1
|
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
|
2
|
Gu X, Zhu Y, Zhao C, Cao Y, Wang J, Zhang Q, Li L. TNFSF15 facilitates the differentiation of CD11b + myeloid cells into vascular pericytes in tumors. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0245. [PMID: 37921408 PMCID: PMC10690882 DOI: 10.20892/j.issn.2095-3941.2023.0245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/13/2023] [Indexed: 11/04/2023] Open
Abstract
OBJECTIVE Immature vasculature lacking pericyte coverage substantially contributes to tumor growth, drug resistance, and cancer cell dissemination. We previously demonstrated that tumor necrosis factor superfamily 15 (TNFSF15) is a cytokine with important roles in modulating hematopoiesis and vascular homeostasis. The main purpose of this study was to explore whether TNFSF15 might promote freshly isolated myeloid cells to differentiate into CD11b+ cells and further into pericytes. METHODS A model of Lewis lung cancer was established in mice with red fluorescent bone marrow. After TNFSF15 treatment, CD11b+ myeloid cells and vascular pericytes in the tumors, and the co-localization of pericytes and vascular endothelial cells, were assessed. Additionally, CD11b+ cells were isolated from wild-type mice and treated with TNFSF15 to determine the effects on the differentiation of these cells. RESULTS We observed elevated percentages of bone marrow-derived CD11b+ myeloid cells and vascular pericytes in TNFSF15-treated tumors, and the latter cells co-localized with vascular endothelial cells. TNFSF15 protected against CD11b+ cell apoptosis and facilitated the differentiation of these cells into pericytes by down-regulating Wnt3a-VEGFR1 and up-regulating CD49e-FN signaling pathways. CONCLUSIONS TNFSF15 facilitates the production of CD11b+ cells in the bone marrow and promotes the differentiation of these cells into pericytes, which may stabilize the tumor neovasculature.
Collapse
Affiliation(s)
- Xiangxiang Gu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Yipan Zhu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Cancan Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Yixin Cao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Jingying Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Qiangzhe Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| | - Luyuan Li
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, and Haihe Laboratory of Cell Ecosystem, Tianjin 300350, China
| |
Collapse
|
3
|
Zhao CC, Han QJ, Ying HY, Gu XX, Yang N, Li LY, Zhang QZ. TNFSF15 facilitates differentiation and polarization of macrophages toward M1 phenotype to inhibit tumor growth. Oncoimmunology 2022; 11:2032918. [PMID: 35127254 PMCID: PMC8812784 DOI: 10.1080/2162402x.2022.2032918] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Affiliation(s)
- Can-Can Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Qiu-Ju Han
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Hao-Yan Ying
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Xiang-Xiang Gu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Na Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, 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
| | - 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
| |
Collapse
|
4
|
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
|
5
|
Yang GL, Wang S, Zhang S, Liu Y, Liu X, Wang D, Wei H, Xiong J, Zhang ZS, Wang Z, Li LY, Zhang J. A Protective Role of Tumor Necrosis Factor Superfamily-15 in Intracerebral Hemorrhage-Induced Secondary Brain Injury. ASN Neuro 2021; 13:17590914211038441. [PMID: 34596444 PMCID: PMC8642778 DOI: 10.1177/17590914211038441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Destabilization of blood vessels by the activities of vascular endothelial growth factor (VEGF) and matrix metalloproteinases (MMPs) following intracerebral hemorrhage (ICH) has been considered the main causes of aggravated secondary brain injury. Here, we show that tumor necrosis factor superfamily-15 (TNFSF15; also known as vascular endothelial growth inhibitor), an inhibitor of VEGF-induced vascular hyper-permeability, when overexpressed in transgenic mice, exhibits a neuroprotective function post-ICH. In this study, we set-up a collagenase-induced ICH model with TNFSF15-transgenic mice and their transgene-negative littermates. We observed less lesion volume and neural function perturbations, together with less severe secondary injuries in the acute phase that are associated with brain edema and inflammation, including vascular permeability, oxidative stress, microglia/macrophage activation and neutrophil infiltration, and neuron degeneration, in the TNFSF15 group compared with the littermate group. Additionally, we show that there is an inhibition of VEGF-induced elevation of MMP-9 in the perihematomal blood vessels of the TNFSF15 mice following ICH, concomitant with enhanced pericyte coverage of the perihematomal blood vessels. These findings are consistent with the view that TNFSF15 may have a potential as a therapeutic agent for the treatment of secondary injuries in the early phase of ICH.
Collapse
Affiliation(s)
- Gui-Li Yang
- Department of Neurosurgery, 230967Tianjin Medical University General Hospital; Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Shizhao Wang
- 128790North China University of Science and Technology Affiliated Hospital, Tangshan, HeBei Province, China
| | - Shu Zhang
- Department of Neurosurgery, 230967Tianjin Medical University General Hospital; Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Ye Liu
- Department of Neurosurgery, 230967Tianjin Medical University General Hospital; Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Xiao Liu
- Department of Neurosurgery, 230967Tianjin Medical University General Hospital; Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Dong Wang
- Department of Neurosurgery, 230967Tianjin Medical University General Hospital; Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Huijie Wei
- Department of Neurosurgery, 230967Tianjin Medical University General Hospital; Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Jianhua Xiong
- Department of Neurosurgery, 230967Tianjin Medical University General Hospital; Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Zhi-Song Zhang
- State Key Laboratory of Medicinal Chemical Biology, 12538Nankai University College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Tianjin, China
| | - Zengguang Wang
- Department of Neurosurgery, 230967Tianjin Medical University General Hospital; Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| | - Lu-Yuan Li
- State Key Laboratory of Medicinal Chemical Biology, 12538Nankai University College of Pharmacy, Tianjin Key Laboratory of Molecular Drug Research, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, 230967Tianjin Medical University General Hospital; Tianjin Neurological Institute; Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education; Tianjin Key Laboratory of Injuries, Variations and Regeneration of Nervous System, Tianjin, China
| |
Collapse
|
6
|
Zhao H, Zhang Q. Signaling in TNFSF15-mediated Suppression of VEGF Production in Endothelial Cells. Methods Mol Biol 2021; 2248:1-18. [PMID: 33185864 DOI: 10.1007/978-1-0716-1130-2_1] [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] [Indexed: 12/03/2022]
Abstract
Vascular endothelial growth factor (VEGF) plays a pivotal role in promoting neovascularization. Tumor necrosis factor superfamily 15 (TNFSF15) is an antiangiogenic cytokine prominently produced by endothelial cells in a normal vasculature. In this study, Western blot, quantitative polymerase chain reaction (qPCR), and dual luciferase reporter gene assay were used to validate the mechanisms of TNFSF15-mediated suppression of VEGF production in endothelial cells. We report that TNFSF15 inhibits VEGF production via microRNA-29b (miR-29b) targeting the 3'-UTR of VEGF transcript in mouse endothelial cell line bEnd.3. Neutralizing antibody against TNFSF15, 4-3H, inhibits the level of miR-29b and reinvigorates VEGF. In addition, TNFSF15 activates the JNK signaling pathway as well as the transcription factor GATA3, resulting in enhanced miR-29b production. SP600125, an inhibitor of JNK, eradicates TNFSF15-induced GATA3 expression. Moreover, GATA3 siRNA suppressed TNFSF15-induced miR-29b expression. Together, this study provides evidence and method of activation of the JNK-GATA3 signaling pathway by TNFSF15 that suppresses VEGF gene expression, which gives rise to upregulation of miR-29b.
