheral blood. However, TL1A levels were higher in the STEMI and NSTEMI groups than in the control group (P < .05). Moreover, TL1A levels in the coronary blood of the STEMI and NSTEMI groups were higher than in the peripheral blood (P < .05). The expression of TL1A in the coronary blood was the highest in the STEMI group. In addition, TL1A level in the coronary blood was highly correlated with levels in the peripheral blood (correlation coefficient: 0.899, P < .001). The hs-CRP and MPO levels in the coronary and peripheral blood of all the UA, NSTEMI, and STEMI groups were higher than the control group. Plasma IL-10 levels in all the UA, NSTEMI and STEMI groups were lower than those in the control group. Plasma TL1A level was positively correlated with the cTnI level, degree of coronary thrombus burden, occurrence of slow coronary flow / no coronary reflow and MACE, but negatively correlated with the IL-10 level or non-correlated with the Syntax score. CONCLUSION: Plasma TL1A concentration levels can be used as a predictor of inflammatory response and prognosis in patients with ACS. TRIAL REGISTRATION: ClinicalTrials.gov, number: NCT02430025; Unique Protocol ID: FJPH20150101; Brief Title: Fujian Province Cardiovascular Diseases Study (FJCVD).


Subject(s)
Acute Coronary Syndrome/blood , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Aged , Angina, Unstable/blood , Biomarkers , Body Mass Index , Enzyme-Linked Immunosorbent Assay , Female , Humans , Lipids/blood , Male , Middle Aged , Myocardial Infarction/blood , Myocardial Infarction/classification
2.
J Neuroimmunol ; 335: 577020, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31445379

ABSTRACT

TL1A/DR3/DcR3 pathway is an important mediator of inflammatory responses and contributes to the pathogenesis of several chronic inflammatory diseases. Therefore, we analysed PBMC gene expression of these molecules in 30 relapsing-remitting multiple sclerosis (RRMS) patients, 8 secondary progressive MS (SPMS), 9 primary progressive MS (PPMS), 11 clinically isolated syndrome (CIS) patients, and 16 healthy controls (HCs), to evaluate their biomarker potential in MS. The results showed significant decrease in TL1A expression in RRMS compared to other study groups. TL1A as a marker of inflammation, we found its higher expression among treatment näive RRMS patients as compared to HCs and among patients who were treated with DMTs. Moreover, TL1A expression was found to be associated with the clinical and MRI findings of MS patients suggesting its possible involvement in the establishment or preservation of immune system homeostasis or in the regulation of inflammatory activity. Taken together, these findings suggest the TL1A should be evaluated further for its potential as a candidate biomarker of inflammatory activity and the marker of therapeutic response to immunomodulatory treatments in MS.


Subject(s)
Multiple Sclerosis/immunology , Receptors, Tumor Necrosis Factor, Member 25/biosynthesis , Receptors, Tumor Necrosis Factor, Member 6b/biosynthesis , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Adult , Biomarkers/analysis , Female , Follow-Up Studies , Humans , Male , Middle Aged , Receptors, Tumor Necrosis Factor, Member 25/analysis , Receptors, Tumor Necrosis Factor, Member 6b/analysis , Transcriptome , Tumor Necrosis Factor Ligand Superfamily Member 15/analysis
3.
Int J Oncol ; 55(1): 167-178, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31180533

ABSTRACT

Vascular endothelial growth inhibitor (VEGI; also referred to as TNFSF15 or TL1A) is involved in the modulation of vascular homeostasis. VEGI is known to operate via two receptors: Death receptor­3 (DR3) and decoy receptor­3 (DcR3). DR3, which is thus far the only known functional receptor for VEGI, contains a death domain and induces cell apoptosis. DcR3 is secreted as a soluble protein and antagonizes VEGI/DR3 interaction. Overexpression of DcR3 and downregulation of VEGI have been detected in a number of cancers. The aim of the present study was to investigate the effects of sodium valproate (VPA), a histone deacetylase inhibitor, in combination with hydralazine hydrochloride (Hy), a DNA methylation inhibitor, on the expression of VEGI and its related receptors in human osteosarcoma (OS) cell lines and human microvascular endothelial (HMVE) cells. Combination treatment with Hy and VPA synergistically induced the expression of VEGI and DR3 in both OS and HMVE cells, without inducing DcR3 secretion. In addition, it was observed that the combination of VPA and Hy significantly enhanced the inhibitory effect on vascular tube formation by VEGI/DR3 autocrine and paracrine pathways. Furthermore, the VEGI/VEGF­A immune complex was pulled down by immunoprecipitation. Taken together, these findings suggest that DNA methyltransferase and histone deacetylase inhibitors not only have the potential to induce the re­expression of tumor suppressor genes in cancer cells, but also exert anti­angiogenic effects, via enhancement of the VEGI/DR3 pathway and VEGI/VEGF­A interference.


