1
|
Zeng Y, Wang Y, Shi X, Zhao Y, Tang Y, Liu S, Zhu X. Porphyromonas gingivalis outer membrane vesicles augments proliferation and metastasis of oral squamous cell carcinoma cells. BMC Oral Health 2025; 25:701. [PMID: 40348995 PMCID: PMC12065146 DOI: 10.1186/s12903-025-05937-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 04/02/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Porphyromonas gingivalis (P. gingivalis) is closely related to Oral squamous cell carcinoma (OSCC), and P. gingivalis outer membrane vesicles (OMVs) is the main pathogenic factor, which is associated with periodontitis, atherosclerosis and other diseases. However, few studies have reported an association between P. gingivalis OMVs and OSCC. The purpose of this study was to establish the clinical relationship between P. gingivalis and OSCC based on clinical samples. Further, the effect of P. gingivalis OMVs on OSCC was observed with cell model in vitro, and the possible molecular mechanism was discussed. METHODS Immunohistochemistry was used to detect the abundance of P. gingivalis in OSCC and its paired paracancer tissues, and to analyze the correlation between P. gingivalis and clinicopathological parameters of patients. P. gingivalis OMVs were isolated to observe its effects on the proliferation and migration of OSCC cell lines. RNA-seq was performed and the expression of differentially expressed genes (DEGs) was detected by real-time quantitative PCR (RT-qPCR) to explore the potential mechnism of P. gingivalis OMVs on OSCC progression. RESULTS The abundance of P. gingivalis in OSCC was higher than that in para-cancerous tissues, and was positively correlated with the degree of tissue differentiation (P = 0.028), T stage (P < 0.001), and clinical stage (P = 0.011). P. gingivalis OMVs promoted the proliferation and migration of HN6 cells, and promoted the proliferation of CAL27 cells, but had no significant effect on its migration. P. gingivalis OMVs treatment attenuated the expressions of TNFSF15, ZNF292, ATRX, ASPM and KIF20B in CAL27 and HN6 cells. CONCLUSION This study suggests that P. gingivalis may be an indicator of poor prognosis for OSCC. P. gingivalis OMVs may down-regulate the expression of TNFSF15, ZNF292, ATRX, ASPM, KIF20B and participate in the occurrence and development of OSCC.
Collapse
Affiliation(s)
- Yanru Zeng
- Department of Oral Maxillo-Facial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yiyang Wang
- Department of Oral Maxillo-Facial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Xiaona Shi
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yuanhao Zhao
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Yue Tang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Shanshan Liu
- Department of Oral Maxillo-Facial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Oral Maxillo-Facial Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaofeng Zhu
- Department of Oral Maxillo-Facial Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Oral Maxillo-Facial Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
2
|
Abdelgalil AA, Monir R, Elmetwally M, Ghattas MH, Bazeed FB, Mesbah NM, Abo-Elmatty DM, Mehanna ET. The Relation of VEGFA, VEGFR2, VEGI, and HIF1A Genetic Variants and Their Serum Protein Levels with Breast Cancer in Egyptian Patients. Biochem Genet 2024; 62:547-573. [PMID: 37392242 DOI: 10.1007/s10528-023-10419-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 06/07/2023] [Indexed: 07/03/2023]
Abstract
Breast cancer is the most common type of cancer in Egyptian females. Polymorphisms in the angiogenesis pathway have been implicated previously in cancer risk and prognosis. The aim of the current study was to determine whether certain polymorphisms in the genes of vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor receptor 2 (VEGFR2), vascular endothelial growth inhibitor (VEGI), and hypoxia-inducible factor-1α (HIF1A) associated with breast cancer development. The study included 154 breast cancer patients and 132 apparently healthy age-matched females as a control group. VEGFA rs25648 genotyping was performed using (ARMS) PCR technique; while VEGFR2 rs2071559, VEGI rs6478106, and HIF-1α rs11549465 were genotyped by the PCR-RFLP method. Serum levels of VEGF, VEGFR2, VEGI, and HIF1A proteins in breast cancer patients and controls were measured by ELISA. There was a significant association between the VEGFA rs25648 C allele and breast cancer risk (OR 2.5, 95% CI 1.7-3.6, p < 0.001). VEGFA rs25648 C/C genotype was statistically significantly higher in breast cancer patients vs. control (p < 0.001). Participants with the T/T and T/C VEGFR2 rs2071559 genotypes had 5.46 and 5 higher odds, respectively, of having breast cancer than those with the C/C genotype. For the VEGI rs6478106 polymorphism, there was a higher proportion of C allele in breast cancer patients vs. control (p = 0.003). Moreover, the C/C genotype of VEGI rs6478106 was statistically significantly higher in breast cancer patients vs. control (p = 0.001). There was no significant difference in genotypes and allele frequencies of HIF1A rs11549465 polymorphism between breast cancer cases and control individuals (p > 0.05). Serum levels of VEGFA, VEGI, and HIF1A were considerably greater in women with breast cancer than in the control (p < 0.001). In conclusion, the genetic variants VEGFA rs25648, VEGFR2 rs2071559, and VEGI rs6478106 revealed a significant association with increased breast cancer risk in Egyptian patients.
Collapse
Affiliation(s)
- Amani A Abdelgalil
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| | - Rehan Monir
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Medical Biochemistry, Faculty of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mohamed Elmetwally
- Department of Surgical Oncology, Oncology Center, Mansoura University, Mansoura, Egypt
| | - Maivel H Ghattas
- Department of Medical Biochemistry, Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Fagr B Bazeed
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Noha M Mesbah
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Dina M Abo-Elmatty
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Eman T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
3
|
Luo Z, Wan R, Liu S, Feng X, Peng Z, Wang Q, Chen S, Shang X. Mechanisms of exercise in the treatment of lung cancer - a mini-review. Front Immunol 2023; 14:1244764. [PMID: 37691942 PMCID: PMC10483406 DOI: 10.3389/fimmu.2023.1244764] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Lung cancer constitutes a formidable menace to global health and well-being, as its incidence and mortality rate escalate at an alarming pace. In recent years, research has indicated that exercise has potential roles in both the prevention and treatment of lung cancer. However, the exact mechanism of the coordinating effect of exercise on lung cancer treatment is unclear, limiting the use of exercise in clinical practice. The purpose of this review is to explore the mechanisms through which exercise exerts its anticancer effects against lung cancer. This review will analyze the biological basis of exercise's anticancer effects on lung cancer, with a focus on aspects such as the tumor microenvironment, matrix regulation, apoptosis and angiogenesis. Finally, we will discuss future research directions and potential clinical applications.
Collapse
Affiliation(s)
- Zhiwen Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Renwen Wan
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Shan Liu
- Department of Endocrinology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinting Feng
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhen Peng
- Department of Sports Medicine, Shanghai General Hospital, Shanghai, China
| | - Qing Wang
- Department of Orthopaedics, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, Jiangsu, China
| | - Shiyi Chen
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiliang Shang
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Lu HJ, Chuang CY, Su CW, Chen MK, Yang WE, Yeh CM, Tang CH, Lin CW, Yang SF. Role of TNFSF15 variants in oral cancer development and clinicopathologic characteristics. J Cell Mol Med 2022; 26:5452-5462. [PMID: 36226563 PMCID: PMC9639028 DOI: 10.1111/jcmm.17569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/02/2022] [Accepted: 09/20/2022] [Indexed: 12/04/2022] Open
Abstract
Tumour necrosis family superfamily (TNFSF) member 15 (TNFSF15), encoded by TNFSF15, regulates immune responses and inflammation. However, the roles of TNFSF15 single‐nucleotide variants (SNVs; formerly SNPs) in oral cavity squamous cell carcinoma (OCSCC) remain unclear. This case–control study included 2523 participants (1324 patients with OCSCC [52.5%] and 1199 healthy controls [47.5%]). The effects of TNFSF15 rs3810936, rs6478108 and rs6478109 on cancer development and prognosis were analysed by real‐time PCR genotype assay. The Genotype‐Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA) databases were used to validate our findings. The results demonstrated that the patients with altered TNFSF15 SNVs had poorer histological differentiation than did those with wild‐type alleles. TNFSF15 SNVs were significantly associated with moderate‐to‐poor histological differentiation in univariate logistic regression. In the GTEx database, the expression of altered TNFSF15 SNVs in whole blood was lower than that of wild‐type alleles. However, the expression of altered SNVs in the upper aerodigestive mucosa was higher than that of wild‐type alleles. In the TCGA database, the patients with higher TNFSF15 expression had shorter overall survival than did those with lower TNFSF15 expression, especially for human papillomavirus‐negative and advanced staging groups. In conclusion, although TNFSF15 SNVs did not affect OCSCC development, the patients with altered TNFSF15 SNVs exhibited poorer histological differentiation. The patients with higher TNFSF15 expression had poorer prognosis than did those with lower TNFSF15 expression.
Collapse
Affiliation(s)
- Hsueh-Ju Lu
- Division of Hematology and Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Yi Chuang
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Otolaryngology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chun-Wen Su
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Mu-Kuan Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan.,Oral cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Wei-En Yang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Ming Yeh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Oral cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
5
|
Li J, Xie R, Jiang F, Li Y, Zhu Y, Liu Z, Liao M, Liu Y, Meng X, Chen S, Yu J, Du M, Wang X, Chen Y, Yan H. Tumor necrosis factor ligand-related molecule 1A maintains blood-retinal barrier via modulating SHP-1-Src-VE-cadherin signaling in diabetic retinopathy. FASEB J 2021; 35:e22008. [PMID: 34679191 DOI: 10.1096/fj.202100807rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/03/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022]
Abstract
An impaired blood-retinal barrier (BRB) leads to diabetic macular edema (DME), which is a major complication of Diabetic retinopathy (DR). Mediators such as inflammation cause BRB breakdown. However, the explicit mechanism of its disruption is largely unknown. In this study, we identified tumor necrosis factor ligand-related molecule 1A (TL1A) as a crucial factor which protect retinal endothelial cells integrity in DR. By providing both human and mouse data, we show that TL1A is significantly decreased in the retinas of DME patients and diabetic rodents. We further demonstrate that the loss of TL1A accelerated diabetes-induced retinal barrier breakdown. TL1A supplementation protects the diabetic retina against BRB breakdown. Mechanistically, TL1A stabilize intracellular junctions and protect vascular integrity by blocking SHP1-Src-regulated VE-cadherin phosphorylation. Collectively, our findings reveal that loss of TL1A in the retina leads to increased vascular permeability in DR, and that TL1A treatment is of potential therapeutic interest for the treatment of DME.
Collapse
Affiliation(s)
- Jianan Li
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Molecular ophthalmology, Tianjin Medical University, Tianjin, China.,Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Ruotian Xie
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Feng Jiang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yiming Li
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Molecular ophthalmology, Tianjin Medical University, Tianjin, China
| | - Yanfang Zhu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Molecular ophthalmology, Tianjin Medical University, Tianjin, China
| | - Zhiheng Liu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, China
| | - Mengyu Liao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Molecular ophthalmology, Tianjin Medical University, Tianjin, China
| | - Yuanyuan Liu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangda Meng
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Song Chen
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinguo Yu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mei Du
- Laboratory of Molecular ophthalmology, Tianjin Medical University, Tianjin, China.,Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaohong Wang
- Laboratory of Molecular ophthalmology, Tianjin Medical University, Tianjin, China.,Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yupeng Chen
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Institute of Urology, Tianjin Medical University, Tianjin, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China.,Laboratory of Molecular ophthalmology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
6
|
Zhu L, Ding Z, Li X, Wei H, Chen Y. Research Progress of Radiolabeled Asn-Gly-Arg (NGR) Peptides for Imaging and Therapy. Mol Imaging 2021; 19:1536012120934957. [PMID: 32862776 PMCID: PMC7466889 DOI: 10.1177/1536012120934957] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Asn-Gly-Arg (NGR) motifs have vasculature-homing properties via interactions with the aminopeptidase N (CD13) expressed on tumor neovasculature. Numerous NGR peptides with different molecular scaffolds have been exploited for targeted delivery of different compounds for imaging and therapy. When conjugated with NGR, complexes recognize the CD13 receptor expressed on the tumor vasculature, which improves the specificity to tumor and avoids systematic toxic reactions. Both preclinical and clinical studies performed with these products suggest that NGR-mediated vascular targeting is an effective strategy for delivering bioactive amounts of cytokines to tumor endothelial cells. For molecular imaging, radiolabeled peptides have been the most successful approach and have been translated into clinic. This review describes current data on radiolabeled tumor vasculature-homing NGR peptides for imaging and therapy.