Collapse
Affiliation(s)
- Huanyu Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Qiangzhe Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.
| |
Collapse
|
7
|
Ding Y, Gao S, Shen J, Bai T, Yang M, Xu S, Gao Y, Zhang Z, Li L. TNFSF15 facilitates human umbilical cord blood haematopoietic stem cell expansion by activating Notch signal pathway. J Cell Mol Med 2020; 24:11146-11157. [PMID: 32910534 PMCID: PMC7576288 DOI: 10.1111/jcmm.15626] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 12/24/2022] Open
Abstract
The lack of efficient ex vivo expansion methods restricts clinical use of haematopoietic stem cells (HSC) for the treatment of haematological malignancies and degenerative diseases. Umbilical cord blood (UCB) serves as an alternative haematopoietic stem cell source. However, currently what limits the use of UCB‐derived HSC is the very low numbers of haematopoietic stem and progenitor cells available for transplantation in a single umbilical cord blood unit. Here, we report that TNFSF15, a member of the tumour necrosis factor superfamily, promotes the expansion of human umbilical cord blood (UCB)‐derived HSC. TNFSF15‐treated UCB‐HSC is capable of bone marrow engraftment as demonstrated with NOD/SCID or NOD/Shi‐SCID/IL2Rgnull (NOG) mice in both primary and secondary transplantation. The frequency of repopulating cells occurring in the injected tibiae is markedly higher than that in vehicle‐treated group. Additionally, signal proteins of the Notch pathway are highly up‐regulated in TNFSF15‐treated UCB‐HSC. These findings indicate that TNFSF15 is useful for in vitro expansion of UCB‐HSC for clinical applications. Furthermore, TNFSF15 may be a hopeful selection for further UCB‐HSC application or study.
Collapse
Affiliation(s)
- Yahui Ding
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Shan Gao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Jian Shen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Tairan Bai
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Ming Yang
- Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shiqi Xu
- Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yingdai Gao
- Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Zhisong Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Luyuan Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| |
Collapse
|
8
|
VEGI Improves Outcomes in the Early Phase of Experimental Traumatic Brain Injury. Neuroscience 2020; 438:60-69. [PMID: 32380270 DOI: 10.1016/j.neuroscience.2020.04.042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/23/2020] [Accepted: 04/25/2020] [Indexed: 12/14/2022]
Abstract
Excessive expression of vascular endothelial growth factor (VEGF) is a common cause of blood-brain barrier (BBB) breakdown that triggers severe complications following traumatic brain injury (TBI). It has been shown that inhibition of VEGF activities may attenuate cerebral edema in pathological conditions. Vascular endothelial growth inhibitor (VEGI; also known as TNFSF15), a cytokine produced largely by vascular endothelial cells, is capable of downregulating VEGF expression and inhibiting VEGF receptor-2 (VEGFR2) activation. In this study we found that TBI can cause breakdown of BBB and sharp increases of VEGF/VEGI and Angpt2/Angpt1 ratios in the injured tissues. VEGI treatment resulted in a marked decrease of BBB permeability and concomitant restoration of normal ratios of VEGF/VEGI and Angpt2/Angpt1. Consistently, alleviated edema, decreased neuron cell death, and improved neurological functions were observed when the experimental animals were treated with VEGI in the early phase of TBI. Our findings suggest that administration of VEGI recombinant protein at early phases of TBI is beneficial to stabilization of the disease conditions.
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
|
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
|
11
|
Kumanishi S, Yamanegi K, Nishiura H, Fujihara Y, Kobayashi K, Nakasho K, Futani H, Yoshiya S. Epigenetic modulators hydralazine and sodium valproate act synergistically in VEGI-mediated anti-angiogenesis and VEGF interference in human osteosarcoma and vascular endothelial cells. Int J Oncol 2019; 55:167-178. [PMID: 31180533 DOI: 10.3892/ijo.2019.4811] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 05/14/2019] [Indexed: 11/06/2022] Open
Abstract
Vascular endothelial growth inhibitor (VEGI; also referred to as TNFSF15 or TL1A) is involved in the modulation of vascular homeostasis. VEGI is known to operate via two receptors: Death receptor‑3 (DR3) and decoy receptor‑3 (DcR3). DR3, which is thus far the only known functional receptor for VEGI, contains a death domain and induces cell apoptosis. DcR3 is secreted as a soluble protein and antagonizes VEGI/DR3 interaction. Overexpression of DcR3 and downregulation of VEGI have been detected in a number of cancers. The aim of the present study was to investigate the effects of sodium valproate (VPA), a histone deacetylase inhibitor, in combination with hydralazine hydrochloride (Hy), a DNA methylation inhibitor, on the expression of VEGI and its related receptors in human osteosarcoma (OS) cell lines and human microvascular endothelial (HMVE) cells. Combination treatment with Hy and VPA synergistically induced the expression of VEGI and DR3 in both OS and HMVE cells, without inducing DcR3 secretion. In addition, it was observed that the combination of VPA and Hy significantly enhanced the inhibitory effect on vascular tube formation by VEGI/DR3 autocrine and paracrine pathways. Furthermore, the VEGI/VEGF‑A immune complex was pulled down by immunoprecipitation. Taken together, these findings suggest that DNA methyltransferase and histone deacetylase inhibitors not only have the potential to induce the re‑expression of tumor suppressor genes in cancer cells, but also exert anti‑angiogenic effects, via enhancement of the VEGI/DR3 pathway and VEGI/VEGF‑A interference.