Subject(s)
Bone Neoplasms/drug therapy , Hydralazine/pharmacology , Osteosarcoma/drug therapy , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Valproic Acid/pharmacology , Vascular Endothelial Growth Factor A/metabolism , Bone Neoplasms/blood supply , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Drug Synergism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Osteosarcoma/blood supply , Osteosarcoma/genetics , Osteosarcoma/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor, Member 25/biosynthesis , Receptors, Tumor Necrosis Factor, Member 25/genetics , Transcription, Genetic/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics
4.
Life Sci ; 231: 116536, 2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31176785

ABSTRACT

AIMS: TL1A was reported to contribute to the susceptibility to ulcerative colitis (UC). However, the molecular mechanisms of TL1A in UC development are poorly understood. We aimed to investigate the role of TL1A in colitis, and reveal the regulatory mechanism of TL1A in chronic colitis development. MAIN METHODS: Wild-type mice and transgenic mice with overexpressing TL1A in lymphocytes were used to construct chronic DSS colitis models. To investigate the molecular mechanism in vitro, CD4+ T cells were sorted from spleens and mesenteric lymph node cells to induce Th9 cells. Biopsy specimens from ulcerative colitis patients were collected for in vivo validation. KEY FINDINGS: The elevated TL1A expression in chronic DSS colitis models exacerbated intestinal inflammation. The differentiation of Th9 cells, IL-9 secretion and production of TGF-ß, IL-4 and PU.1 was significantly enhanced in transgenic mice with TL1A overexpression. In vitro results showed that TL1A enhanced the Th9 cells, IL-9 and PU.1 production, while TL1A antibodies inhibited their production. In human translational studies, patients with ulcerative colitis with elevated TL1A expression also exhibited more serious inflammation with higher levels of Th9 cells, IL-9 and PU.1 expression. SIGNIFICANCE: We presented a possible mechanism of TL1A in UC development that TL1A may promote the differentiation of Th9 cells and enhanced IL-9 secretion by up-regulating the expression of TGF-ß, IL-4 and PU.1, which provided a novel perspective to study the UC pathogenesis, and indicated that targeting of TL1A signal pathway may by a likely strategy for the treatment of chronic colitis.


Subject(s)
Colitis/immunology , Interleukin-9/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/immunology , Animals , Cell Differentiation/immunology , Colitis/chemically induced , Colitis/pathology , Cytokines/immunology , Cytokines/metabolism , Glutathione/metabolism , Interleukin-17/immunology , Interleukin-1beta/metabolism , Interleukin-9/metabolism , Intestinal Mucosa/immunology , Intestines/immunology , Intestines/pathology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction , T-Lymphocyte Subsets/immunology , Transforming Growth Factor beta/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Tumor Necrosis Factor-alpha/metabolism
5.
Anticancer Res ; 37(8): 4269-4275, 2017 08.
Article in English | MEDLINE | ID: mdl-28739718

ABSTRACT

BACKGROUND: Vascular endothelial growth inhibitor (VEGI) is a member of the tumor necrosis factor superfamily, identified as an anti-angiogenic cytokine. However, the effect of VEGI on epithelial-mesenchymal transition (EMT) in renal cell carcinoma (RCC) is still unknown. MATERIALS AND METHODS: In this study, protein VEGI174 was designed and synthesized. Renal cell carcinoma A498 cells were implanted into immune-deficient mice to establish tumor models. Two groups were included: control group treated with saline, and VEGI174-treated group. Data of tumor growth were collected every 3 to 4 days. Two weeks later, the tumor specimens were harvested for immunohistochemical staining of EMT markers (E-cadherin, N-cadherin, vimentin). RESULTS: Compared to the saline-treated group, the VEGI174-treated group showed significant inhibition of tumor growth (p<0.05). The expression of E-cadherin was significantly higher in the VEGI174-treated group compared to the saline-treated group (p<0.01). However, the expression of N-cadherin and vimentin were reduced in the VEGI174-treated group. CONCLUSION: Our findings indicate that VEGI174 prevents progression and tumor metastasis through inhibiting EMT in RCC in vivo. This may provide a new approach for the treatment of RCC.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic/drug effects , Growth Inhibitors/administration & dosage , Humans , Mice , Tumor Necrosis Factor Ligand Superfamily Member 15/antagonists & inhibitors , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Xenograft Model Antitumor Assays
6.
Cell Physiol Biochem ; 41(5): 2077-2090, 2017.
Article in English | MEDLINE | ID: mdl-28456797