Collapse
Affiliation(s)
- Liqin Zhu
- Department of Nuclear Medicine, 556508The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Zhikai Ding
- Department of Nuclear Medicine, 556508The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, People's Republic of China
| | - Xingliang Li
- Institute of Nuclear Physics and Chemistry, China Academy of Engineering Physics, Mianyang, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, People's Republic of China
| | - Hongyuan Wei
- Department of Nuclear Medicine, 556508The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Institute of Nuclear Physics and Chemistry, China Academy of Engineering Physics, Mianyang, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, People's Republic of China
| | - Yue Chen
- Department of Nuclear Medicine, 556508The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, Sichuan, People's Republic of China
| |
Collapse
|
7
|
VEGI downregulation is correlated with nodal metastasis and poor prognosis in lung adenocarcinoma. Mol Clin Oncol 2020; 14:25. [PMID: 33335733 PMCID: PMC7739847 DOI: 10.3892/mco.2020.2187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 07/11/2020] [Indexed: 11/12/2022] Open
Abstract
Although the incidence of lung cancer is increasing worldwide, the molecular mechanisms for its tumorigenesis, progression and prognosis remain unknown. As a member of the tumor necrosis factor superfamily, vascular endothelial growth inhibitor (VEGI) is involved in the development and progression of many malignant diseases. In the present study, the expression of VEGI and CD31 was examined via immunohistochemistry in non-small cell lung cancer (NSCLC) tissues obtained from 150 patients with NSCLC. The inhibitory effect of VEGI on tumor-associated blood vessel formation and growth was investigated by determining the relationship between VEGI protein expression and microvascular density (MVD). Prognostic significance was evaluated using the Kaplan-Meier method. VEGI expression was downregulated or lost in 68.7% (103/150) of patients with NSCLC, an effect that was more prevalent in adenocarcinoma (AC), 76.0% (57/75), than in squamous cell carcinoma, 61.3% (46/75). A significant negative correlation was indentified between VEGI expression and lymphovascular invasion (P=0.039) and lymph node metastasis (P=0.017) in AC tissue. Additionally, MVD was significantly lower in the VEGI-rich group compared with the VEGI-poor group. The downregulation of VEGI expression was also associated with poorer overall survival (P=0.011) in patients with AC. The present study therefore provides evidence that VEGI may be a new and effective prognostic marker of lung AC.
Collapse
|
8
|
Ali EMT, Abdallah HI, El-Sayed SM. Histomorphological, VEGF and TGF-β immunoexpression changes in the diabetic rats' ovary and the potential amelioration following treatment with metformin and insulin. J Mol Histol 2020; 51:287-305. [PMID: 32399705 DOI: 10.1007/s10735-020-09880-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 05/05/2020] [Indexed: 12/29/2022]
Abstract
Diabetes mellitus (DM) affects the ovary by reducing the number and diameters of ovarian follicles and increasing atretic follicles. Follicular growth and diameters depend on VEGF production. Hyperglycemia causes ovarian stromal and follicular degeneration then fibrosis by activating TGF-β. Insulin and metformin promote development of ovarian follicles and reduce atretic follicles. Therefore, the present study investigates the ovarian VEGF and TGF-β immune-expression and its variations in diabetic, insulin and metformin-treated rats. Forty adult female albino rats were divided equally into four groups: control, diabetic (STZ-induced diabetes), diabetic metformin-treated group (100 mg/kg/day orally/eight weeks) and diabetic insulin-treated group (5 U insulin /day). Ovarian sections were stained with hematoxylin and eosin, Masson's trichrome, immunohistochemistry for VEGF and TGF-β. The diabetic group showed noticeable atrophic and degenerative changes in cortex and medulla as well as increased density and distribution of the collagenous fibers. The number and diameter of primary, secondary and tertiary follicles were decreased. However, the number of atretic follicles and corpus luteum was increased. Significant decrease in the surface area percentage of VEGF immuno-expression and significant increase in TGF-β immuno-expression surface area percentage were detected. By treating animals with metformin and insulin, there was restoration of the ovarian histological structure more or less as in control. DM negatively affects the histological and morphometric parameters of ovaries. Furthermore, insulin showed more beneficial effects than metformin in hindering these complications by modifying the expression of VEGF and TGF-β.
Collapse
Affiliation(s)
- Eyad M T Ali
- Department of Anatomy, Faculty of medicine, Taibah University, Madinah, Kingdom of Saudi Arabia. .,Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Hesham I Abdallah
- Department of Anatomy, Faculty of medicine, Taibah University, Madinah, Kingdom of Saudi Arabia.,Department of Anatomy, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sayed M El-Sayed
- Department of Anatomy, Faculty of medicine, Taibah University, Madinah, Kingdom of Saudi Arabia.,Department of Anatomy, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
9
|
Wang T, Li J, Xie R, Wang J, Zhang W, Jiang F, Du M, Wang X, Huang B, Brant R, Zhang C, Yan H. Intraocular tumour necrosis factor ligand related molecule 1 A links disease progression of proliferative diabetic retinopathy after primary vitrectomy. Clin Exp Pharmacol Physiol 2020; 47:966-976. [PMID: 32064668 DOI: 10.1111/1440-1681.13284] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/17/2020] [Accepted: 02/13/2020] [Indexed: 11/30/2022]
Abstract
Tumour necrosis factor ligand related molecule 1 A (TL1A), a member of tumour necrosis factor superfamily, has been identified as a crucial regulator for vascular homeostasis and inflammation. However, the function of TL1A in diabetic retinopathy (DR) is largely unknown. This study aims to examine levels of TL1A in serum and intraocular fluid in patients with proliferative diabetic retinopathy (PDR), and to explore the correlation of intraocular TL1A with the prognosis of PDR progression after primary vitrectomy. Seventy-five patients (75 eyes) with PDR who underwent pars plana vitrectomy (PPV) and 19 patients (19 eyes) who received vitrectomy for idiopathic macular holes (IMH) as non-diabetic control group were enrolled in this prospective study. Serum, aqueous and vitreous fluid samples were collected during cataract and PPV surgery. Protein expressions of TL1A as well as other angiogenic and inflammatory cytokines in serum and intraocular fluid were measured. Correlations of intraocular TL1A concentrations with inflammatory cytokines were analyzed. We found both aqueous and vitreous TL1A levels were significantly higher in the PDR group than in control group (Paqueous = 0.026; Pvitreous <0.001). Angiogenic and inflammatory cytokines such as VEGF, IL-6, IL-8, MCP-1, MIP-1α, and MIP-1β were significantly higher in intraocular fluid in PDR group than in controls, which MCP-1 and MIP-1α showed positive correlation with intraocular TL1A levels. There is no significant difference in the levels of serum TL1A as well as other inflammatory cytokines between PDR patients and controls. Intraocular levels of TL1A were significantly lower in PDR progression group than in the stable group (Paqueous <0.001; Pvitreous <0.001). Multivariate logistic regression analyses revealed that lower levels of intraocular TL1A was an important risk factor for predicting PDR progression after primary PPV (ORaqueous = 0.717, Paqueous = 0.001; ORvitreous = 0.684; Pvitreous = 0.002). In conclusion, TL1A and multiple inflammatory cytokines were highly enriched in the intraocular fluid of PDR patients compared with the controls. Lower levels of intraocular TL1A were associated with development of PDR complications after primary PPV and might be used as prognostic factor in predicting the vitrectomy outcome in PDR patients.
Collapse
Affiliation(s)
- Tian Wang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianan Li
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ruotian Xie
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiaxing Wang
- Department of Ophthalmology, Emory University, Atlanta, GA, USA
| | - Wei Zhang
- Tianjin Eye Hospital, Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Clinical College of Ophthalmology Tianjin Medical University, Tianjin, China
| | - Feng Jiang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mei Du
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaohong Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Bo Huang
- Department of Ophthalmology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Rodrigo Brant
- Department of Ophthalmology and Visual Sciences, Federal University of São Paulo, São Paulo, Brazil
| | - Cheng Zhang
- Department of Ophthalmology & Visual Sciences, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
10
|
Gao Y, Wang Y, Wang X, Wang Y, Zhang X, Sun X. TNF-like ligand 1A is associated with progression and prognosis of human gastric cancer. Onco Targets Ther 2019; 12:7715-7723. [PMID: 31571922 PMCID: PMC6756834 DOI: 10.2147/ott.s210939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022] Open
Abstract
Purpose This study aimed to investigate the function of TNF-like ligand 1A (TL1A) in the tumorigenesis and progression of gastric cancer (GC). Methods RNA-seq gene expression and clinical information for GC patients were obtained from The Cancer Genome Atlas (TCGA) database. Differentially expressed genes (DEGs) between GC tissue samples and normal controls were screened with the edgeR package. Identification of gene co-expression and functional enrichment analyses were performed with Pearson’s correlation analysis and gene set enrichment analysis (GSEA), respectively. Lastly, survival analysis was performed using the Kaplan-Meier method with the log rank test. Results TL1A expression in GC tissue samples were significantly higher than that in normal controls (LogFC=1.07 and P=8.90E-07). Moreover, 215 genes, co-expressed with TL1A, and 21 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were obtained. Next, the miRNA-lncRNA/mRNA network, comprising 7 miRNAs, 27 lncRNAs, and 21 mRNAs, was constructed based on key genes from intersections between co-expression analysis and GSEA. In addition, survival analysis results demonstrated that TL1A (P=2.6e−07) was significantly associated with the overall survival (OS) of GC patients. Conclusion TL1A was involved in the tumorigenesis and progression of GC, and was significantly associated with the OS of GC patients.
Collapse
Affiliation(s)
- Yaxian Gao
- Department of Immunology, China Medical University, Shenyang, Liaoning 110000, People's Republic of China.,Department of Immunology, Chengde Medical College, Chengde, Hebei 067000, People's Republic of China
| | - Yuanyuan Wang
- Department of Anesthesiology, The Fourth Affiliated Hospital, China Medical University, Shenyang, Liaoning 110000, People's Republic of China
| | - Xiao Wang
- Department of Immunology, China Medical University, Shenyang, Liaoning 110000, People's Republic of China
| | - Yongwei Wang
- Department of Anatomy, Chengde Medical College, Chengde, Hebei 067000, People's Republic of China
| | - Xiaoqing Zhang
- Department of Immunology, China Medical University, Shenyang, Liaoning 110000, People's Republic of China
| | - Xun Sun
- Department of Immunology, China Medical University, Shenyang, Liaoning 110000, People's Republic of China
| |
Collapse
|
11
|
Vascular Endothelial Growth Inhibitor, a Cytokine of the Tumor Necrosis Factor Family, is Associated With Epithelial-Mesenchymal Transition in Renal Cell Carcinoma. Appl Immunohistochem Mol Morphol 2019; 26:727-733. [PMID: 28362712 DOI: 10.1097/pai.0000000000000517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Previous studies have revealed that the activation of the epithelial-mesenchymal transition (EMT) endows metastatic properties upon cancer cells to promote invasion and migration. In this study, immunohistochemical analysis was performed in 50 cases of clear cell renal cell carcinoma (RCC) and paired normal kidney tissues. We detected the expression of vascular endothelial growth inhibitor (VEGI) and EMT markers (E-cadherin, fibronectin, and Slug) and recorded the clinical, pathologic, and follow-up (median follow-up: 79.0 mo) information. The expression of VEGI and E-cadherin was significantly lower in RCC tissues compared with normal kidney tissues (P<0.001). However, the expression of fibronectin and Slug was higher in RCC tissues (P<0.05). VEGI and EMT marker expression marginally differed in tumor size and stage. Significant differences were found in the pathologic grade (P<0.05). The Spearman correlation analysis suggested a positive correlation between VEGI and E-cadherin (r=0.451, P<0.01). A negative correlation was shown between VEGI and fibronectin (r=-0.465, P<0.01). There was also a negative correlation between VEGI and Slug (r=-0.758, P<0.01). During the 79.0 months (range, 7 to 119 mo) of follow-up, 6 patients died due to RCC, and the tumor-free survival rate was 88% (44/50). We did not find a significant correlation between VEGI/EMT markers (E-cadherin, fibronectin, and Slug) and overall survival (P>0.05). Our findings indicate that VEGI plays an important role in EMT in RCC. It suggests that VEGI may be investigated as a disease biomarker and therapeutic target in RCC.