Collapse
Affiliation(s)
- Shunsuke Kumanishi
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Koji Yamanegi
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Hiroshi Nishiura
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Yuki Fujihara
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Kenta Kobayashi
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Keiji Nakasho
- 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
| | - Shinichi Yoshiya
- Department of Orthopedic Surgery, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
12
|
Qiu Y, Chen C, Zhang J, Chen M, Gong H, Gong L, Du L, Wang R. VEGF attenuates lung injury by inducing homing of CD133+ progenitors via VEGFR1. Biochem Biophys Res Commun 2019; 511:650-657. [DOI: 10.1016/j.bbrc.2019.02.071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 02/14/2019] [Indexed: 12/22/2022]
|
13
|
Berendam SJ, Koeppel AF, Godfrey NR, Rouhani SJ, Woods AN, Rodriguez AB, Peske JD, Cummings KL, Turner SD, Engelhard VH. Comparative Transcriptomic Analysis Identifies a Range of Immunologically Related Functional Elaborations of Lymph Node Associated Lymphatic and Blood Endothelial Cells. Front Immunol 2019; 10:816. [PMID: 31057546 PMCID: PMC6478037 DOI: 10.3389/fimmu.2019.00816] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 03/27/2019] [Indexed: 12/11/2022] Open
Abstract
Lymphatic and blood vessels are formed by specialized lymphatic endothelial cells (LEC) and blood endothelial cells (BEC), respectively. These endothelial populations not only form peripheral tissue vessels, but also critical supporting structures in secondary lymphoid organs, particularly the lymph node (LN). Lymph node LEC (LN-LEC) also have been shown to have important immunological functions that are not observed in LEC from tissue lymphatics. LN-LEC can maintain peripheral tolerance through direct presentation of self-antigen via MHC-I, leading to CD8 T cell deletion; and through transfer of self-antigen to dendritic cells for presentation via MHC-II, resulting in CD4 T cell anergy. LN-LEC also can capture and archive foreign antigens, transferring them to dendritic cells for maintenance of memory CD8 T cells. The molecular basis for these functional elaborations in LN-LEC remain largely unexplored, and it is also unclear whether blood endothelial cells in LN (LN-BEC) might express similar enhanced immunologic functionality. Here, we used RNA-Seq to compare the transcriptomic profiles of freshly isolated murine LEC and BEC from LN with one another and with freshly isolated LEC from the periphery (diaphragm). We show that LN-LEC, LN-BEC, and diaphragm LEC (D-LEC) are transcriptionally distinct from one another, demonstrating both lineage and tissue-specific functional specializations. Surprisingly, tissue microenvironment differences in gene expression profiles were more numerous than those determined by endothelial cell lineage specification. In this regard, both LN-localized endothelial cell populations show a variety of functional elaborations that suggest how they may function as antigen presenting cells, and also point to as yet unexplored roles in both positive and negative regulation of innate and adaptive immune responses. The present work has defined in depth gene expression differences that point to functional specializations of endothelial cell populations in different anatomical locations, but especially the LN. Beyond the analyses provided here, these data are a resource for future work to uncover mechanisms of endothelial cell functionality.
Collapse
Affiliation(s)
- Stella J. Berendam
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Alexander F. Koeppel
- Department of Public Health Sciences and Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Nicole R. Godfrey
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Sherin J. Rouhani
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Amber N. Woods
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Anthony B. Rodriguez
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - J. David Peske
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Kara L. Cummings
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Stephen D. Turner
- Department of Public Health Sciences and Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Victor H. Engelhard
- Department of Microbiology, Immunology, and Cancer Biology, Carter Immunology Center, University of Virginia School of Medicine, Charlottesville, VA, United States
- *Correspondence: Victor H. Engelhard
| |
Collapse
|
14
|
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
|
15
|
Counterbalance: modulation of VEGF/VEGFR activities by TNFSF15. Signal Transduct Target Ther 2018; 3:21. [PMID: 30101034 PMCID: PMC6085396 DOI: 10.1038/s41392-018-0023-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/23/2018] [Accepted: 05/31/2018] [Indexed: 01/11/2023] Open
Abstract
Vascular hyperpermeability occurs in angiogenesis and several pathobiological conditions, producing elevated interstitial fluid pressure and lymphangiogenesis. How these closely related events are modulated is a fundamentally important question regarding the maintenance of vascular homeostasis and treatment of disease conditions such as cancer, stroke, and myocardial infarction. Signals mediated by vascular endothelial growth factor receptors, noticeably VEGFR-1, −2, and −3, are centrally involved in the promotion of both blood vessel and lymphatic vessel growth. These signaling pathways are counterbalanced or, in the case of VEGFR3, augmented by signals induced by tumor necrosis factor superfamily-15 (TNFSF15). TNFSF15 can simultaneously downregulate membrane-bound VEGFR1 and upregulate soluble VEGFR1, thus changing VEGF/VEGFR1 signals from pro-angiogenic to anti-angiogenic. In addition, TNFSF15 inhibits VEGF-induced VEGFR2 phosphorylation, thereby curbing VEGFR2-mediated enhancement of vascular permeability. Third, and perhaps more interestingly, TNFSF15 is capable of stimulating VEGFR3 gene expression in lymphatic endothelial cells, thus augmenting VEGF-C/D-VEGFR3-facilitated lymphangiogenesis. We discuss the intertwining relationship between the actions of TNFSF15 and VEGF in this review. The ability of tumor necrosis factor superfamily-15 (TNFSF15) protein to balance the actions of vascular endothelial growth factors (VEGFs) highlights new therapeutic strategies for the treatment of diseases that disrupt the circulatory system. Gui-Li Yang at the Tianjin Neurological Institute and Lu-Yuan Li at Nankai University describe the mechanisms through which TNFSF15 inhibits blood vessel growth mediated by VEGF receptor-1 (VEGFR1) and counterbalances the increase in vascular permeability mediated by VEGFR2. Interestingly, TNFSF15 enhances the effects of VEGFR3 on the formation of lymphatic vessels by promoting VEGFR3 gene expression in lymphatic endothelial cells. Further research will determine whether TNFSF15′s unique capacity to regulate the properties of both blood and lymph vessels can be harnessed to improve the treatment of conditions such as cancer, stroke, myocardial infarction and lymphoedema.