ABSTRACT

BACKGROUND/AIMS: To investigate the associations between the rs1250569 (zinc finger MIZ-type containing 1, ZMIZ1), rs1042522 (tumour protein p53, TP53), and rs10114470 (tumour necrosis factor-like cytokine 1A, TL1A) polymorphisms and the development of inflammatory bowel disease (IBD) in a Chinese (Han) population. We analysed the expression of genes that predispose patients to Crohn's disease (CD) and ulcerative colitis (UC). METHODS: A total of 381 IBD patients and 517 healthy controls were recruited into our study. Polymorphisms at the three loci were genotyped using polymerase chain reaction-ligation detection reactions (PCR-LDR). Genotype-phenotype correlations were analysed. Blood and gut samples were obtained and analysed using quantitative real-time PCR (qRT-PCR), western blot analysis, and immunohistochemistry to investigate the mRNA and protein levels and in situ expression of genes found to predispose patients to IBD. Furthermore, the expression of susceptible genes was further verified using a mouse dextran sulphate sodium (DSS)-induced acute colitis model. RESULTS: No significant association was detected between rs1250569 and rs1042522 genotypes and CD or UC susceptibility. However, the frequency of allele A of rs1250569 was much higher in CD patients than that in healthy controls (55.03% vs. 48.48%, respectively; p = 0.044). The mutation rates at rs10114470 were dramatically lower at both the genotype and allele level in patients than those in healthy controls (p = 0.002 at both the genotype and allele level). Additionally, increased ZMIZ1 and TL1A levels were detected in intestinal samples obtained from both IBD patients and DSS-treated mice. CONCLUSION: rs1250569 (ZMIZ1) and rs10114470 (TL1A) are two novel loci that indicate susceptibility to IBD in Han-Chinese patients. Consistent with previous studies, TL1A expression levels were higher in Chinese Han IBD patients and DSS-treated mice. Most importantly, we found that ZMIZ1 expression was markedly higher in both IBD patients and mice with experimentally induced colitis, suggesting that ZMIZ1 plays important roles in the pathogenesis of IBD.


Subject(s)
Alleles , Colitis, Ulcerative/genetics , Gene Expression Regulation , Genetic Loci , Genetic Predisposition to Disease , Transcription Factors/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Asian People/ethnology , Asian People/genetics , China/ethnology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/ethnology , Colitis, Ulcerative/metabolism , Crohn Disease/ethnology , Crohn Disease/genetics , Crohn Disease/metabolism , Disease Models, Animal , Female , Humans , Intracellular Signaling Peptides and Proteins/biosynthesis , Intracellular Signaling Peptides and Proteins/genetics , Male , Mice , Middle Aged , RNA-Binding Proteins , Real-Time Polymerase Chain Reaction , Transcription Factors/biosynthesis , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis
7.
Genet Mol Res ; 14(2): 5462-7, 2015 May 22.
Article in English | MEDLINE | ID: mdl-26125742

ABSTRACT

We investigated the effect of atorvastatin on vascular endothelial growth inhibitor (VEGI) expression in rats with diabetic retinopathy. Wistar rats were divided into a blank group and diabetic model group, which was further randomly divided into treatment and control groups. Rats in the treatment group received 10 mg/kg atorvastatin daily, while rats in the blank and control groups received normal saline. Rats were randomly euthanized at 3 or 6 months. Immunohistochemical staining was used to determine changes in VEGI and vascular endothelial growth factor, interleukin-4, and tumor necrosis factor α levels in rats with diabetic retinopathy. Survival rate in the treatment group was 84% (63/75) after 6 months, which was significantly higher than that in the control group (P < 0.05); rats in the control group showed the lowest survival rate. Survival in the treatment group was higher than that in the control group but not significant compared with the blank group after 3 months. VEGI, vascular endothelial growth factor, tumor necrosis factor α, and interluekin-4 expression was lower than that in the control group, but higher than the blank group after 3 months. The expression of each factor decreased to the blank group level in the treatment group and was significantly lower than that in the control group after 6 months (P < 0.05). Expression in control and blank groups was similar at 3 and 6 months. Atorvastatin can inhibit VEGI and vascular endothelial growth factor expression to protect rats from diabetic retinopathy.


Subject(s)
Atorvastatin/administration & dosage , Diabetes Mellitus, Experimental/drug therapy , Diabetic Retinopathy/drug therapy , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetic Retinopathy/genetics , Diabetic Retinopathy/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Interleukin-4/biosynthesis , Rats , Rats, Wistar , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Vascular Endothelial Growth Factor A/biosynthesis
8.
Int J Oncol ; 42(5): 1664-73, 2013 May.
Article in English | MEDLINE | ID: mdl-23545578