Collapse
|
12
|
Yang G, Han Z, Xiong J, Wang S, Wei H, Qin T, Xiao H, Liu Y, Xu L, Qi J, Zhang Z, Jiang R, Zhang J, Li L. Inhibition of intracranial hemangioma growth and hemorrhage by TNFSF15. FASEB J 2019; 33:10505-10514. [PMID: 31242765 DOI: 10.1096/fj.201802758rrr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Gui‐Li Yang
- Key Laboratory of Post‐Neuroinjury RepairRegeneration in Central Nervous SystemMinistry of EducationTianjin Neurological InstituteTianjin Medical University General Hospital Tianjin China
| | - Zhenying Han
- Key Laboratory of Post‐Neuroinjury RepairRegeneration in Central Nervous SystemMinistry of EducationTianjin Neurological InstituteTianjin Medical University General Hospital Tianjin China
| | - Jianhua Xiong
- Key Laboratory of Post‐Neuroinjury RepairRegeneration in Central Nervous SystemMinistry of EducationTianjin Neurological InstituteTianjin Medical University General Hospital Tianjin China
| | - Shizhao Wang
- Key Laboratory of Post‐Neuroinjury RepairRegeneration in Central Nervous SystemMinistry of EducationTianjin Neurological InstituteTianjin Medical University General Hospital Tianjin China
| | - Huijie Wei
- Key Laboratory of Post‐Neuroinjury RepairRegeneration in Central Nervous SystemMinistry of EducationTianjin Neurological InstituteTianjin Medical University General Hospital Tianjin China
| | - Ting‐Ting Qin
- Tianjin Medical UniversityCancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for Cancer Tianjin China
| | - Huaiyuan Xiao
- Tianjin Medical UniversityCancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for Cancer Tianjin China
| | - Ye Liu
- Key Laboratory of Post‐Neuroinjury RepairRegeneration in Central Nervous SystemMinistry of EducationTianjin Neurological InstituteTianjin Medical University General Hospital Tianjin China
| | - Li‐Xia Xu
- State Key Laboratory of Medicinal Chemical BiologyNankai University College of PharmacyTianjin Key Laboratory of Molecular Drug Research Tianjin China
| | - Jian‐Wei Qi
- State Key Laboratory of Medicinal Chemical BiologyNankai University College of PharmacyTianjin Key Laboratory of Molecular Drug Research Tianjin China
| | - Zhi‐Song Zhang
- State Key Laboratory of Medicinal Chemical BiologyNankai University College of PharmacyTianjin Key Laboratory of Molecular Drug Research Tianjin China
| | - Rongcai Jiang
- Key Laboratory of Post‐Neuroinjury RepairRegeneration in Central Nervous SystemMinistry of EducationTianjin Neurological InstituteTianjin Medical University General Hospital Tianjin China
| | - Jianning Zhang
- Key Laboratory of Post‐Neuroinjury RepairRegeneration in Central Nervous SystemMinistry of EducationTianjin Neurological InstituteTianjin Medical University General Hospital Tianjin China
| | - Lu‐Yuan Li
- State Key Laboratory of Medicinal Chemical BiologyNankai University College of PharmacyTianjin Key Laboratory of Molecular Drug Research Tianjin China
| |
Collapse
|
13
|
Gao H, Niu Z, Zhang Z, Wu H, Xie Y, Yang Z, Li A, Jia Z, Zhang X. TNFSF15 promoter polymorphisms increase the susceptibility to small cell lung cancer: a case-control study. BMC MEDICAL GENETICS 2019; 20:29. [PMID: 30736740 PMCID: PMC6368786 DOI: 10.1186/s12881-019-0762-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/31/2019] [Indexed: 12/30/2022]
Abstract
BACKGROUND Tumor necrosis factor superfamily member 15 (TNFSF15) is closely related to tumorigenesis and development. This study aimed to investigate the correlations between TNFSF15 polymorphisms and genetic susceptibility to lung cancer. METHODS This case-control study included 209 small cell lung cancer patients (SCLC), 340 non- small cell lung cancer patients (NSCLC) and 460 health controls. TNFSF15-638 A > G and - 358 T > C polymorphisms were genotyped by polymerase chain reaction-restrictive fragment length polymorphism (PCR-RFLP) analysis. Odds ratio (OR) and 95% confidence interval (95% CI) were estimated by unconditional logistic regression. RESULTS Our results showed that subjects carrying the TNFSF15-638GG genotype or -358CC genotype were more likely to develop SCLC (-638GG, OR = 1.84, 95%CI = 1.13-2.99; -358CC, OR = 2.44, 95%CI = 1.46-4.06), but not NSCLC (P > 0.05). In stratified analysis, -638GG genotype was related to SCLC among males (OR = 1.95, 95%CI = 1.09-3.45, P = 0.023) and older patients (OR = 2.93, 95%CI = 1.44-8.68, P = 0.006). However, -358CC genotype was associated with SCLC among females (OR = 8.42, 95%CI = 2.22-31.89, P = 0.002) and older subjects with OR (95%CI) of 11.04 (3.57-34.15) (P < 0.001). Moreover, TNFSF15 -358CC was linked with a higher risk of SCLC among non-smokers (OR = 2.54, 95%CI = 1.20-5.35, P = 0.015) but not among smokers (OR = 1.88, 95%CI = 0.92-3.84, P = 0.086). CONCLUSION These findings highlight the importance of TNFSF15 polymorphisms in the development of SCLC.
Collapse
Affiliation(s)
- Hui Gao
- College of Life Science, North China University of Science and Technology, Tangshan, 063210 China
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 China
| | - Zeren Niu
- College of Life Science, North China University of Science and Technology, Tangshan, 063210 China
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 China
| | - Zhi Zhang
- Affliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, 063000 China
| | - Hongjiao Wu
- College of Life Science, North China University of Science and Technology, Tangshan, 063210 China
| | - Yuning Xie
- College of Life Science, North China University of Science and Technology, Tangshan, 063210 China
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 China
| | - Zhenbang Yang
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, 063210 China
| | - Ang Li
- College of Life Science, North China University of Science and Technology, Tangshan, 063210 China
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 China
| | - Zhenxian Jia
- College of Life Science, North China University of Science and Technology, Tangshan, 063210 China
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 China
| | - Xuemei Zhang
- College of Life Science, North China University of Science and Technology, Tangshan, 063210 China
| |
Collapse
|
14
|
Xu W, Xu Z, Huang L, Qin EQ, Zhang JL, Zhao P, Tu B, Shi L, Li WG, Chen WW. Transcriptome Sequencing Identifies Novel Immune Response Genes Highly Related to the Severity of Human Adenovirus Type 55 Infection. Front Microbiol 2019; 10:130. [PMID: 30787914 PMCID: PMC6372566 DOI: 10.3389/fmicb.2019.00130] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 01/21/2019] [Indexed: 01/01/2023] Open
Abstract
Human adenovirus type 55 (HAdV-55) is considered a highly virulent pathogen causing severe and even deadly pneumonia in immunocompetent people. The mechanisms of HAdV-55-induced initiation and progression of severe pneumonia remain ambiguous. In the current study, we endeavored to identify novel immune response genes which are substantially involved in the pathogenesis of severe inflammation in HAdV-55-infected patients. HAdV-55-infected patients with upper respiratory tract symptoms (minor patients) and pneumonia (severe patients) were enrolled. Through transcriptome sequencing and quantitative real-time PCR, the peripheral blood mononuclear cells of the patients were analyzed. We found that the expression of eight genes, including Il18, Il36b, Il17rc, Tnfsf10, Tnfsf11, Tnfsf14, Tnfsf15, and Il1a, were closely correlated with the severity of HAdV-55 infection. Most of these genes belong to interleukin-1 family or tumor necrosis factor (TNF) superfamily, respectively. The changes in gene expression were confirmed by Western blot assay. Our data will be crucial for deepening the understanding of the pathogenic mechanisms of severe pneumonia in HAdV-55 infection.
Collapse
Affiliation(s)
- Wen Xu
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing, China
| | - Zhe Xu
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing, China
| | - Lei Huang
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing, China
| | - En-Qiang Qin
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing, China
| | - Jie-Li Zhang
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing, China
| | - Peng Zhao
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing, China
| | - Bo Tu
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing, China
| | - Lei Shi
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing, China
| | - Wen-Gang Li
- Radiation Oncology Center, 302 Military Hospital of China, Beijing, China
| | - Wei-Wei Chen
- Treatment and Research Center for Infectious Diseases, 302 Military Hospital of China, Beijing, China
| |
Collapse
|
15
|
Zhao Q, Hong B, Liu T, Ji Y, Tang X, Gong K, Ye L, Yang Y, Zhang N. VEGI174 protein and its functional domain peptides exert antitumour effects on renal cell carcinoma. Int J Oncol 2018; 54:390-398. [PMID: 30431089 DOI: 10.3892/ijo.2018.4632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 10/24/2018] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth inhibitor (VEGI) has been identified as an anti‑angiogenic cytokine. However, the effects of VEGI174 protein, and its functional domain peptides V7 and V8, on renal cell carcinoma (RCC) remain unknown. In the present study, the protein and peptides were biosynthesised as experimental agents. The A498 and 786‑O RCC cell lines, and an established mouse xenograft model, were separately treated with VEGI174, V7 or V8. Cellular functions, including proliferation, migration and invasion, were subsequently detected. Cell migration and invasion were monitored using the xCELLigence system. Furthermore, tumour growth and mouse behaviours, including mobility, appetite and body weight, were assessed. The results demonstrated that VEGI174, V7 and V8 inhibited the proliferation, migration and invasion of A498 and 786‑O cell lines when administered at concentrations of 1 and 100 pM, 10 nM and 1 µM. The inhibitory effects exhibited dose‑ and time‑dependent antitumour activity. Furthermore, VEGI174, V7 and V8 inhibited tumour growth in A498 and 786‑O xenograft mice. In the A498 xenografts, the tumour growth inhibition (TGI) rates in the VEGI174‑, V7‑ and V8‑treated groups were 71, 20 and 31%, respectively. In the 786‑O xenografts, the TGI rates in the VEGI174‑, V7‑ and V8‑treated groups were 34, 26 and 31%, respectively. There was no significant loss in body weight and no cases of mortality were observed for all treated mice. In conclusion, VEGI174, V7 and V8 exhibited potential antitumour effects and were well tolerated in vivo. V7 and V8, as functional domain peptides of the VEGI174 protein, may be studied for the future treatment of RCC.