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
|
Danastas K, Whittington CM, Dowland SN, Combes V, Murphy CR, Lindsay LA. Ovarian Hyperstimulation Reduces Vascular Endothelial Growth Factor-A During Uterine Receptivity. Reprod Sci 2018; 26:259-268. [PMID: 29621956 DOI: 10.1177/1933719118768703] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The angiogenic factor vascular endothelial growth factor-A (VEGFA) plays a critical role during early pregnancy in many species including the rat, and any alterations in VEGFA levels can severely impact blastocyst implantation rates. The rat ovarian hyperstimulation (OH) model is useful in studying how the induction of superovulation affects VEGFA levels and endometrial receptivity to blastocyst implantation. The present study shows that the major isoform in the rat uterus, Vegf188, is reduced at the time of receptivity in OH compared to normal pregnancy, whereas there is no change in Vegf164 and Vegf120 messenger RNA (mRNA). The VEGFA receptor 2 (VEGFR2) protein levels are also reduced at the time of receptivity in OH. Our ovariectomy studies show that Vegf164, Vegf188, and Vegf120 are significantly decreased by estrogen, and, to a lesser extent progesterone, when compared to control animals. Although no change in the percentage of endometrial blood vessels was seen across all stages of pregnancy, at the time of receptivity in OH pregnancies, blood vessels were typically larger compared to other stages. The altered progesterone-estrogen ratio seen in OH, taken together with our ovariectomy studies, explains the changes to Vegfa mRNA in OH at the time of receptivity. Since VEGFA is important during implantation, the changes to Vegfa and VEGFR2 levels in the endometrium may help explain the observed lower endometrial receptivity following OH. This study aimed to analyse how ovarian hyperstimulation alters the levels of vascular endothleial growth factor and its major receptor, VEGFR2 in the uterus in a rat model.
Collapse
Affiliation(s)
- Kevin Danastas
- 1 Anatomy and Histology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Camilla M Whittington
- 2 School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales, Australia
| | - Samson N Dowland
- 1 Anatomy and Histology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Valery Combes
- 3 School of Life Sciences, The University of Technology Sydney, Sydney, New South Wales, Australia
| | - Christopher R Murphy
- 1 Anatomy and Histology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | - Laura A Lindsay
- 1 Anatomy and Histology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
18
|
Hahne M, Schumann P, Mursell M, Strehl C, Hoff P, Buttgereit F, Gaber T. Unraveling the role of hypoxia-inducible factor (HIF)-1α and HIF-2α in the adaption process of human microvascular endothelial cells (HMEC-1) to hypoxia: Redundant HIF-dependent regulation of macrophage migration inhibitory factor. Microvasc Res 2018; 116:34-44. [DOI: 10.1016/j.mvr.2017.09.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/12/2017] [Accepted: 09/19/2017] [Indexed: 11/26/2022]
|
19
|
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
|
20
|
Yan Y, Song D, Liu L, Meng X, Qi C, Wang J. The relationship of plasma decoy receptor 3 and coronary collateral circulation in patients with coronary artery disease. Life Sci 2017; 189:84-88. [PMID: 28942284 DOI: 10.1016/j.lfs.2017.09.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 09/07/2017] [Accepted: 09/19/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Previously, decoy receptor 3 (DcR3) was found to be a potential angiogenetic factor, while the relationship of DcR3 with coronary collateral circulation formation has not been investigated. In this study, we aimed to investigate whether plasma decoy receptor 3 levels was associated with CCC formation and evaluate its predictive power for CCC status in patients with coronary artery disease. METHODS Among patients who underwent coronary angiography with coronary artery disease and had a stenosis of ≥90% were included in our study. Collateral degree was graded according to Rentrope Cohen classification. Patients with grade 2 or 3 collateral degree were enrolled in good CCC group and patients with grade 0 or 1 collateral degree were enrolled in poor CCC group. RESULTS Plasma DcR3 level was significantly higher in good CCC group (328.00±230.82 vs 194.84±130.63ng/l, p<0.01) and positively correlated with Rentrope grade (p<0.01). In addition, plasma DcR3 was also positively correlated with VEGF-A. Both ROC (receiver operating characteristic curve) and multinomial logistical regression analysis showed that plasma DcR3 displayed potent predictive power for CCC status. CONCLUSIONS Higher plasma DcR3 level was related to better CCC formation and displayed potent predictive power for CCC status.
Collapse
Affiliation(s)
- Youyou Yan
- Department of Cardiology, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Dandan Song
- Department of Clinical Laboratory, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Lulu Liu
- Department of Cardiology, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Xiuping Meng
- Department of Endodontics, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Chao Qi
- Department of Cardiology, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun 130041, China
| | - Junnan Wang
- Department of Cardiology, The Second Hospital, Jilin University, No. 218 Ziqiang Street, Changchun 130041, China.
| |
Collapse
|
21
|
Shi X, Zhang W, Yin L, Chilian WM, Krieger J, Zhang P. Vascular precursor cells in tissue injury repair. Transl Res 2017; 184:77-100. [PMID: 28284670 PMCID: PMC5429880 DOI: 10.1016/j.trsl.2017.02.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/25/2016] [Accepted: 02/14/2017] [Indexed: 12/22/2022]
Abstract
Vascular precursor cells include stem cells and progenitor cells giving rise to all mature cell types in the wall of blood vessels. When tissue injury occurs, local hypoxia and inflammation result in the generation of vasculogenic mediators which orchestrate migration of vascular precursor cells from their niche environment to the site of tissue injury. The intricate crosstalk among signaling pathways coordinates vascular precursor cell proliferation and differentiation during neovascularization. Establishment of normal blood perfusion plays an essential role in the effective repair of the injured tissue. In recent years, studies on molecular mechanisms underlying the regulation of vascular precursor cell function have achieved substantial progress, which promotes exploration of vascular precursor cell-based approaches to treat chronic wounds and ischemic diseases in vital organ systems. Verification of safety and establishment of specific guidelines for the clinical application of vascular precursor cell-based therapy remain major challenges in the field.