ABSTRACT

Vascular endothelial growth inhibitor (VEGI) has been associated with tumor-related vasculature in certain malignancies. However, its implication in renal cell carcinoma (RCC), an angiogenesis-dependent tumor, remains unknown. In the present study, we investigated the role played by VEGI in RCC. The expression of VEGI was examined in human renal tissue and RCC cell lines using immunohistochemical staining and RT-PCR, respectively. The biological impact of modifying the expression of VEGI in RCC cells was evaluated using in vitro and in vivo models. We show that VEGI mRNA is expressed in a wide variety of human RCC cell lines, all of normal renal and most of RCC tissue specimens. VEGI protein expression was observed in normal renal tubular epithelial cells, but was decreased or absent in RCC specimens, particularly in tumors with high grade. Moreover, forced expression of VEGI led to an inhibition of vascular endothelial tube formation, decrease in the motility and adhesion of RCC cells in vitro. Interestingly, forced expression of VEGI had no bearing on growth, apoptosis and invasive capacity of RCC cells. However, tumor growth was reduced in xenograft models. Immunohistochemical staining showed that microvessel density decreased in VEGI forced expression xenograft tumor samples. Taken together, our findings showed that the expression of VEGI is decreased in RCC, particularly in tumors with higher grade. Together with its inhibitory effect on cellular motility, adhesion, vascular endothelial tube formation and tumor growth in vivo, this suggests that VEGI functions mainly through inhibition of angiogenesis and is a negative regulator of aggressiveness during the development and progression of RCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Endothelium, Vascular/pathology , Neovascularization, Pathologic/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Animals , Apoptosis/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Endothelium, Vascular/growth & development , Endothelium, Vascular/metabolism , Gene Expression Regulation, Neoplastic , Humans , Mice , Neoplasm Staging , Neovascularization, Pathologic/pathology , RNA, Messenger/biosynthesis , Transplantation, Heterologous , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Gut ; 62(7): 985-94, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22684480

ABSTRACT

OBJECTIVES: The postinfectious irritable bowel syndrome (PI-IBS) suggests that impaired resolution of inflammation could cause IBS symptoms. The authors hypothesised that polymorphisms in genes whose expression were altered by gastroenteritis might be linked to IBS with diarrhoea (IBS-D) which closely resembles PI-IBS. DESIGN: Part 1: 25 healthy volunteers (HVs), 21 patients 6 months after Campylobacter jejuni infection, 37 IBS-D and 19 IBS with constipation (IBS-C) underwent rectal biopsy for gene expression analysis and peripheral blood mononuclear cell cytokine production assessment. Part 2: Polymorphisms in genes whose expression was altered in Part 1 were assessed in 179 HV, 179 IBS-D, 122 IBS-C and 41 PI-IBS. RESULTS: Part 1: Mucosal expression of seven genes was altered in IBS: CCL11, CCL13, Calpain 8 and TNFSF15 increased while NR1D1, GPR161 and GABRE decreased with similar patterns after infection with C jejuni. Part 2: The authors assessed 21 known single nucleotide polymorphisms (SNPs) in these seven genes and one SNP in each of the TNFα and IL-10 genes. Three out of five TNFSF15 SNPs (rs6478108, rs6478109 and rs7848647) showed reduced minor allele frequency (MAF) (0.28, 0.27 and 0.27) in subjects with IBS-D compared with HV (0.38, 0.36 and 0.37; p=0.007, 0.015 and 0.007, respectively) confirming others recent findings. The authors also replicated the previously reported association of the TNFα SNP rs1800629 with PI-IBS which showed an increase in the MAF at 0.30 versus 0.19 for HV (p=0.04). CONCLUSION: IBS-D and PI-IBS patients are associated with TNFSF15 and TNFα genetic polymorphisms which also predispose to Crohn's disease suggesting possible common underlying pathogenesis.


Subject(s)
Irritable Bowel Syndrome/genetics , Polymorphism, Single Nucleotide , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Tumor Necrosis Factor-alpha/genetics , Adult , Cytokines/biosynthesis , Female , Gene Expression Profiling/methods , Gene Expression Regulation , Gene Frequency , Genetic Association Studies/methods , Genetic Predisposition to Disease , Genotype , Helicobacter Infections/complications , Helicobacter Infections/genetics , Helicobacter Infections/metabolism , Helicobacter pylori , Humans , Intestinal Absorption/physiology , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/microbiology , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Oligonucleotide Array Sequence Analysis/methods , Phenotype , Rectum/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis
10.
Gynecol Oncol ; 128(2): 391-6, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23142075