Collapse
Affiliation(s)
- Qiang Zhao
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Baoan Hong
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, Beijing 100034, P.R. China
| | - Tiezhu Liu
- Department of Urology, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163001, P.R. China
| | - Yongpeng Ji
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Xinxin Tang
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Institute of Urology, Peking University, Beijing 100034, P.R. China
| | - Lin Ye
- Metastasis and Angiogenesis Research Group, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Yong Yang
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| | - Ning Zhang
- Department of Urology, Beijing Institute for Cancer Research, Beijing Cancer Hospital, Beijing 100142, P.R. China
| |
Collapse
|
16
|
Qin T, Huang D, Liu Z, Zhang X, Jia Y, Xian CJ, Li K. Tumor necrosis factor superfamily 15 promotes lymphatic metastasis via upregulation of vascular endothelial growth factor-C in a mouse model of lung cancer. Cancer Sci 2018; 109:2469-2478. [PMID: 29890027 PMCID: PMC6113425 DOI: 10.1111/cas.13665] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/29/2018] [Accepted: 06/01/2018] [Indexed: 12/16/2022] Open
Abstract
Lymphatic metastasis is facilitated by lymphangiogenic growth factor vascular endothelial growth factor-C (VEGFC) that is secreted by some primary tumors. We previously identified tumor necrosis factor superfamily 15 (TNFSF15), a blood vascular endothelium-derived cytokine, in lymphatic endothelial cells, as a key molecular modulator during lymphangiogenesis. However, the effect of TNFSF15 on tumor lymphatic metastasis and the underlying molecular mechanisms remain unclear. We report here that TNFSF15, which is known to inhibit primary tumor growth by suppressing angiogenesis, can promote lymphatic metastasis through facilitating lymphangiogenesis in tumors. Mice bearing tumors induced by A549 cells stably overexpressing TNFSF15 exhibited a significant increase in densities of lymphatic vessels and a marked enhancement of A549 tumor cells in newly formed lymphatic vessels in the primary tumors as well as in lymph nodes. Treatment of A549 cells with TNFSF15 results in upregulation of VEGFC expression, which can be inhibited by siRNA gene silencing of death domain-containing receptor-3 (DR3), a cell surface receptor for TNFSF15. In addition, TNFSF15/DR3 signaling pathways in A549 cells include activation of NF-κB during tumor lymphangiogenesis. Our data indicate that TNFSF15, a cytokine mainly produced by blood endothelial cells, facilitates tumor lymphangiogenesis by upregulating VEGFC expression in A549 cells, contributing to lymphatic metastasis in tumor-bearing mice. This finding also suggests that TNFSF15 may have potential as an indicator for prognosis evaluation.
Collapse
Affiliation(s)
- Tingting Qin
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Dingzhi Huang
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Zhujun Liu
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaoling Zhang
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yanan Jia
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Kai Li
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.,Tianjin's Clinical Research Center for Cancer, Tianjin, China.,Department of Thoracic Oncology, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
17
|
Zhang K, Cai HX, Gao S, Yang GL, Deng HT, Xu GC, Han J, Zhang QZ, Li LY. TNFSF15 suppresses VEGF production in endothelial cells by stimulating miR-29b expression via activation of JNK-GATA3 signals. Oncotarget 2018; 7:69436-69449. [PMID: 27589684 PMCID: PMC5342489 DOI: 10.18632/oncotarget.11683] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 08/25/2016] [Indexed: 02/05/2023] Open
Abstract
Vascular endothelial cell growth factor (VEGF) plays a pivotal role in promoting neovascularization. VEGF gene expression in vascular endothelial cells in normal tissues is maintained at low levels but becomes highly up-regulated in a variety of disease settings including cancers. Tumor necrosis factor superfamily 15 (TNFSF15; VEGI; TL1A) is an anti-angiogenic cytokine prominently produced by endothelial cells in a normal vasculature. We report here that VEGF production in mouse endothelial cell line bEnd.3 can be inhibited by TNFSF15 via microRNA-29b (miR-29b) that targets the 3'-UTR of VEGF transcript. Blocking TNFSF15 activity by using either siRNA against the TNFSF15 receptor known as death domain-containing receptor-3 (DR3; TNFRSF25), or a neutralizing antibody 4-3H against TNFSF15, led to inhibition of miR-29b expression and reinvigoration of VEGF production. In addition, we found that TNFSF15 activated the JNK signaling pathway as well as the transcription factor GATA3, resulting in enhanced miR-29b production. Treatment of the cells either with SP600125, an inhibitor of JNK, or with JNK siRNA, led to eradication of TNFSF15-induced GATA3 expression. Moreover, GATA3 siRNA suppressed TNFSF15-induced miR-29b expression. These findings suggest that VEGF gene expression can be suppressed by TNFSF15-stimulated activation of the JNK-GATA3 signaling pathway which gives rise to up-regulation of miR-29b.
Collapse
Affiliation(s)
- Kun Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital, Sichuan University, Chengdu, China
| | - Hong-Xing Cai
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital, Sichuan University, Chengdu, China
| | - Shan Gao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital, Sichuan University, Chengdu, China
| | - Gui-Li Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital, Sichuan University, Chengdu, China
| | - Hui-Ting Deng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital, Sichuan University, Chengdu, China
| | - Guo-Ce Xu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital, Sichuan University, Chengdu, China
| | - Jihong Han
- Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital, Sichuan University, Chengdu, China.,College of Life Sciences, Nankai University, Tianjin, China
| | - Qiang-Zhe Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital, Sichuan University, Chengdu, China
| | - Lu-Yuan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Zhao J, Cheng F, Jia P, Cox N, Denny JC, Zhao Z. An integrative functional genomics framework for effective identification of novel regulatory variants in genome-phenome studies. Genome Med 2018; 10:7. [PMID: 29378629 PMCID: PMC5789733 DOI: 10.1186/s13073-018-0513-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 01/04/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Genome-phenome studies have identified thousands of variants that are statistically associated with disease or traits; however, their functional roles are largely unclear. A comprehensive investigation of regulatory mechanisms and the gene regulatory networks between phenome-wide association study (PheWAS) and genome-wide association study (GWAS) is needed to identify novel regulatory variants contributing to risk for human diseases. METHODS In this study, we developed an integrative functional genomics framework that maps 215,107 significant single nucleotide polymorphism (SNP) traits generated from the PheWAS Catalog and 28,870 genome-wide significant SNP traits collected from the GWAS Catalog into a global human genome regulatory map via incorporating various functional annotation data, including transcription factor (TF)-based motifs, promoters, enhancers, and expression quantitative trait loci (eQTLs) generated from four major functional genomics databases: FANTOM5, ENCODE, NIH Roadmap, and Genotype-Tissue Expression (GTEx). In addition, we performed a tissue-specific regulatory circuit analysis through the integration of the identified regulatory variants and tissue-specific gene expression profiles in 7051 samples across 32 tissues from GTEx. RESULTS We found that the disease-associated loci in both the PheWAS and GWAS Catalogs were significantly enriched with functional SNPs. The integration of functional annotations significantly improved the power of detecting novel associations in PheWAS, through which we found a number of functional associations with strong regulatory evidence in the PheWAS Catalog. Finally, we constructed tissue-specific regulatory circuits for several complex traits: mental diseases, autoimmune diseases, and cancer, via exploring tissue-specific TF-promoter/enhancer-target gene interaction networks. We uncovered several promising tissue-specific regulatory TFs or genes for Alzheimer's disease (e.g. ZIC1 and STX1B) and asthma (e.g. CSF3 and IL1RL1). CONCLUSIONS This study offers powerful tools for exploring the functional consequences of variants generated from genome-phenome association studies in terms of their mechanisms on affecting multiple complex diseases and traits.
Collapse
Affiliation(s)
- Junfei Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA
| | - Feixiong Cheng
- Center for Cancer Systems Biology and Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
- Center for Complex Networks Research, Northeastern University, Boston, MA, 02215, USA
| | - Peilin Jia
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA
| | - Nancy Cox
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Joshua C Denny
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, 7000 Fannin St. Suite 820, Houston, TX, 77030, USA.
- Human Genetics Center, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Spoletini M, Taurone S, Tombolini M, Minni A, Altissimi G, Wierzbicki V, Giangaspero F, Parnigotto PP, Artico M, Bardella L, Agostinelli E, Pastore FS. Trophic and neurotrophic factors in human pituitary adenomas (Review). Int J Oncol 2017; 51:1014-1024. [PMID: 28902350 DOI: 10.3892/ijo.2017.4120] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 07/17/2017] [Indexed: 11/06/2022] Open
Abstract
The pituitary gland is an organ that functionally connects the hypothalamus with the peripheral organs. The pituitary gland is an important regulator of body homeostasis during development, stress, and other processes. Pituitary adenomas are a group of tumors arising from the pituitary gland: they may be subdivided in functional or non-functional, depending on their hormonal activity. Some trophic and neurotrophic factors seem to play a key role in the development and maintenance of the pituitary function and in the regulation of hypothalamo-pituitary-adrenocortical axis activity. Several lines of evidence suggest that trophic and neurotrophic factors may be involved in pituitary function, thus suggesting a possible role of the trophic and neurotrophic factors in the normal development of pituitary gland and in the progression of pituitary adenomas. Additional studies might be necessary to better explain the biological role of these molecules in the development and progression of this type of tumor. In this review, in light of the available literature, data on the following neurotrophic factors are discussed: ciliary neurotrophic factor (CNTF), transforming growth factors β (TGF‑β), glial cell line-derived neurotrophic factor (GDNF), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), vascular endothelial growth inhibitor (VEGI), fibroblast growth factors (FGFs) and epidermal growth factor (EGF) which influence the proliferation and growth of pituitary adenomas.