Collapse
Affiliation(s)
- Xin Shi
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Weihong Zhang
- Department of Basic Medicine, School of Nursing, Zhengzhou University, Zhengzhou, Henan Province, PR China
| | - Liya Yin
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - William M Chilian
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Jessica Krieger
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio
| | - Ping Zhang
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, Ohio.
| |
Collapse
|
22
|
Kwok J, O'Shea M, Hume DA, Lengeling A. Jmjd6, a JmjC Dioxygenase with Many Interaction Partners and Pleiotropic Functions. Front Genet 2017; 8:32. [PMID: 28360925 PMCID: PMC5352680 DOI: 10.3389/fgene.2017.00032] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/27/2017] [Indexed: 12/20/2022] Open
Abstract
Lysyl hydroxylation and arginyl demethylation are post-translational events that are important for many cellular processes. The jumonji domain containing protein 6 (JMJD6) has been reported to catalyze both lysyl hydroxylation and arginyl demethylation on diverse protein substrates. It also interacts directly with RNA. This review summarizes knowledge of JMJD6 functions that have emerged in the last 15 years and considers how a single Jumonji C (JmjC) domain-containing enzyme can target so many different substrates. New links and synergies between the three main proposed functions of Jmjd6 in histone demethylation, promoter proximal pause release of polymerase II and RNA splicing are discussed. The physiological context of the described molecular functions is considered and recently described novel roles for JMJD6 in cancer and immune biology are reviewed. The increased knowledge of JMJD6 functions has wider implications for our general understanding of the JmjC protein family of which JMJD6 is a member.
Collapse
Affiliation(s)
- Janice Kwok
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Edinburgh, UK
| | - Marie O'Shea
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Edinburgh, UK
| | - David A Hume
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Edinburgh, UK
| | - Andreas Lengeling
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Edinburgh, UK
| |
Collapse
|
23
|
Yang GL, Zhao Z, Qin TT, Wang D, Chen L, Xiang R, Xi Z, Jiang R, Zhang ZS, Zhang J, Li LY. TNFSF15 inhibits VEGF-stimulated vascular hyperpermeability by inducing VEGFR2 dephosphorylation. FASEB J 2017; 31:2001-2012. [PMID: 28183800 DOI: 10.1096/fj.201600800r] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 01/17/2017] [Indexed: 02/05/2023]
Abstract
Vascular hyperpermeability is critical in ischemic diseases, including stroke and myocardial infarction, as well as in inflammation and cancer. It is well known that the VEGF-VEGFR2 signaling pathways are pivotal in promoting vascular permeability; however, counterbalancing mechanisms that restrict vascular permeability to maintain the integrity of blood vessels are not yet fully understood. We report that TNF superfamily member 15 (TNFSF15), a cytokine largely produced by vascular endothelial cells and a specific inhibitor of the proliferation of these same cells, can inhibit VEGF-induced vascular permeability in vitro and in vivo, and that death receptor 3 (DR3), a cell surface receptor of TNFSF15, mediates TNFSF15-induced dephosphorylation of VEGFR2. Src homology region 2 domain-containing phosphatase-1 (SHP-1) becomes associated with DR3 upon TNFSF15 interaction with the latter. In addition, a protein complex consisting of VEGFR2, DR3, and SHP-1 is formed in response to the effects of TNFSF15 and VEGF on endothelial cells. It is plausible that this protein complex provides a structural basis for the molecular mechanism in which TNFSF15 induces the inhibition of VEGF-stimulated vascular hyperpermeability.-Yang, G.-L., Zhao, Z., Qin, T.-T., Wang, D., Chen, L., Xiang, R., Xi, Z., Jiang, R., Zhang, Z.-S., Zhang, J., Li. L.-Y. TNFSF15 inhibits VEGF-stimulated vascular hyperpermeability by inducing VEGFR2 dephosphorylation.
Collapse
Affiliation(s)
- Gui-Li Yang
- Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Collaborative Innovation Center for Biotherapy and Tianjin Key Laboratory of Molecular Drug Research.,Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and
| | - Zilong Zhao
- Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and
| | - Ting-Ting Qin
- Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Collaborative Innovation Center for Biotherapy and Tianjin Key Laboratory of Molecular Drug Research
| | - Dong Wang
- Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and
| | - Lijuan Chen
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Rong Xiang
- Department of Immunology, Medical School of Nankai University, and
| | - Zhen Xi
- Department of Chemical Biology, College of Chemistry, Nankai University, Tianjin, China
| | - Rongcai Jiang
- Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and
| | - Zhi-Song Zhang
- Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Collaborative Innovation Center for Biotherapy and Tianjin Key Laboratory of Molecular Drug Research,
| | - Jianning Zhang
- Key Laboratory of Post-Neuroinjury Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; and
| | - Lu-Yuan Li
- Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Collaborative Innovation Center for Biotherapy and Tianjin Key Laboratory of Molecular Drug Research,
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
Janelidze S, Hertze J, Nägga K, Nilsson K, Nilsson C, Wennström M, van Westen D, Blennow K, Zetterberg H, Hansson O. Increased blood-brain barrier permeability is associated with dementia and diabetes but not amyloid pathology or APOE genotype. Neurobiol Aging 2016; 51:104-112. [PMID: 28061383 PMCID: PMC5754327 DOI: 10.1016/j.neurobiolaging.2016.11.017] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/07/2016] [Accepted: 11/25/2016] [Indexed: 01/09/2023]
Abstract
Blood-brain barrier (BBB) dysfunction might be an important component of many neurodegenerative disorders. In this study, we investigated its role in dementia using large clinical cohorts. The cerebrospinal fluid (CSF)/plasma albumin ratio (Qalb), an indicator of BBB (and blood-CSF barrier) permeability, was measured in a total of 1015 individuals. The ratio was increased in patients with Alzheimer's disease, dementia with Lewy bodies or Parkinson's disease dementia, subcortical vascular dementia, and frontotemporal dementia compared with controls. However, this measure was not changed during preclinical or prodromal Alzheimer's disease and was not associated with amyloid positron emission tomography or APOE genotype. The Qalb was increased in diabetes mellitus and correlated positively with CSF biomarkers of angiogenesis and endothelial dysfunction (vascular endothelial growth factor, intracellular adhesion molecule 1, and vascular cell adhesion molecule 1). In healthy elderly, high body mass index and waist-hip ratio predicted increased Qalb 20 years later. In summary, BBB permeability is increased in major dementia disorders but does not relate to amyloid pathology or APOE genotype. Instead, BBB impairment may be associated with diabetes and brain microvascular damage.