ABSTRACT

OBJECTIVE: The prognostic role of vascular endothelial growth factor (VEGF) in ovarian cancer remains inconclusive. This meta-analysis aimed to explore the association between VEGF overexpression and survival outcomes in ovarian cancer patients. METHODS: Studies were identified from PubMed and EMBASE searches performed on January 2nd, 2011. After careful review, survival data were extracted from eligible studies. A meta-analysis was performed to generate combined hazard ratio (HR) for progression-free survival (PFS) and overall survival (OS) in serum and tumor tissue studies. RESULTS: Sixteen studies with 1111 patients were analyzed. Elevated serum VEGF was significantly associated with poor PFS [HR 2.46, 95% CI (1.84, 3.29)] and OS [HR 2.21, 95% CI (1.57, 3.13)]. No significant heterogeneity existed in serum studies. Similarly, tissue VEGF overexpression was associated with poor PFS [HR 1.63, 95% CI (1.09, 2.42)] and OS [HR 1.70, 95% CI (1.01, 2.87)]. However, significant heterogeneity was found in tissue studies, with I(2) of 44% for PFS and 64% for OS. Studies were stratified into subgroups by International Federation of Gynecology and Obstetrics (FIGO) stages. Subgroup analyses showed that high tissue VEGF was significantly associated with shorter PFS [HR 5.34, 95% CI (1.95, 14.59)] and OS [HR 6.13, 95% CI (2.47, 15.26)] in studies where predominantly early-stage patients were included, but not in studies with a majority of advanced-stage patients. Subgroup analysis was not performed in serum studies because all those studies enrolled more patients in advanced stages than early stages. CONCLUSIONS: Overexpression of VEGF in primary tumor and serum associates with poor PFS and OS for patients with ovarian cancer. The association between high tissue VEGF level and poor prognosis exists in early stage patients, but not in advanced stage patients.


Subject(s)
Ovarian Neoplasms/blood , Ovarian Neoplasms/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Tumor Necrosis Factor Ligand Superfamily Member 15/blood , Disease-Free Survival , Female , Humans , Prognosis , Survival Rate
11.
Am J Pathol ; 180(4): 1454-64, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22330679

ABSTRACT

The expression of death receptor 3 (DR3), a member of the tumor necrosis factor (TNF) receptor superfamily, is up-regulated in human tubular epithelial cells (TECs) during renal injury, but its function in this setting remains unknown. We used cisplatin to induce renal injury in wild-type (DR3(+/+)) or congenitally deficient DR3(-/-) mice to examine the in vivo role of DR3. Cisplatin induced the expression of DR3, its ligand, TNF-like ligand 1A (TL1A), and TNF in TECs, as observed in human renal injury. Cisplatin increased apoptotic death of DR3(-/-) TECs by twofold compared with DR3(+/+) TECs, whereas it reduced the number of tubules expressing phospho-NF-κBp65(Ser276) by 50% at 72 hours. Similar degrees of induction of DR3, TL1A, and TNF, and changes in apoptosis and phospho-NF-κBp65(Ser276), were obtained in mouse kidney organ cultures treated with cisplatin for 3 hours, suggesting a direct effect on TECs. TNF was implicated in mediating cisplatin-induced tubular damage given that the in vivo co-administration of GM6001, an inhibitor of TNF maturation and release, significantly reduced TNF production and tubular damage. Moreover, TNF exacerbated, whereas TL1A reduced, cisplatin-induced apoptosis in the DR3(+/+) mouse proximal tubule cell line, TKPTS. Our data demonstrate that cisplatin-induced nephrotoxicity is mitigated by DR3 signaling, suggesting that this occurs by antagonizing pro-apoptotic signals induced by TNF. Therefore, activating DR3 may be beneficial in reducing acute kidney injury.


Subject(s)
Acute Kidney Injury/pathology , Receptors, Tumor Necrosis Factor, Member 25/physiology , Tumor Necrosis Factor-alpha/physiology , Acute Kidney Injury/chemically induced , Acute Kidney Injury/metabolism , Animals , Apoptosis/drug effects , Cisplatin/pharmacology , Dipeptides/pharmacology , Drug Interactions , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Kidney Tubules/pathology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/pathology , Ligands , Mice , Mice, Mutant Strains , NF-kappa B/metabolism , Organ Culture Techniques , Phosphorylation/drug effects , Protease Inhibitors/pharmacology , Receptors, Tumor Necrosis Factor, Member 25/biosynthesis , Receptors, Tumor Necrosis Factor, Member 25/deficiency , Signal Transduction/physiology , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation/drug effects
12.
J Immunol ; 186(7): 4175-82, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21368235

ABSTRACT

The nervous system influences immune responses through the release of neural factors such as neuropeptides. Among them, the tachykinin substance P (SP) signals via the neurokinin 1 receptor (NK-1R), which is expressed by various immune cells. We thereby analyzed in this paper whether tachykinins may participate in human CD4(+) Th cell polarization. We report that SP and hemokinin-1 (HK-1) upregulate IL-17A and IFN-γ production by human memory CD4(+) T cells without affecting IL-4 and IL-10 production. SP and HK-1 switch non-Th17-committed CD4(+) memory T cells into bona fide Th17 cells and Th1/Th17 cells. In contrast, SP and HK-1 do not modulate the polarization of naive CD4(+) T cells. SP- and HK-1-induced Th17 cell generation is mediated through NK-1R and requires the presence of monocytes. SP and HK-1 trigger IL-1ß, IL-6, and TNF-α production, upregulate IL-23 production, and enhance TNF-like 1A expression on monocyte surface. Neutralization experiments demonstrated that IL-1ß, IL-23, and TNF-like 1A are involved in the SP- and HK-1-induced Th17 cell. The other members of the tachykinin family, neurokinins A and B, have no effect on the differentiation of naive and memory T cells. These results thereby show that SP and HK-1 are novel Th17 cell-inducing factors that may act locally on memory T cells to amplify inflammatory responses.