Collapse
Affiliation(s)
- Marialuisa Spoletini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, 'Sapienza' University of Rome, Rome, Italy
| | - Samanta Taurone
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | - Mario Tombolini
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | - Antonio Minni
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | | | | | - Felice Giangaspero
- Department of Radiology, Oncology and Anatomic Pathology, 'Sapienza' University of Rome, Rome, Italy
| | - Pier Paolo Parnigotto
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling (TES) Onlus, Padua, Italy
| | - Marco Artico
- Department of Sensory Organs, 'Sapienza' University of Rome, Rome, Italy
| | - Lia Bardella
- Department of Neurology and Psychiatry, 'Sapienza' University of Rome, Rome, Italy
| | - Enzo Agostinelli
- Department of Biochemical Sciences 'A. Rossi Fanelli', 'Sapienza' University of Rome, Rome, Italy
| | - Francesco Saverio Pastore
- Department of Systems' Medicine, Division of Neurosurgery, University of Rome 'Tor Vergata', Rome, Italy
| |
Collapse
|
20
|
Zhang N, Hong B, Lian W, Zhou C, Chen S, Du X, Deng X, Duoerkun S, Li Q, Yang Y, Gong K. Vascular endothelial growth inhibitor 174 and its functional domains inhibit epithelial-mesenchymal transition in renal cell carcinoma cells in vitro. Int J Mol Med 2017; 40:569-575. [PMID: 28656288 DOI: 10.3892/ijmm.2017.3033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 06/08/2017] [Indexed: 11/06/2022] Open
Abstract
The present study was carried out to investigate the effects of vascular endothelial growth inhibitor 174 (VEGI174) and its functional domains (V7 and V8) on epithelial‑mesenchymal transition (EMT) in renal cell carcinoma (RCC) cells in vitro. The RCC cell lines A498 and 786‑O were used in this study. Based on our preliminary study, we selected full‑length VEGI174 and its functional domains (V7 and V8) as the target genes in this study. Plasmids containing VEGI174, V7 or V8 transgenes were constructed and transfected into A498 and 786‑O cell lines. Cytological activity was tested during cell culture. Quantitative PCR and western blot analysis were performed to determine the expression levels of EMT markers (E‑cadherin, vimentin, β‑catenin and Slug). Overexpression of VEGI174, V7 or V8 did not have a significant influence on cell viability (P>0.05). The mRNA level of E‑cadherin was significantly upregulated, while that of vimentin was downregulated in A498VEGIexp, A498V7exp, A498V8exp, 786‑OVEGIexp, 786‑OV7exp and 786‑OV8exp cells compared with the cells containing the empty plasmid controls (P<0.05). The western blot results showed that changes in protein expression levels were consistent with the changes in mRNA expression. Both the mRNA and protein expression levels of β‑catenin and Slug were downregulated in the A498VEGIexp, A498V7exp, A498V8exp, 786‑OVEGIexp, 786‑OV7exp and 786‑OV8exp cells. In conclusion, overexpression of VEGI174, V7 or V8 inhibited EMT in A498 and 786‑O cells. Notably, V7 and V8 are two effective functional domains of VEGI174 that have the potential to be studied for peptide synthesis and the treatment of RCC.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Urology, Peking University Cancer Hospital, Beijing Institute for Cancer Research, Beijing 100142, P.R. China
| | - Baoan Hong
- Department of Urology, Peking University First Hospital, Beijing 100034, P.R. China
| | - Wenyong Lian
- Department of Urology, Xinjiang Production and Construction Corps First Division Hospital, Aksu, Xinjiang 843000, P.R. China
| | - Changhua Zhou
- School of Pharmaceutical Sciences, Center for Cellular and Structural Biology, Sun Yat-Sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Siqi Chen
- School of Pharmaceutical Sciences, Center for Cellular and Structural Biology, Sun Yat-Sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Xin Du
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, P.R. China
| | - Xiaohu Deng
- Department of Urology, Karamay People's Hospital, Karamay, Xinjiang 834000, P.R. China
| | - Shayiremu Duoerkun
- Department of Urology, Hami District Central Hospital, Hami, Xinjiang 839000, P.R. China
| | - Qing Li
- School of Pharmaceutical Sciences, Center for Cellular and Structural Biology, Sun Yat-Sen University, Guangzhou, Guangdong 510006, P.R. China
| | - Yong Yang
- Department of Urology, Peking University Cancer Hospital, Beijing Institute for Cancer Research, Beijing 100142, P.R. China
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Beijing 100034, P.R. China
| |
Collapse
|
21
|
Deng HT, Liu HL, Zhai BB, Zhang K, Xu GC, Peng XM, Zhang QZ, Li LY. Vascular endothelial growth factor suppresses TNFSF15 production in endothelial cells by stimulating miR-31 and miR-20a expression via activation of Akt and Erk signals. FEBS Open Bio 2016; 7:108-117. [PMID: 28097093 PMCID: PMC5221472 DOI: 10.1002/2211-5463.12171] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/18/2016] [Accepted: 11/26/2016] [Indexed: 02/05/2023] Open
Abstract
Tumor necrosis factor superfamily‐15 (TNFSF15; VEGI; TL1A) is a negative modulator of angiogenesis for blood vessel homeostasis and is produced by endothelial cells in a mature vasculature. It is known to be downregulated by vascular endothelial growth factor (VEGF), a major regulator of neovascularization but the mechanism of this interaction is unclear. Here we report that VEGF is able to stimulate the production of two microRNAs, miR‐20a and miR‐31, which directly target the 3′‐UTR of TNFSF15. Additionally, we show that two VEGF‐stimulated cell growth signals, Erk and Akt, are responsible for promoting the expression of miR‐20a and miR‐31. Treatment of human umbilical vein endothelial cells (HUVECs) with Akt inhibitor LY294002 results in diminished miR‐20a and miR‐31 production, while Erk inhibitor U0126 prevented VEGF‐stimulated expression of miR‐20a but not that of miR‐31. Furthermore, inactivation of either Erk or Akt signals restores TNFSF15 gene expression. In an angiogenesis assay, elevated miR‐20a or miR‐31 levels in HUVECs leads to enhancement of capillary‐like tubule formation in vitro, whereas lowered miR‐20a and miR‐31 levels results in an inhibition. These findings are consistent with the view that miR‐20a and miR‐31 mediate VEGF‐induced downregulation of TNFSF15. Targeting these microRNA molecules may therefore provide an effective approach to inhibit angiogenesis.
Collapse
Affiliation(s)
- Hui-Ting Deng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research Nankai University China; Collaborative Innovation Center for Biotherapy Nankai University West China Hospital Sichuan University Chengdu China
| | - Hai-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research Nankai University China; Collaborative Innovation Center for Biotherapy Nankai University West China Hospital Sichuan University Chengdu China
| | - Bei-Bei Zhai
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research Nankai University China; Collaborative Innovation Center for Biotherapy Nankai University West China Hospital Sichuan University Chengdu China
| | - Kun Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research Nankai University China; Collaborative Innovation Center for Biotherapy Nankai University West China Hospital Sichuan University Chengdu China
| | - Guo-Ce Xu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research Nankai University China; Collaborative Innovation Center for Biotherapy Nankai University West China Hospital Sichuan University Chengdu China
| | - Xue-Mei Peng
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research Nankai University China; Collaborative Innovation Center for Biotherapy Nankai University West China Hospital Sichuan University Chengdu China
| | - Qiang-Zhe Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research Nankai University China; Collaborative Innovation Center for Biotherapy Nankai University West China Hospital Sichuan University Chengdu China
| | - Lu-Yuan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research Nankai University China; Collaborative Innovation Center for Biotherapy Nankai University West China Hospital Sichuan University Chengdu China
| |
Collapse
|
22
|
Zhang ZH, Chen QZ, Jiang F, Townsend TA, Mao CJ, You CY, Yang WH, Sun ZY, Yu JG, Yan H. Changes in TL1A levels and associated cytokines during pathogenesis of diabetic retinopathy. Mol Med Rep 2016; 15:573-580. [PMID: 28000874 PMCID: PMC5364842 DOI: 10.3892/mmr.2016.6048] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 10/04/2016] [Indexed: 01/08/2023] Open
Abstract
Tumor necrosis factor (TNF) ligand related molecule 1A (TL1A), also termed TNF superfamily member 15 and vascular endothelial growth inhibitor is important for tumorigenicity and autoimmunity. However, the function of TL1A in diabetic retinopathy (DR) remains to be elucidated. The present study established a diabetes mellitus (DM) rat model to investigate TL1A, vascular endothelial growth factor (VEGF), tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β) expression levels in the retina, vitreous and serum of rats with DM at different stages (1 month group, 3 month group and 6 month group). The present study determined that TL1A expression levels in the retina and vitreous from the DM 1 month group were significantly lower compared with the control group. However, TL1A levels in the retina and vitreous were significantly increased in advanced stages of DM compared with the control group. Furthermore, the levels of VEGF in the retina and vitreous were significantly higher in the DM groups compared with the control group. The expression levels of TNF-α and IL-1β in the retina and vitreous were significantly higher in DM 3 month and 6 month groups compared with the control group. It is of note that the expression levels of TL1A were significantly lower in the DM 1 and 3 month groups compared with the control group; however, they were significantly increased in the DM 6 month group compared with the DM 3 month group. The expression levels of VEGF, TNF-α and IL-1β in blood serum have been observed to exhibit similar expression change dynamics as those of the retina and vitreous. Therefore, these findings suggest that TL1A may be a protective factor of DR, and may provide a rationale for the development of novel therapeutic strategies to treat DR.
Collapse
Affiliation(s)
- Zhu-Hong Zhang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Qing-Zhong Chen
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Feng Jiang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Todd A Townsend
- Division of Genetic and Molecular Toxicology, US Food and Drug Administration, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | - Chun-Jie Mao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Cai-Yun You
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Wen-Hui Yang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zhi-Yong Sun
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jin-Guo Yu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
23
|
Han S, Liu L, Xu F, Chen S, Yuan W, Fu Z, Li D, Li D. A case-control study about the association between vascular endothelial growth inhibitor gene polymorphisms and breast cancer risk in female patients in Northeast China. Chin J Cancer Res 2016; 28:435-43. [PMID: 27647972 PMCID: PMC5018539 DOI: 10.21147/j.issn.1000-9604.2016.04.07] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Objective The inhibition of the neovascularization in tumors is a potential therapeutic target of cancer. Vascular endothelial growth inhibitor (VEGI) is a member of the TNF superfamily which has the ability to suppress the formation of new vessels in tumors. In order to study the association between VEGI gene polymorphisms and breast cancer risk, a case-control study was conducted in Chinese Han women in Northeast China. Methods Our study involved 708 female breast cancer patients and 685 healthy volunteers. Four SNPs of VEGI gene were analyzed through the polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) method. The association between VEGI gene polymorphisms and breast cancer risk was analyzed in our study. The relation between VEGI gene variants and clinical features of breast cancer including lymph node (LN) metastasis, estrogen receptor (ER), progestrogen receptor (PR), tumor protein 53 (p53), human epidermal growth factor receptor 2 (Her-2) and triple negative (ER-/PR-/Her-2-) status was analyzed as well. Results We found that the CT genotype and T allele of rs6478106 were more frequent in patients than in controls. There was also a statistical difference in the distribution of Crs6478106Grs4263839 haplotype between patients and controls. In addition, SNP rs6478106 and rs4979462 were related with the Her-2 status. Conclusions Our results suggest that VEGI gene variants may be related to the breast cancer risk and the clinical features of breast cancer in Chinese Han women in Northeast China.
Collapse
Affiliation(s)
| | - Lei Liu
- Department of Immunology; College of Bioinformatics Science and Technology
| | | | | | - Weiguang Yuan
- Department of Immunology; Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin 150081, China
| | | | - Dalin Li
- Department of Surgery, the Third Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Dianjun Li
- Department of Immunology; Institute of Cancer Prevention and Treatment, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
24
|
Jiang F, Chen Q, Huang L, Wang Y, Zhang Z, Meng X, Liu Y, Mao C, Zheng F, Zhang J, Yan H. TNFSF15 Inhibits Blood Retinal Barrier Breakdown Induced by Diabetes. Int J Mol Sci 2016; 17:ijms17050615. [PMID: 27120595 PMCID: PMC4881442 DOI: 10.3390/ijms17050615] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Revised: 04/07/2016] [Accepted: 04/11/2016] [Indexed: 12/21/2022] Open
Abstract
Tumor necrosis factor superfamily 15 (TNFSF15) is an endogenous neovascularization inhibitor and an important negative regulator of vascular homeostasis. This study aimed to explore the potential role of TNFSF15 in diabetic retinopathy. Vitreous TNFSF15 and VEGF levels in proliferative diabetic retinopathy (PDR) patients were detected by ELISA. Retinal expression of TNFSF15 and the content of tight junction proteins (TJPs) in rats were detected by immunohistochemistry and Western blot, respectively. The blood retinal barrier (BRB) permeability was evaluated using Evans Blue (EB) dye. The TNFSF15/VEGF ratio was decreased in the vitreous fluid of patients with PDR relative to the controls, even though the expression levels of TNFSF15 were higher. TNFSF15 was dramatically decreased one month later after diabetes induction (p < 0.001), and then increased three months later and thereafter. TNFSF15 treatment significantly protected the BRB in the diabetic animals. Diabetes decreased TJPs levels in the retina, and these changes were inhibited by TNFSF15 treatment. Moreover, TNFSF15 decreased activation of VEGF both in mRNA and protein levels caused by diabetes. These results indicate that TNFSF15 is an important inhibitor in the progression of DR and suggest that the regulation of TNFSF15 shows promise for the development of diabetic retinopathy treatment strategies.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Qingzhong Chen
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Liming Huang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Ying Wang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Zhuhong Zhang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Xiangda Meng
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Yuanyuan Liu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Chunjie Mao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Fang Zheng
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Jingkai Zhang
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
25
|
YU MIAO, LU GUIHUA, ZHU XUN, HUANG ZHIBIN, FENG CHONG, FANG RONG, WANG YESONG, GAO XIUREN. Downregulation of VEGF and upregulation of TL1A expression induce HUVEC apoptosis in response to high glucose stimuli. Mol Med Rep 2016; 13:3265-72. [DOI: 10.3892/mmr.2016.4924] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 12/30/2015] [Indexed: 11/05/2022] Open
|
26
|
Ma W, Shao Y, Yang W, Li G, Zhang Y, Zhang M, Zuo C, Chen K, Wang J. Evaluation of (188)Re-labeled NGR-VEGI protein for radioimaging and radiotherapy in mice bearing human fibrosarcoma HT-1080 xenografts. Tumour Biol 2016; 37:9121-9. [PMID: 26768609 DOI: 10.1007/s13277-016-4810-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/06/2016] [Indexed: 11/25/2022] Open
Abstract
Vascular endothelial growth inhibitor (VEGI) is an anti-angiogenic protein, which includes three isoforms: VEGI-174, VEGI-192, and VEGI-251. The NGR (asparagine-glycine-arginine)-containing peptides can specifically bind to CD13 (Aminopeptidase N) receptor which is overexpressed in angiogenic blood vessels and tumor cells. In this study, a novel NGR-VEGI fusion protein was prepared and labeled with (188)Re for radioimaging and radiotherapy in mice bearing human fibrosarcoma HT-1080 xenografts. Single photon emission computerized tomography (SPECT) imaging results revealed that (188)Re-NGR-VEGI exhibits good tumor-to-background contrast in CD13-positive HT-1080 tumor xenografts. The CD13 specificity of (188)Re-NGR-VEGI was further verified by significant reduction of tumor uptake in HT-1080 tumor xenografts with co-injection of the non-radiolabeled NGR-VEGI protein. The biodistribution results demonstrated good tumor-to-muscle ratio (4.98 ± 0.25) of (188)Re-NGR-VEGI at 24 h, which is consistent with the results from SPECT imaging. For radiotherapy, 18.5 MBq of (188)Re-NGR-VEGI showed excellent tumor inhibition effect in HT-1080 tumor xenografts with no observable toxicity, which was confirmed by the tumor size change and hematoxylin and eosin (H&E) staining of major mouse organs. In conclusion, these data demonstrated that (188)Re-NGR-VEGI has the potential as a theranostic agent for CD13-targeted tumor imaging and therapy.