Collapse
Affiliation(s)
- Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden
| | - Joakim Hertze
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Katarina Nägga
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Karin Nilsson
- Memory Clinic, Skåne University Hospital, Malmö, Sweden
| | - Christer Nilsson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Department of Neurology, Skåne University Hospital, Lund, Sweden
| | | | - Malin Wennström
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Wallenberg Laboratory, Malmö, Sweden
| | - Danielle van Westen
- Department of Clinical Sciences, Diagnostic radiology, Lund University, Lund, Sweden; Imaging and Function, Skåne University Health Care, Lund, Sweden
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Molecular Neuroscience, UCL Institute of Neurology, Queen Square, London, UK
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences Malmö, Lund University, Lund, Sweden; Memory Clinic, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
27
|
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
|
28
|
Kanki K, Ii M, Terai Y, Ohmichi M, Asahi M. Bone Marrow-Derived Endothelial Progenitor Cells Reduce Recurrent Miscarriage in Gestation. Cell Transplant 2016; 25:2187-2197. [PMID: 27513361 DOI: 10.3727/096368916x692753] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bone marrow-derived endothelial progenitor cells (EPCs) have been shown to contribute to not only angiogenesis in ischemic tissue but also neovascularization in uterine endometrium formation. Reduced neovascularization and elevation of serum soluble Flt1, a functional blockage of VEGF, in the development of placenta is thought to be one of the major causes of repeated miscarriages in gestation. We then examined whether transfusion of VEGF-expressing extrinsic EPCs prevented frequent miscarriage via its promotional effect on neovascularization with a VEGFeNOS signaling pathway in a mouse miscarriage model. The results showed that systemic EPC transfusion significantly reduced the rate of miscarriage, and EPCs were frequently observed in the miscarriage placenta. In contrast, only a few EPCs were detected in the placenta of normal gestation. The vascular pattern was irregular, and vessel size was small in the miscarriage placenta compared with that of normal gestation. The placental vascular pattern in miscarriage tended to be normalized with increased vessel size up to a similar level as normal gestation by EPC recruitment. For the mechanistic insight, since soluble Flt1 inhibits EPC functions, it was suggested that the increased soluble Flt1 could suppress the recruited EPC functional activity in the miscarriage placenta. In vitro experiments by soluble Flt1 treatment in cultured EPCs suggested that the vascular abnormality could be partly due to the inhibition of eNOS expression by the increased amounts of soluble Flt1. These findings from animal experiments indicated that autologous EPC therapy may be a novel therapy to prevent miscarriage in high-risk pregnancies, such as preeclampsia.
Collapse
|
29
|
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
|
30
|
Aprelikova O, Chen K, El Touny LH, Brignatz-Guittard C, Han J, Qiu T, Yang HH, Lee MP, Zhu M, Green JE. The epigenetic modifier JMJD6 is amplified in mammary tumors and cooperates with c-Myc to enhance cellular transformation, tumor progression, and metastasis. Clin Epigenetics 2016; 8:38. [PMID: 27081402 PMCID: PMC4831179 DOI: 10.1186/s13148-016-0205-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 04/06/2016] [Indexed: 01/23/2023] Open
Abstract
Background Oncogene overexpression in primary cells often triggers the induction of a cellular safeguard response promoting senescence or apoptosis. Secondary cooperating genetic events are generally required for oncogene-induced tumorigenesis to overcome these biologic obstacles. We employed comparative genomic hybridization for eight genetically engineered mouse models of mammary cancer to identify loci that might harbor genes that enhance oncogene-induced tumorigenesis. Results Unlike many other mammary tumor models, the MMTV-Myc tumors displayed few copy number variants except for amplification of distal mouse chromosome 11 in 80 % of the tumors (syntenic to human 17q23-qter often amplified in human breast cancer). Analyses of candidate genes located in this region identified JMJD6 as an epigenetic regulatory gene that cooperates with Myc to enhance tumorigenesis. It suppresses Myc-induced apoptosis under varying stress conditions through inhibition of p19ARF messenger RNA (mRNA) and protein, leading to reduced levels of p53. JMJD6 binds to the p19ARF promoter and exerts its inhibitory function through demethylation of H4R3me2a. JMJD6 overexpression in MMTV-Myc cell lines increases tumor burden, induces EMT, and greatly enhances tumor metastasis. Importantly, we demonstrate that co-expression of high levels of JMJD6 and Myc is associated with poor prognosis for human ER+ breast cancer patients. Conclusions A novel epigenetic mechanism has been identified for how JMJD6 cooperates with Myc during oncogenic transformation. Combined high expression of Myc and JMJD6 confers a more aggressive phenotype in mouse and human tumors. Given the pleiotropic pro-tumorigenic activities of JMJD6, it may be useful as a prognostic factor and a therapeutic target for Myc-driven mammary tumorigenesis. Electronic supplementary material The online version of this article (doi:10.1186/s13148-016-0205-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Olga Aprelikova
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Building 37, Room 4054, 37 Convent Dr., Bethesda, MD 20892 USA
| | - Kenny Chen
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Building 37, Room 4054, 37 Convent Dr., Bethesda, MD 20892 USA
| | - Lara H El Touny
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Building 37, Room 4054, 37 Convent Dr., Bethesda, MD 20892 USA
| | - Constance Brignatz-Guittard
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Building 37, Room 4054, 37 Convent Dr., Bethesda, MD 20892 USA
| | - Justin Han
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Building 37, Room 4054, 37 Convent Dr., Bethesda, MD 20892 USA
| | - Tinghu Qiu
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Building 37, Room 4054, 37 Convent Dr., Bethesda, MD 20892 USA