Subject(s)
Cell Differentiation/immunology , Immunologic Memory , Interleukin-1beta/biosynthesis , Interleukin-23/biosynthesis , Monocytes/immunology , Substance P/physiology , Tachykinins/physiology , Th17 Cells/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Cell Communication/genetics , Cell Communication/immunology , Cell Differentiation/genetics , Cell Polarity/genetics , Cell Polarity/immunology , Cells, Cultured , Humans , Immunologic Memory/genetics , Inflammation Mediators/physiology , Interleukin-1beta/genetics , Interleukin-1beta/physiology , Interleukin-23/genetics , Interleukin-23/physiology , Monocytes/metabolism , Monocytes/pathology , Th17 Cells/metabolism , Th17 Cells/pathology , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Tumor Necrosis Factor Ligand Superfamily Member 15/physiology
13.
Anticancer Res ; 30(10): 3939-44, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21036706

ABSTRACT

Induction of angiogenic responses by multiple factors, a crucial step in tumor growth and metastasis, is not completely understood. Recently, involvement of the cytoskeletal actin-binding proteins in angiogenesis has been suggested as a target for anti-neovascular cancer therapy in vitro. In this study, the expression of filamin A (FLNA) and vascular endothelial growth factor (VEGF) in paraffin-embedded tumor samples from patients with well-characterized lung tumors was immunohistochemically analyzed and compared with clinical variables and survival outcome. A positive expression of FLNA and VEGF was detected in the cytoplasm of tumor cells in 66 (48.2%) and 69 (50.4%) of the 137 patients with lung cancer, respectively (p<0.0001). A significant difference was observed between FLNA expression and VEGF expression. Although our findings do not suggest that the expression of FLNA alone plays an independent prognostic role, the angiogenesis pathway mediated by FLNA appears to be responsible for controlling the growth of lung tumors.


Subject(s)
Contractile Proteins/biosynthesis , Lung Neoplasms/metabolism , Microfilament Proteins/biosynthesis , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Adult , Aged , Aged, 80 and over , Cohort Studies , Cytoplasm/metabolism , Female , Filamins , Humans , Immunohistochemistry , Lung Neoplasms/blood supply , Lung Neoplasms/pathology , Male , Middle Aged , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Paraffin Embedding , Retrospective Studies
14.
Anticancer Res ; 30(1): 87-95, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20150621

ABSTRACT

BACKGROUND: Vascular endothelial growth inhibitor (VEGI) has been implicated in the regulation of tumour-related vasculature in certain solid tumours. However, its role in urothelial tumours is still unclear. In the present study, the role played by VEGI in urothelial tumours of the bladder was investigated. MATERIALS AND METHODS: The expression of VEGI was examined in cancer human bladder tissues and in a serial of cell lines using immunochemical staining and RT-PCR respectively. The biological impact of modifying VEGI expression in urothelial cancer cells was evaluated using in vitro models. RESULTS: VEGI mRNA was expressed in a wide variety of human cell lines. VEGI expression was seen in normal urothelial and stromal cells, but was found to be reduced or absent in urothelial cancer cells. The positive staining in normal tissue (6/7) was significantly higher than that of urothelial cancer tissues (2/12), p=0.006. Moreover, overexpression of VEGI reduced the motility and adhesion of urothelial cancer cells in vitro. However, the overexpression of VEGI had no bearing on the growth and invasion of urothelial cancer cells. CONCLUSION: VEGI has an inhibitory effect on cellular motility and adhesion in bladder cancer cells. Taken together with the expression pattern of VEGI in urothelial cancer of the bladder, it suggests that VEGI functions as a negative regulator of aggressiveness during development and progression of bladder cancer.