Collapse
Affiliation(s)
- Wenhui Ma
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC 103, Los Angeles, CA, 90033-9061, USA
| | - Yahui Shao
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
- Department of Nuclear Medicine, General Hospital of Jinan Military Region, Jinan, Shandong, China
| | - Weidong Yang
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Guiyu Li
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Yingqi Zhang
- The State Key Laboratory of Cancer Biology, Department of Biopharmaceutics, School of Pharmacy, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Mingru Zhang
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China
| | - Changjing Zuo
- Department of Nuclear Medicine, Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Kai Chen
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC 103, Los Angeles, CA, 90033-9061, USA.
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, The Fourth Military Medical University, 127 West Changle Road, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
27
|
Sağsöz H, Saruhan BG, Erdoğan S. Functional roles of angiogenic factors and receptors on non-endothelial cells in the oropharyngeal cavity of the chukar partridge (Alectoris chukar). ACTA ZOOL-STOCKHOLM 2015. [DOI: 10.1111/azo.12149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Hakan Sağsöz
- Department of Histology and Embryology; Faculty of Veterinary Medicine; Dicle University; 21280 Diyarbakir Turkey
| | - Berna G. Saruhan
- Department of Histology and Embryology; Faculty of Veterinary Medicine; Dicle University; 21280 Diyarbakir Turkey
| | - Serkan Erdoğan
- Department of Anatomy; Faculty of Veterinary Medicine; Namık Kemal University; 59030 Tekirdağ Turkey
| |
Collapse
|
28
|
Yamanegi K, Kawabe M, Futani H, Nishiura H, Yamada N, Kato-Kogoe N, Kishimoto H, Yoshiya S, Nakasho K. Sodium valproate, a histone deacetylase inhibitor, modulates the vascular endothelial growth inhibitor-mediated cell death in human osteosarcoma and vascular endothelial cells. Int J Oncol 2015; 46:1994-2002. [PMID: 25778932 DOI: 10.3892/ijo.2015.2924] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 02/12/2015] [Indexed: 11/06/2022] Open
Abstract
The level of vascular endothelial growth inhibitor (VEGI) has been reported to be negatively associated with neovascularization in malignant tumors. The soluble form of VEGI is a potent anti-angiogenic factor due to its effects in inhibiting endothelial cell proliferation. This inhibition is mediated by death receptor 3 (DR3), which contains a death domain in its cytoplasmic tail capable of inducing apoptosis that can be subsequently blocked by decoy receptor 3 (DcR3). We investigated the effects of sodium valproate (VPA) and trichostatin A (TSA), histone deacetylase inhibitors, on the expression of VEGI and its related receptors in human osteosarcoma (OS) cell lines and human microvascular endothelial (HMVE) cells. Consequently, treatment with VPA and TSA increased the VEGI and DR3 expression levels without inducing DcR3 production in the OS cell lines. In contrast, the effect on the HMVE cells was limited, with no evidence of growth inhibition or an increase in the DR3 and DcR3 expression. However, VPA-induced soluble VEGI in the OS cell culture medium markedly inhibited the vascular tube formation of HMVE cells, while VEGI overexpression resulted in enhanced OS cell death. Taken together, the HDAC inhibitor has anti-angiogenesis and antitumor activities that mediate soluble VEGI/DR3-induced apoptosis via both autocrine and paracrine pathways. This study indicates that the HDAC inhibitor may be exploited as a therapeutic strategy modulating the soluble VEGI/DR3 pathway in osteosarcoma patients.
Collapse
Affiliation(s)
- Koji Yamanegi
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Mutsuki Kawabe
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiroyuki Futani
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiroshi Nishiura
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Naoko Yamada
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Nahoko Kato-Kogoe
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiromitsu Kishimoto
- Department of Oral and Maxillofacial Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Shinichi Yoshiya
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Keiji Nakasho
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
29
|
Zhang Z, Yu D, Lu J, Zhai K, Cao L, Rao J, Liu Y, Zhang X, Guo Y. Functional genetic variants of TNFSF15 and their association with gastric adenocarcinoma: a case-control study. PLoS One 2014; 9:e108321. [PMID: 25251497 PMCID: PMC4176965 DOI: 10.1371/journal.pone.0108321] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 08/19/2014] [Indexed: 01/26/2023] Open
Abstract
The purpose of this study was to identify functional genetic variants in the promoter of tumor necrosis factor superfamily member 15 (TNFSF15) and evaluate their effects on the risk of developing gastric adenocarcinoma. Forty DNA samples from healthy volunteers were sequenced to identify single nucleotide polymorphisms (SNPs) in the TNFSF15 promoter. Two TNFSF15 SNPs (−358T>C and −638A>G) were identified by direct sequencing. Next, genotypes and haplotypes of 470 gastric adenocarcinoma patients and 470 cancer-free controls were analyzed. Odds ratios (ORs) and 95% confidence intervals (CIs) were estimated by logistic regression. Serologic tests for Helicobacter pylori infection were measured by enzyme-linked immuno-sorbent assay (ELISA). Subjects carrying the TNFSF15 −358CC genotype were at an elevated risk for developing gastric adenocarcinoma, compared with those with the −358TT genotype (OR 1.42, 95% CI, 1.10 to 2.03). H. pylori infection was a risk factor for developing gastric adenocarcinoma (OR 2.31, 95% CI, 1.76 to 3.04). In the H. pylori infected group, subjects with TNFSF15 −358CC genotype were at higher risks for gastric adenocarcinoma compared with those carrying −358TT genotype (OR: 2.01, 95%CI: 1.65 to 4.25), indicating that H. pylori infection further influenced gastric adenocarcinoma susceptibility. The −358 T>C polymorphism eliminates a nuclear factor Y (NF-Y) binding site and the −358C containing haplotypes showed significantly decreased luciferase expression compared with −358T containing haplotypes. Collectively these findings indicate that functional genetic variants in TNFSF15 may play a role in increasing susceptibility to gastric adenocarcinoma.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Chemotherapy and Radiotherapy, Tangshan Gongren Hospital, Tangshan, China
- Institute of Molecular Genetics, College of Life Science, Hebei United University, Tangshan, China
| | - Dianke Yu
- Department of Etiology of Carcinogenesis, Cancer Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie Lu
- Beijing Key Laboratory for Pediatric Otolaryngology, Head and Neck Science, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Kan Zhai
- Department of Etiology of Carcinogenesis, Cancer Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Cao
- Institute of Molecular Genetics, College of Life Science, Hebei United University, Tangshan, China
| | - Juan Rao
- Institute of Molecular Genetics, College of Life Science, Hebei United University, Tangshan, China
| | - Yingwen Liu
- Institute of Molecular Genetics, College of Life Science, Hebei United University, Tangshan, China
| | - Xuemei Zhang
- Institute of Molecular Genetics, College of Life Science, Hebei United University, Tangshan, China
- * E-mail: (YG); (XZ)
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Otolaryngology, Head and Neck Science, Beijing Pediatric Research Institute, Beijing Children’s Hospital, Capital Medical University, Beijing, China
- * E-mail: (YG); (XZ)
| |
Collapse
|
30
|
Ma W, Li G, Wang J, Yang W, Zhang Y, Conti PS, Chen K. In vivo NIRF imaging-guided delivery of a novel NGR-VEGI fusion protein for targeting tumor vasculature. Amino Acids 2014; 46:2721-32. [PMID: 25182731 DOI: 10.1007/s00726-014-1828-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 08/17/2014] [Indexed: 12/11/2022]
Abstract
Pathological angiogenesis is crucial in tumor growth, invasion and metastasis. Previous studies demonstrated that the vascular endothelial growth inhibitor (VEGI), a member of the tumor necrosis factor superfamily, can be used as a potent endogenous inhibitor of tumor angiogenesis. Molecular probes containing the asparagine-glycine-arginine (NGR) sequence can specifically bind to CD13 receptor which is overexpressed on neovasculature and several tumor cells. Near-infrared fluorescence (NIRF) optical imaging for targeting tumor vasculature offers a noninvasive method for early detection of tumor angiogenesis and efficient monitoring of response to anti-tumor vasculature therapy. The aim of this study was to develop a new NIRF imaging probe on the basis of an NGR-VEGI protein for the visualization of tumor vasculature. The NGR-VEGI fusion protein was prepared from prokaryotic expression, and its function was characterized in vitro. The NGR-VEGI protein was then labeled with a Cy5.5 fluorophore to afford Cy5.5-NGR-VEGI probe. Using the NIRF imaging technique, we visualized and quantified the specific delivery of Cy5.5-NGR-VEGI protein to subcutaneous HT-1080 fibrosarcoma tumors in mouse xenografts. The Cy5.5-NGR-VEGI probe exhibited rapid HT-1080 tumor targeting, and highest tumor-to-background contrast at 8 h post-injection (pi). Tumor specificity of Cy5.5-NGR-VEGI was confirmed by effective blocking of tumor uptake in the presence of unlabeled NGR-VEGI (20 mg/kg). Ex vivo NIRF imaging further confirmed in vivo imaging findings, demonstrating that Cy5.5-NGR-VEGI displayed an excellent tumor-to-muscle ratio (18.93 ± 2.88) at 8 h pi for the non-blocking group and significantly reduced ratio (4.92 ± 0.75) for the blocking group. In conclusion, Cy5.5-NGR-VEGI provided highly sensitive, target-specific, and longitudinal imaging of HT-1080 tumors. As a novel theranostic protein, Cy5.5-NGR-VEGI has the potential to improve cancer treatment by targeting tumor vasculature.