| | - Howard H Yang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Building 37, Room 4054, 37 Convent Dr., Bethesda, MD 20892 USA
| | - Maxwell P Lee
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Building 37, Room 4054, 37 Convent Dr., Bethesda, MD 20892 USA
| | - Min Zhu
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Building 37, Room 4054, 37 Convent Dr., Bethesda, MD 20892 USA
| | - Jeffrey E Green
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Building 37, Room 4054, 37 Convent Dr., Bethesda, MD 20892 USA
| |
Collapse
|
31
|
Li M, Jia Q, Chen T, Zhao Z, Chen J, Zhang J. The role of vascular endothelial growth factor and vascular endothelial growth inhibitor in clinical outcome of traumatic brain injury. Clin Neurol Neurosurg 2016; 144:7-13. [PMID: 26945876 DOI: 10.1016/j.clineuro.2016.02.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 02/16/2016] [Accepted: 02/23/2016] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Tumor necrosis factor superfamily-15 (TNFSF15) also known as vascular endothelial growth inhibitor (VEGI) is a cytokine that modulates anti-angiogenesis and inflammation. Vascular endothelial growth factor (VEGF) promotes angiogenesis and vascular permeability following traumatic brain injury (TBI). The balance of VEGF and VEGI may play a key role in the maintenance of vascular and immune system homeostasis in the brain. However, the dynamic changes of circulating VEGF and VEGI after traumatic brain injury (TBI) and the correlation between plasma VEGF and plasma VEGI remains obscure. In this study, we were to investigate whether circulating VEGF and VEGI can be used as prognostic markers for patients with TBI. PATIENTS AND METHODS A prospective clinical study was conducted in two neurosurgical intensive care units of Tianjin Medical University General Hospital and Tianjin Huanhu Hospital (Tianjin, China). 40 patients and 30 healthy controls were recruited. The recruited subjects were aged over 18 with randomized gender and GCS. 1mL of blood was withdrawn on 1, 4, 7, 14, and 21days after TBI. Blood samples were centrifuged at 3000rpm and the supernatants were used to measure VEGF and VEGI by ELISA kit. RESULTS 1) Circulating VEGF in TBI patients was decreased on the 1st day after TBI, then climbed up on the 4th day, reaching a maximum level on the14th day after TBI, as compared to normal controls. VEGF level returned to normal level on 21th day after TBI. 2) Circulating VEGI in TBI patients was decreased on the 1st and 4th day after TBI, then climbed up on the 7th day after TBI, reaching a maximum level on 14th day after TBI, as compared to normal controls. VEGI levels declined to normal level on 21th day after TBI. 3) There was a significant positive correlation between circulating VEGF and VEGI. 4) However, TBI patients whose conditions had improved exhibited lower VEGF levels 7days after TBI when compared to TBI patients whose condition had deteriorated. Survivors exhibited higher VEGI levels 7days after TBI when compared to non-survivors. 5)TBI patients whose condition had improved exhibited higher VEGI levels when compared to TBI patients whose condition had deteriorated 21days after TBI. Patients with mild TBI exhibited higher VEGI levels than those with moderate and severe TBI 21days after TBI. 6) A lower rate of recovery and higher hospital mortality were found in patients with VEGF/VEGI ratio≥2.366 as compared to those with VEGF/VEGI ratio<2.366 7days after TBI. CONCLUSIONS 1) VEGF level positively correlates with VEGI after TBI. 2) The elevation of VEGF exhibits an adverse effect from 4 to 14days after TBI while it has an advantageous effect from 14 to 21days after TBI. Increasing VEGI levels are beneficial in recovery after TBI. Controlling the ratio of VEGF/VEGI may benefit the clinical outcome following TBI.
Collapse
Affiliation(s)
- Min Li
- Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Qiang Jia
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin 300060, PR China
| | - Tongheng Chen
- Department of Neurosurgery, Second Hospital of Tianjin Medical University, Tianjin 300211, PR China
| | - Zilong Zhao
- Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit MI 48202, USA
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, PR China.
| |
Collapse
|
32
|
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
|
33
|
Flamini V, Jiang WG, Lane J, Cui YX. Significance and therapeutic implications of endothelial progenitor cells in angiogenic-mediated tumour metastasis. Crit Rev Oncol Hematol 2016; 100:177-89. [PMID: 26917455 DOI: 10.1016/j.critrevonc.2016.02.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 11/30/2015] [Accepted: 02/15/2016] [Indexed: 01/16/2023] Open
Abstract
Cancer conveys profound social and economic consequences throughout the world. Metastasis is responsible for approximately 90% of cancer-associated mortality and, when it occurs, cancer becomes almost incurable. During metastatic dissemination, cancer cells pass through a series of complex steps including the establishment of tumour-associated angiogenesis. The human endothelial progenitor cells (hEPCs) are a cell population derived from the bone marrow which are required for endothelial tubulogenesis and neovascularization. They also express abundant inflammatory cytokines and paracrine angiogenic factors. Clinically hEPCs are highly correlated with relapse, disease progression, metastasis and treatment response in malignancies such as breast cancer, ovarian cancer and non-small-cell lung carcinoma. It has become evident that the hEPCs are involved in the angiogenesis-required progression and metastasis of tumours. However, it is not clear in what way the signalling pathways, controlling the normal cellular function of human BM-derived EPCs, are hijacked by aggressive tumour cells to facilitate tumour metastasis. In addition, the actual roles of hEPCs in tumour angiogenesis-mediated metastasis are not well characterised. In this paper we reviewed the clinical relevance of the hEPCs with cancer diagnosis, progression and prognosis. We further summarised the effects of tumour microenvironment on the hEPCs and underlying mechanisms. We also hypothesized the roles of altered hEPCs in tumour angiogenesis and metastasis. We hope this review may enhance our understanding of the interaction between hEPCs and tumour cells thus aiding the development of cellular-targeted anti-tumour therapies.