Subject(s)
Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Urinary Bladder Neoplasms/metabolism , Urinary Bladder Neoplasms/pathology , Cell Adhesion/physiology , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Movement/physiology , Humans , Immunohistochemistry , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Urinary Bladder Neoplasms/genetics , Urothelium/pathology
15.
Protein Sci ; 19(3): 449-57, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20052682

ABSTRACT

Methods to prepare pure, bioactive recombinant human vascular endothelial growth inhibitor (rhVEGI), a potent inhibitor of angiogenesis potentially applicable in antiangiogenic cancer therapy, are in urgent demand for preclinical investigation as well as future clinical trials of the protein. Here, we report expression and purification of rhVEGI-192, a recombinant VEGI isoform, comparatively using host strains BL21 (DE3) pLysS and Origami B (DE3) with IPTG-induction and autoinduction techniques. Our study identified that a combined use of Origami B (DE3) strain and autoinduction expression system gave rise to a high yield of purified rhVEGI-192 at 105.38 mg/L culture by immobilized-metal affinity chromatography on Ni-NTA column. The antiangiogenic activity was effectively restored after the insoluble fractions being dissolved in 8M urea and subsequently subjected to a gradient-dialysis refolding process. Functional tests demonstrated that the purified rhVEGI-192 potently inhibited endothelial growth, induced endothelial apoptosis and suppressed neovascularization in chicken chorioallantoic membrane, indicating that the developed method allows preparation of rhVEGI-192 with high yield, solubility, and bioactivity. Most importantly, our study also demonstrates that VEGI-192 is capable of forming polymeric structure, which is possibly required for its antiangiogenic activity.


Subject(s)
Angiogenesis Inhibitors/biosynthesis , Neovascularization, Physiologic/drug effects , Recombinant Proteins/biosynthesis , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Cell Culture Techniques , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Chromatography, Affinity , Humans , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 15/chemistry , Tumor Necrosis Factor Ligand Superfamily Member 15/pharmacology
16.
Cytokine ; 46(1): 72-8, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19251437

ABSTRACT

Vascular endothelial growth inhibitor TNFSF15 (TL1A), a ligand for TNFRSF25 (DR3) and decoy receptor TNFRSF6B (DcR3), is expressed in human pulmonary arterial (HPAEC) and lung microvascular (HMVEC) endothelial cells where it might modulate inflammation and sickle vasculopathy. Pulmonary disease, endothelial abnormalities and inflammation are prominent features of sickle cell disease (SCD). Butyrate has opposing effects on endogenous TNFSF15 expression in pulmonary endothelium, acting as an inhibitor in HPAEC and an inducer in HMVEC. Similar effects were observed with a known cytokine TNF-alpha in these two cell types. Furthermore the TNFSF15 promoter utilized different combinations of cis-elements for its expression in these two cell types. AP1-like and G-rich sequence elements were critical for promoter activity in large vessel HPAEC while AP1-like and NF-kappaB consensus sequence elements were required in small vessel HMVEC. The requirement of an NF-kappaB sequence element by the TNFSF15 promoter in HMVEC but not in HPAEC supported the notion that HMVEC might be a target of inflammation and vasoocclusion in SCD. The dual effects of butyrate-dependant TNFSF15 regulation in lung endothelium may help in identify inflammatory pathways and understand the role of HMVEC in pathogenesis of vasoocclusion in SCD.


Subject(s)
Endothelium, Vascular/drug effects , Gene Expression Regulation , Lung/drug effects , Pulmonary Artery/drug effects , Sodium Oxybate/pharmacology , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Cell Line , Cells, Cultured , Endothelium, Vascular/cytology , Humans , Ligands , Lung/metabolism , Microcirculation , Models, Biological , NF-kappa B/metabolism , Promoter Regions, Genetic , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Tumor Necrosis Factor-alpha/metabolism
17.
J Immunol ; 179(6): 3742-51, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17785811

ABSTRACT

Angiogenesis is an essential component of chronic inflammation that is linked to carcinogenesis. In this study, we report that human vascular endothelial growth inhibitor (VEGI, TNF superfamily 15), an endothelial cell-produced antiangiogenic cytokine, induces mouse dendritic cell (DC) maturation, a critical event in inflammation-initiated immunity. VEGI-stimulated bone marrow-derived immature DCs display early activation of maturation signaling molecules NF-kappaB, STAT3, p38, and JNK, and cytoskeleton reorganization and dendrite formation. The activation signals are partially inhibited by using a neutralizing Ab against death domain-containing receptor-3 (DR3) or a truncated form of DR3 consisting of the extracellular domain, indicating an involvement of DR3 in the transmission of VEGI activity. A VEGI isoform, TL1A, does not induce similar activities under otherwise identical experimental conditions. Additionally, the cells reveal significantly enhanced expression of mature DC-specific marker CD83, secondary lymphoid tissue-directing chemokine receptor CCR7, the MHC class-II protein (MHC-II), and costimulatory molecules CD40, CD80, and CD86. Functionally, the cells exhibit decreased Ag endocytosis, increased cell surface distribution of MHC-II, and increased secretion of IL-12 and TNF. Moreover, VEGI-stimulated DCs are able to facilitate the differentiation of CD4+ naive T cells in cocultures. These findings suggest that the anticancer activity of VEGI arises from coupling the inhibition of endothelial cell growth with the promotion of the adaptive immune mechanisms through the stimulation of DC maturation.