Collapse
Affiliation(s)
- Wenhui Ma
- Department of Radiology, Molecular Imaging Center, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC 103, Los Angeles, CA, 90033-9061, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Lu Y, Gu X, Chen L, Yao Z, Song J, Niu X, Xiang R, Cheng T, Qin Z, Deng W, Li LY. Interferon-γ produced by tumor-infiltrating NK cells and CD4+ T cells downregulates TNFSF15 expression in vascular endothelial cells. Angiogenesis 2014; 17:529-40. [PMID: 24141405 DOI: 10.1007/s10456-013-9397-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Accepted: 10/01/2013] [Indexed: 01/19/2023]
Abstract
Endothelial cells in an established vasculature secrete tumor necrosis factor superfamily-15 (TNFSF15; VEGI; TL1A) that functions as a negative modulator of neovascularization to maintain blood vessel stability. TNFSF15 gene expression diminishes at angiogenesis and inflammation sites such as in cancers and wounds. We reported previously that vascular endothelial growth factor and monocyte chemotactic protein-1 contribute to TNFSF15 downmodulation in ovarian cancer. Here we show that interferon-γ (IFNγ) suppresses TNFSF15 expression in human umbilical vein endothelial cells. This activity is mediated by IFNγ receptor and the transcription factor STAT1. Immunohistochemical analysis of ovarian cancer clinical specimens indicates that TNFSF15 expression diminishes while tumor vascularity increases in specimens with high-grades of IFNγ expression. Since tumor-infiltrating NK and CD4(+) T cells are the main sources of IFNγ in tumor lesions, we isolated these cells from peripheral blood of healthy individuals, treated the cells with ovarian cancer OVCAR3 cell-conditioned media, and found a onefold and tenfold increase of IFNγ production in NK and CD4(+) T cells, respectively, compared with that in vehicle-treated cells. These findings support the view that tumor-infiltrating NK and CD4(+) T cells under the influence of cancer cells significantly increase the production of IFNγ, which in turn inhibits TNFSF15 expression in vascular endothelial cells, shifting the balance of pro- and anti-angiogenic factors toward escalated angiogenesis potential in the tumor.
Collapse
Affiliation(s)
- Yi Lu
- College of Pharmacy, State Key Laboratory of Medicinal Chemical Biology and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Gao Y, Ge Z, Zhang Z, Bai Z, Ma X, Wang Y. Vascular endothelial growth inhibitor affects the invasion, apoptosis and vascularisation in breast cancer cell line MDA-MB-231. Chin Med J (Engl) 2014; 127:1947-1953. [PMID: 24824261 DOI: 10.3760/cma.j.issn.0366-6999.20130794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND Breast cancer is one of the most common malignant female diseases worldwide. It is a significant threat to every woman's health. Vascular endothelial growth inhibitor (VEGI) is known to be abundant in endothelial cells. According to previous literature, overexpression of VEGI has been shown to inhibit tumor neovascularisation and progression in cellular and animal models, but there has been limited research on the significance of VEGI in the breast cancer. METHODS In our study, cell lines MDA-MB-231 were first constructed in which VEGI mediated by lentivirus over-expressed. The effects of VEGI over-expression on MDA-MB-231 cells were investigated both in vitro and in vivo. The expression of VEGI in the MDA-MB-231 cells after infection of lentivirus was analyzed using real-time PCR and Western blotting. The effect of the biological characteristics of MDA-MB-231 cells was assessed by growth, invasion, adhesion, and migration assay with subcutaneous tumor-bearing nude mice models. Then the growth curves of the subcutaneous tumors were studied. Expressions of VEGI, CD31 and CD34 in the tumors were analyzed by immunohistochemistry and apoptosis was detected by flow cytometry and immunohistochemistry. RESULTS Infection of MDA-MB-231 cells within the lentivirus resulted in approximately a 1 000-fold increase in the expression of VEGI. As can be seen in the invasion, adhesion and migration assay, the over-expression of VEGI can inhibit the ability of MDA-MB-231 cells during migration, adhesion and invasion. The volume of the subcutaneous tumor in the over-expression group was distinctly and significantly less than that of the control groups. Immunohistochemistry analysis of the tumor biopsies clearly showed the expression of VEGI in the over-expression group increased while CD31 and CD34 decreased significantly. In vitro and in vivo, the early apoptosis rate and the apoptosis index were increased within the VEGI over-expression group as compared with the control group. CONCLUSIONS Taken together, recombinant lentivirus that were successfully constructed, demonstrated up-regulated VEGI gene expression in breast cancer cells. Lentivirus-mediated over-expression of VEGI weakened the ability of the breast cancer cell migration, adhesion and invasion. Over-expression of VEGI diminished the tumorigenic capacity of breast cancer cells in vivo. Up-regulation of VEGI gene expression however inhibited breast cancer MDA-MB-231 cell in the early apoptosis.
Collapse
Affiliation(s)
- Yinguang Gao
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhicheng Ge
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhongtao Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zhigang Bai
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Xuemei Ma
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yu Wang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
33
|
TNFSF15 inhibits vasculogenesis by regulating relative levels of membrane-bound and soluble isoforms of VEGF receptor 1. Proc Natl Acad Sci U S A 2013; 110:13863-8. [PMID: 23918400 DOI: 10.1073/pnas.1304529110] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mouse bone marrow-derived Lin(-)-Sca-1(+) endothelial progenitor cell (EPC) has pluripotent abilities such as supporting neovascularization. Vascular endothelial growth factor (VEGF) receptor 1 (VEGFR1) (Flt1) recognizes various VEGF isoforms and is critically implicated in a wide range of physiological and pathological settings, including vasculogenesis. Mouse EPC expresses two isoforms of VEGFR1: mFlt1, which transmits ligand-induced signals; and sFlt1, which acts as a negative regulator by sequestering ligands of VEGF receptors. How the relative levels of mFlt1 and sFlt1 are regulated is not yet clear. We report here that tumor necrosis factor superfamily 15 (TNFSF15) (also known as VEGI or TL1A), an endothelial cell-secreted cytokine, simultaneously promotes mFlt1 degradation and up-regulates sFlt1 expression in EPC, giving rise to disruption of VEGF- or PlGF-induced activation of eNOS and MAPK p38 and effective inhibition of VEGF-driven, EPC-supported vasculogenesis in a murine Matrigel implant model. TNFSF15 treatment of EPC cultures facilitates Akt deactivation-dependent, ubiquitin-assisted degradation of mFlt1 and stimulates sFlt1 expression by activating the PKC, Src, and Erk1/2 signaling pathway. Additionally, TNFSF15 promotes alternative splicing of the Flt1 gene in favor of sFlt1 production by down-regulating nuclear protein Jumonji domain-containing protein 6 (Jmjd6), thus alleviating Jmjd6-inhibited sFlt1 expression. These findings indicate that TNFSF15 is a key component of a molecular mechanism that negatively modulates EPC-supported vasculogenesis through regulation of the relative levels of mFlt1 and sFlt1 in EPC.
Collapse
|
34
|
Zhang N, Wu P, Shayiremu D, Wu L, Shan H, Ye L, Zhao X, Cai J, Jiang WG, Gong K, Yang Y. Suppression of renal cell carcinoma growth in vivo by forced expression of vascular endothelial growth inhibitor. Int J Oncol 2013; 42:1664-73. [PMID: 23545578 DOI: 10.3892/ijo.2013.1877] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Accepted: 02/20/2013] [Indexed: 11/06/2022] Open
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.
Collapse
Affiliation(s)
- Ning Zhang
- Department of Urology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Ge Z, Sanders AJ, Ye L, Mansel RE, Jiang WG. Expression of death receptor-3 in human breast cancer and its functional effects on breast cancer cells in vitro. Oncol Rep 2013; 29:1356-64. [PMID: 23443464 DOI: 10.3892/or.2013.2259] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 07/16/2012] [Indexed: 11/05/2022] Open
Abstract
Death receptor-3 (DR3) plays controversial roles in cancer. Currently, DR3 is known to be a functional receptor of vascular endothelial growth inhibitor (VEGI). The role of DR3 in breast cancer remains unclear. The present study investigated DR3 expression in a clinical cohort of breast cancer patients and its role in breast cancer cells in vitro. The expression of DR3 was examined in a breast cancer cohort using quantitative PCR (Q-PCR) and immunohistochemistry (IHC) in comparison to the patients' data. In vitro function of DR3 was examined through the targeting of this molecule in MCF7 and MDA-MB-231 breast cancer cells using ribozyme transgene technology. Decreased DR3 expression was noted in breast cancer tissues compared to normal tissues and decreased expression of DR3 was generally associated with a poorer prognosis as well as a significantly shorter long-term survival (p=0.038). Targeting of DR3 in vitro in breast cancer cell lines resulted in impaired migratory rates compared to respective control cells. Collectively, these data suggest a complex role for DR3 in breast cancer development and progression.
Collapse
Affiliation(s)
- Zhicheng Ge
- Metastasis and Angiogenesis Research Group, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | | | | | | | | |
Collapse
|
36
|
TNFSF15 Modulates Neovascularization and Inflammation. CANCER MICROENVIRONMENT 2012; 5:237-47. [PMID: 22833050 DOI: 10.1007/s12307-012-0117-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 07/10/2012] [Indexed: 12/14/2022]
Abstract
Tumor necrosis factor superfamily-15 (TNFSF15; also known as VEGI or TL1A) is a unique cytokine that functions in the modulation of vascular homeostasis and inflammation. TNFSF15 is expressed abundantly in established vasculature but is down-regulated at sites of neovascularization such as in cancers and wounds. TNFSF15 inhibits endothelial cell proliferation and endothelial progenitor cell differentiation. Additionally, TNFSF15 stimulates T cell activation, Th1 cytokine production, and dendritic cell maturation. Some of the functions of TNFSF15 are mediated by death receptor-3. We review the experimental evidences on TNFSF15 activities in angiogenesis, vasculogenesis, inflammation, and immune system mobilization.
Collapse
|
37
|
Advancement in the research on vascular endothelial growth inhibitor (VEGI). Target Oncol 2012; 7:87-90. [DOI: 10.1007/s11523-012-0206-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 01/11/2012] [Indexed: 11/26/2022]
|
38
|
Deng W, Gu X, Lu Y, Gu C, Zheng Y, Zhang Z, Chen L, Yao Z, Li LY. Down-modulation of TNFSF15 in ovarian cancer by VEGF and MCP-1 is a pre-requisite for tumor neovascularization. Angiogenesis 2011; 15:71-85. [PMID: 22210436 DOI: 10.1007/s10456-011-9244-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Accepted: 12/08/2011] [Indexed: 01/18/2023]
Abstract
Persistent inflammation and neovascularization are critical to cancer development. In addition to upregulation of positive control mechanisms such as overexpression of angiogenic and inflammatory factors in the cancer microenvironment, loss of otherwise normally functioning negative control mechanisms is likely to be an important attribute. Insights into the down-modulation of such negative control mechanisms remain largely unclear, however. We show here that tumor necrosis factor superfamily-15 (TNFSF15), an endogenous inhibitor of neovascularization, is a critical component of the negative control mechanism that operates in normal ovary but is missing in ovarian cancer. We show in clinical settings that TNFSF15 is present prominently in the vasculature of normal ovary but diminishes in ovarian cancer as the disease progresses. Vascular endothelial growth factor (VEGF) produced by cancer cells and monocyte chemotactic protein-1 (MCP-1) produced mainly by tumor-infiltrating macrophages and regulatory T cells effectively inhibits TNFSF15 production by endothelial cells in vitro. Using a mouse syngeneic tumor model, we demonstrate that silencing TNFSF15 by topical shRNA treatments prior to and following mouse ovarian cancer ID8 cell inoculation greatly facilitates angiogenesis and tumor growth, whereas systemic application of recombinant TNFSF15 inhibits angiogenesis and tumor growth. Our findings indicate that downregulation of TNFSF15 by cancer cells and tumor infiltrating macrophages and lymphocytes is a pre-requisite for tumor neovascularization.
Collapse
Affiliation(s)
- Weimin Deng
- State Key Laboratory of Medicinal Chemical Biology, Tianjin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zhou J, Yang Z, Tsuji T, Gong J, Xie J, Chen C, Li W, Amar S, Luo Z. LITAF and TNFSF15, two downstream targets of AMPK, exert inhibitory effects on tumor growth. Oncogene 2011; 30:1892-900. [PMID: 21217782 PMCID: PMC3431012 DOI: 10.1038/onc.2010.575] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2010] [Revised: 11/15/2010] [Accepted: 11/16/2010] [Indexed: 12/30/2022]
Abstract
Lipopolysaccharide (LPS)-induced tumor necrosis factor (TNF) α factor (LITAF) is a multiple functional molecule whose sequence is identical to the small integral membrane protein of the lysosome/late endosome. LITAF was initially identified as a transcription factor that activates transcription of proinflammatory cytokine in macrophages in response to LPS. Mutations of the LITAF gene are associated with a genetic disease, called Charcot-Marie-Tooth syndrome. Recently, we have reported that mRNA levels of LITAF and TNF superfamily member 15 (TNFSF15) are upregulated by 5' adenosine monophosphate (AMP)-activated protein kinase (AMPK). The present study further assesses their biological functions. Thus, we show that 5-aminoimidazole-4-carboxamide ribonucleoside (AICAR), a pharmacological activator of AMPK, increases the abundance of LITAF and TNFSF15 in LNCaP and C4-2 prostate cancer cells, which is abrogated by small hairpin RNA (shRNA) or the dominant-negative mutant of AMPK α1 subunit. Our data further demonstrate that AMPK activation upregulates the transcription of LITAF. Intriguingly, silencing LITAF by shRNA enhances proliferation, anchorage-independent growth of these cancer cells and tumor growth in the xenograft model. In addition, our study reveals that LITAF mediates the effect of AMPK by binding to a specific sequence in the promoter region. Furthermore, we show that TNFSF15 remarkably inhibits the growth of prostate cancer cells and bovine aortic endothelial cells in vitro, with a more potent effect toward the latter. In conjuncture, intratumoral injection of TNFSF15 significantly reduces the size of tumors and number of blood vessels and induces changes that are characteristic of tumor cell differentiation. Therefore, our studies for the first time establish the regulatory axis of AMPK-LITAF-TNFSF15 and also suggest that LITAF may function as a tumor suppressor.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Zhanmin Yang
- College of Life Sciences, Shaanxi Normal University, Xi’an, Shaanxi,710062, China
| | - Takanori Tsuji
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Jun Gong
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Jian Xie
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Changyan Chen
- Department of Radiation Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Wande Li
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| | - Salomon Amar
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston, MA
| | - Zhijun Luo
- Department of Biochemistry, Boston University School of Medicine, Boston, MA
| |
Collapse
|
40
|
Sağsöz H, Saruhan BG. The expression of vascular endothelial growth factor and its receptors (flt1/fms, flk1/KDR, flt4) and vascular endothelial growth inhibitor in the bovine uterus during the sexual cycle and their correlation with serum sex steroids. Theriogenology 2011; 75:1720-34. [PMID: 21396695 DOI: 10.1016/j.theriogenology.2011.01.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 01/07/2011] [Accepted: 01/13/2011] [Indexed: 12/13/2022]
Abstract
The present study was conducted to demonstrate of the immunohistochemical localization of vascular endothelial growth factor (VEGF) and its receptors (flt1/fms, flk1/KDR and flt4) as well as vascular endothelial growth inhibitor (VEGI) and to determine the correlation of VEGF and its receptors and VEGI with serum sex steroids (estrogen and progesterone) in the bovine uterus during the sexual cycle. The stage of the estrous cycle in 30 Holstein cattle was assessed based on the gross and histological appearance of the ovaries and uterus and on blood steroid hormone levels. Tissue samples obtained from the uterus were fixed in 10% formaldehyde for routine histological processing. During both follicular and luteal phases, positive cytoplasmic and membrane staining was achieved for VEGF and its receptors (flt1/fms, flk1/KDR and flt4) as well as VEGI in the luminal and glandular epithelial cells, the connective tissue and smooth muscle cells, and the vascular endothelial cells and smooth muscle cells in the uterus. The intensity, proportional and total scores determined for VEGF and its receptors (flt1/fms and flt4) as well as VEGI were greater in the luminal and glandular epithelial cells compared to the connective tissue and smooth muscle cells (P < 0.05). Furthermore, the number and intensity of the flk1/KDR positive cells were greater among the connective tissue cells compared to the luminal and glandular epithelial cells (P < 0.05). As a result, it was determined that the expression of VEGF and its receptors as well as VEGI in the bovine uterus during the follicular and luteal phases varied with different cell types. This suggests that depending on the stage of the sexual cycle, these factors may mediate the establishment of an appropriate environment for the nutritional supply and implantation of the embryo primarily due to the stimulation of angiogenesis but also through the increase in the secretory activity of the epithelial cells in the uterus. Furthermore, this indicates that ovarian steroid hormones play a significant role in regulating the expression of VEGF and its receptors as well as VEGI.
Collapse
Affiliation(s)
- H Sağsöz
- Dicle University, Department of Histology and Embryology, 21280, Diyarbakir-Turkey.
| | | |
Collapse
|
41
|
Liang PH, Tian F, Lu Y, Duan B, Stolz DB, Li LY. Vascular endothelial growth inhibitor (VEGI; TNFSF15) inhibits bone marrow-derived endothelial progenitor cell incorporation into Lewis lung carcinoma tumors. Angiogenesis 2011; 14:61-8. [PMID: 21188501 PMCID: PMC3042043 DOI: 10.1007/s10456-010-9195-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Accepted: 12/08/2010] [Indexed: 12/17/2022]
Abstract
Bone marrow (BM)-derived endothelial progenitor cells (EPC) have a critical role in tumor neovascularization. Vascular endothelial growth inhibitor (VEGI) is a member of the TNF superfamily (TNFSF15). We have shown that recombinant VEGI suppresses tumor angiogenesis by specifically eliminating proliferating endothelial cells (EC). We report here that treatment of tumor bearing mice with recombinant VEGI leads to a significantly decreased population of BM-derived EPC in the tumors. We transplanted whole bone marrow from green fluorescent protein (GFP) transgenic mice into C57BL/6 recipient mice, which were then inoculated with Lewis lung carcinoma (LLC) cells. Intraperitoneal injection of recombinant VEGI led to significant inhibition of tumor growth and decrease of vasculature density compared to vehicle-treated mice. Tumor implantation yielded a decrease of BM-derived EPC in the peripheral blood, while VEGI-treatment resulted in an initial delay of such decrease. Analysis of the whole bone marrow showed a decrease of Lin(-)-c-Kit(+)-Sca-1(+) hematopoietic stem cell (HSC) population in tumor bearing mice; however, VEGI-treatment caused a significant increase of this cell population. In addition, the number of BM-derived EPC in VEGI-treated tumors was notably less than that in the vehicle-treated group, and most of the apoptotic cells in the VEGI-treated tumors were of bone marrow origin. These findings indicate that VEGI inhibits BM-derived EPC mobilization and prevents their incorporation into LLC tumors by inducing apoptosis specifically of BM-derived cells, resulting in the inhibition of EPC-supported tumor vasculogenesis and tumor growth.
Collapse
Affiliation(s)
- Paulina H. Liang
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Fang Tian
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Yi Lu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | - Biyan Duan
- College of Pharmacy, Nankai University, Tianjin, China
| | - Donna B. Stolz
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lu-Yuan Li
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA. University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA. College of Pharmacy, Nankai University, Tianjin, China
| |
Collapse
|
42
|
Ge Z, Sanders AJ, Ye L, Jiang WG. Aberrant expression and function of death receptor-3 and death decoy receptor-3 in human cancer. Exp Ther Med 2011; 2:167-172. [PMID: 22977485 DOI: 10.3892/etm.2011.206] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 01/17/2011] [Indexed: 12/15/2022] Open
Abstract
Death receptor-3 (DR3) and death decoy receptor-3 (DcR3) are both members of the tumour necrosis factor receptor (TNFR) superfamily. The TNFR superfamily contains eight death domain-containing receptors, including TNFR1 (also called DR1), Fas (also called DR2), DR3, DR4, DR5, DR6, NGFR and EDAR. Upon the binding of these receptors with their corresponding ligands, the death domain recruits various proteins that mediate both the death and proliferation of cells. Receptor function is negatively regulated by decoy receptors (DcR1, DcR2, DcR3 and OPG). DR3/DcR3 are a pair of positive and negative players with which vascular endothelial growth inhibitor (VEGI) interacts. VEGI has been suggested to be a potential tumour suppressor. The inhibitory effects of VEGI on cancer are manifested in three main areas: a direct effect on cancer cells, an anti-angiogenic effect on endothelial cells, and the stimulation of dendritic cell maturation. A recent study indicated that DR3 may be a new receptor for E-selectin, which has been reported to be associated with cancer metastasis. DcR3 is a soluble receptor, highly expressed in various tumours, which lacks an apparent transmembrane segment, prevents cytokine response through ligand binding and neutralization, and is an inhibitor of apoptosis. DcR3 serves as a decoy receptor for FasL, LIGHT and VEGI. The cytokine LIGHT activates various anti-tumour functions and is expected to be a promising candidate for cancer therapy. Certain tumours may escape FasL-dependent immune-cytotoxic attack by expressing DcR3, which blocks FasL function. DR3/DcR3 play profound roles in regulating cell death and proliferation in cancer. The present review briefly discusses DR3/DcR3 and attempts to elucidate the role of these negative and positive players in cancer.
Collapse
Affiliation(s)
- Zhicheng Ge
- Metastasis and Angiogenesis Research Group, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | | | | | | |
Collapse
|
43
|
Xiao T, Fan JK, Huang HL, Gu JF, Li LY, Liu XY. VEGI-armed oncolytic adenovirus inhibits tumor neovascularization and directly induces mitochondria-mediated cancer cell apoptosis. Cell Res 2009; 20:367-78. [DOI: 10.1038/cr.2009.126] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
44
|
Therapeutic Potential of VEGI/TL1A in Autoimmunity and Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 647:207-15. [DOI: 10.1007/978-0-387-89520-8_15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
45
|
Identification of low-molecular-weight protein (SCP1) from shark cartilage with anti-angiogenesis activity and sequence similarity to parvalbumin. J Pharm Biomed Anal 2007; 46:563-7. [PMID: 18093782 DOI: 10.1016/j.jpba.2007.10.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2007] [Revised: 10/10/2007] [Accepted: 10/23/2007] [Indexed: 11/20/2022]
Abstract
Cartilage was considered as a possible natural source of anti-angiogenesis compounds due to its known avascular nature. In this study, a low-molecular-weight protein with an anti-angiogenesis activity was isolated from shark cartilage using a mild extraction procedure. The protein was purified to homogeneity by gel filtration and electroelution techniques and its N-terminal amino acid sequence was determined. The purified protein, designated as SCP1, represented a molecular weight of 13.7 kDa, pI of 6.9-7 and its N-terminal sequence revealed sequence similarity to alpha parvalbumin family. The protein inhibited angiogenesis when subjected to microvessel sprouting of collagen-embedded rat aortic ring assay. It is suggested that SCP1 could be considered as a new angiogenesis inhibitor derived from shark cartilage.
Collapse
|
46
|
Conway KP, Price P, Harding KG, Jiang WG. The role of vascular endothelial growth inhibitor in wound healing. Int Wound J 2007; 4:55-64. [PMID: 17425548 PMCID: PMC7951696 DOI: 10.1111/j.1742-481x.2006.00295.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Vascular endothelial growth inhibitor (VEGI) is an endothelial cell-specific cytokine and a potent inhibitor of endothelial cell proliferation and angiogenesis. The role of VEGI in angiogenesis related to tissue repair has previously not been investigated. Biopsies from different wound types were analysed by immunohistochemistry and quantitative polymerase chain reaction for the presence of VEGI protein and transcript, respectively. Human vascular endothelial cell line was transfected with VEGI expression plasmid and tested for their in vitro angiogenesis properties. Immunohistochemical staining for VEGI showed reduced expression in the dermal layer of the acute wounds compared with the chronic wound or normal skin. The ability of VEGI to prevent angiogenesis by in vitro assays showed that VEGI acts as a suppressor to the proliferation and microtubule formation of endothelial cells, and the addition of Hepatocyte Growth Factor had little effect on the ability of cell lines expressing the VEGI gene to increase microtubule formation. The aberrant expression of VEGI in different wound types appears to be linked to the outcome of the healing in these wounds. The altered expression of VEGI in chronic wounds constitutes an important target of future therapies.
Collapse
Affiliation(s)
- Kevin P Conway
- Metastasis and Angiogenesis Research Group, Department of Surgery, Cardiff University, College of Medicine, Cardiff, UK.
| | | | | | | |
Collapse
|