Subject(s)
Angiogenesis Inhibitors/physiology , Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endothelium, Vascular/immunology , Endothelium, Vascular/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/physiology , Angiogenesis Inhibitors/biosynthesis , Animals , Antigens, Surface/biosynthesis , Antigens, Surface/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cattle , Cell Differentiation/genetics , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/cytology , Endothelium, Vascular/cytology , Female , Gene Expression Profiling , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis
18.
J Immunol ; 178(11): 7325-33, 2007 Jun 01.
Article in English | MEDLINE | ID: mdl-17513783

ABSTRACT

TNF-like cytokine (TL1A) is a newly identified member of the TNF superfamily of ligands that is important for T cell costimulation and Th1 polarization. However, despite increasing information about its functions, very little is known about expression of TL1A in normal or pathological states. In this study, we report that mononuclear phagocytes appear to be a major source of TL1A in rheumatoid arthritis (RA), as revealed by their strong TL1A expression in either synovial fluids or synovial tissue of rheumatoid factor (RF)-seropositive RA patients, but not RF-/RA patients. Accordingly, in vitro experiments revealed that human monocytes express and release significant amounts of soluble TL1A when stimulated with insoluble immune complexes (IC), polyethylene glycol precipitates from the serum of RF+/RA patients, or with insoluble ICs purified from RA synovial fluids. Monocyte-derived soluble TL1A was biologically active as determined by its capacity to induce apoptosis of the human erythroleukemic cell line TF-1, as well as to cooperate with IL-12 and IL-18 in inducing the production of IFN-gamma by CD4(+) T cells. Because RA is a chronic inflammatory disease with autoimmune etiology, in which ICs, autoantibodies (including RF), and various cytokines contribute to its pathology, our data suggest that TL1A could be involved in its pathogenesis and contribute to the severity of RA disease that is typical of RF+/RA patients.


Subject(s)
Antigen-Antibody Complex/physiology , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , Monocytes/immunology , Monocytes/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Antigen-Antibody Complex/biosynthesis , Antigen-Antibody Complex/isolation & purification , Arthritis, Rheumatoid/pathology , Cell Line, Tumor , Cells, Cultured , Cytokines/physiology , Humans , Immunoprecipitation , Monocytes/chemistry , Monocytes/pathology , Polyethylene Glycols/metabolism , Receptors, IgG/antagonists & inhibitors , Receptors, IgG/physiology , Solubility , Synovial Fluid/chemistry , Synovial Fluid/immunology , Synovial Fluid/metabolism , Synovial Membrane/chemistry , Synovial Membrane/immunology , Synovial Membrane/pathology , Tumor Necrosis Factor Ligand Superfamily Member 15/blood , Tumor Necrosis Factor Ligand Superfamily Member 15/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 15/physiology
19.
Exp Gerontol ; 41(5): 474-81, 2006 May.
Article in English | MEDLINE | ID: mdl-16626901

ABSTRACT

A convenient way to study processes of aging in distinct human tissues consists of a molecular analysis of cells from the tissue in question, that were explanted and grown in vitro until they reach senescence. Using human umbilical vein endothelial cells (HUVEC), we have established an in vitro senescence model for human endothelial cells. A major hallmark of HUVEC in vitro senescence is the increased frequency of apoptotic cell death, which occurs as a determining feature of HUVEC senescence. Senescent endothelial cells are also found in vivo in atherosclerotic lesions, suggesting that the presence of such cells may contribute to the development of vascular pathology. To elucidate mechanisms underlying endothelial cell senescence and age-associated apoptosis, gene expression analyses were carried out. In these experiments, we observed the up-regulation of genes coding for extracellular proteins in senescent HUVEC. In particular, a significant upregulation of interleukin-8, VEGI, and the IGF-binding proteins 3 and 5 was observed. Upregulation of these genes was confirmed by both RT-PCR and Western blot. In the case of interleukin-8, a roughly 50-fold upregulation of the protein was also found in cellular supernatants. The extracellular proteins encoded by these genes are well known for their ability to modulate the apoptotic response of human cells, and in the case of interleukin-8, clear links to the establishment of atherosclerotic lesions have been defined. The results described here support a new model, where changes in the secretome of human endothelial cells contribute to vascular aging and vascular pathology.


Subject(s)
Cellular Senescence/physiology , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Protein Biosynthesis/physiology , Apoptosis , Blotting, Western/methods , Cells, Cultured , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Humans , Insulin-Like Growth Factor Binding Proteins/biosynthesis , Insulin-Like Growth Factor Binding Proteins/genetics , Interleukin-8/biosynthesis , Interleukin-8/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Tumor Necrosis Factor Ligand Superfamily Member 15/biosynthesis , Tumor Necrosis Factor Ligand Superfamily Member 15/genetics , Umbilical Veins/cytology , Umbilical Veins/metabolism , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...