Collapse
Affiliation(s)
- Valentina Flamini
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, UK
| | - Wen G Jiang
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, UK
| | - Jane Lane
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, UK
| | - Yu-Xin Cui
- Cardiff China Medical Research Collaborative, School of Medicine, Cardiff University, UK.
| |
Collapse
|
34
|
Janelidze S, Lindqvist D, Francardo V, Hall S, Zetterberg H, Blennow K, Adler CH, Beach TG, Serrano GE, van Westen D, Londos E, Cenci MA, Hansson O. Increased CSF biomarkers of angiogenesis in Parkinson disease. Neurology 2015; 85:1834-42. [PMID: 26511451 PMCID: PMC4662706 DOI: 10.1212/wnl.0000000000002151] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 07/09/2015] [Indexed: 12/29/2022] Open
Abstract
Objective: To study biomarkers of angiogenesis in Parkinson disease (PD), and how these are associated with clinical characteristics, blood–brain barrier (BBB) permeability, and cerebrovascular disease. Methods: In this cross-sectional analysis, 38 elderly controls and 100 patients with PD (82 without dementia and 18 with dementia) were included from the prospective Swedish BioFinder study. CSF samples were analyzed for the angiogenesis biomarkers vascular endothelial growth factor (VEGF); its receptors, VEGFR-1 and VEGFR-2; placental growth factor (PlGF); angiopoietin 2 (Ang2); and interleukin-8. BBB permeability, white matter lesions (WMLs), and cerebral microbleeds (CMB) were assessed. CSF angiogenesis biomarkers were also measured in 2 validation cohorts: (1) 64 controls and 87 patients with PD with dementia; and (2) 35 controls and 93 patients with neuropathologically confirmed diagnosis of PD with and without dementia. Results: Patients with PD without dementia displayed higher CSF levels of VEGF, PlGF, and sVEGFR-2, and lower levels of Ang2, compared to controls. Similar alterations in VEGF, PlGF, and Ang2 levels were observed in patients with PD with dementia. Angiogenesis markers were associated with gait difficulties and orthostatic hypotension as well as with more pronounced BBB permeability, WMLs, and CMB. Moreover, higher levels of VEGF and PlGF levels were associated with increased CSF levels of neurofilament light (a marker of neurodegeneration) and monocyte chemotactic protein–1 (a marker of glial activation). The main results were validated in the 2 additional cohorts. Conclusions: CSF biomarkers of angiogenesis are increased in PD, and they are associated with gait difficulties, BBB dysfunction, WMLs, and CMB. Abnormal angiogenesis may be important in PD pathogenesis and contribute to dopa-resistant symptoms.
Collapse
Affiliation(s)
- Shorena Janelidze
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - Daniel Lindqvist
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden.
| | - Veronica Francardo
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - Sara Hall
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - Henrik Zetterberg
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - Kaj Blennow
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - Charles H Adler
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - Thomas G Beach
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - Geidy E Serrano
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - Danielle van Westen
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - Elisabet Londos
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - M Angela Cenci
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| | - Oskar Hansson
- From the Clinical Memory Research Unit, Department of Clinical Sciences (S.J., E.L., O.H.), Lund University, Malmö; the Department of Clinical Sciences (D.L., S.H.), Division of Psychiatry (D.L.), Department of Experimental Medical Science (V.F., M.A.C.), and Clinical Sciences, Diagnostic Radiology (D.v.W.), Lund University, Lund; Psychiatry Skåne (D.L.), Lund; the Department of Neurology (S.H.) and Memory Clinic (E.L., O.H.), Skåne University Hospital, Lund; Institute of Neuroscience and Physiology (H.Z., K.B.), Department of Psychiatry and Neurochemistry, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; UCL Institute of Neurology (H.Z.), London, UK; The Torsten Söderberg Professorship in Medicine at the Royal Swedish Academy of Sciences (K.B.), Stockholm, Sweden; Department of Neurology (C.H.A.), Mayo Clinic, Scottsdale; Banner Sun Health Research Institute (T.G.B., G.E.S.), Sun City, AZ; and Imaging and Function (D.v.W.), Skåne University Health Care, Lund, Sweden
| |
Collapse
|
35
|
Broggini T, Wüstner M, Harms C, Stange L, Blaes J, Thomé C, Harms U, Mueller S, Weiler M, Wick W, Vajkoczy P, Czabanka M. NDRG1 overexpressing gliomas are characterized by reduced tumor vascularization and resistance to antiangiogenic treatment. Cancer Lett 2015; 380:568-576. [PMID: 26297987 DOI: 10.1016/j.canlet.2015.06.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 06/07/2015] [Accepted: 06/19/2015] [Indexed: 12/21/2022]
Abstract
Hypoxia-regulated molecules play an important role in vascular resistance to antiangiogenic treatment. N-myc downstream-regulated-gene 1 (NDRG1) is significantly upregulated during hypoxia in glioma. It was the aim of the present study to analyze the role of NDRG1 on glioma angiogenesis and on antiangiogenic treatment. Orthotopically implanted NDRG1 glioma showed reduced tumor growth and vessel density compared to controls. RT-PCR gene array analysis revealed a 30-fold TNFSF15 increase in NDRG1 tumors. Consequently, the supernatant from NDRG1 transfected U87MG glioma cells resulted in reduced HUVEC proliferation, migration and angiogenic response in tube formation assays in vitro. This effect was provoked by increased TNFSF15 promoter activity in NDRG1 cells. Mutations in NF-κB and AP-1 promoter response elements suppressed TNFSF15 promoter activity. Moreover, U87MG glioma NDRG1 knockdown supernatant contained multiple proangiogenic proteins and increased HUVEC spheroid sprouting. Sunitinib treatment of orhotopically implanted mice reduced tumor volume and vessel density in controls; in NDRG1 overexpressing cells no reduction of tumor volume or vessel density was observed. NDRG1 overexpression leads to reduced tumor growth and angiogenesis in experimental glioma via upregulation of TNFSF15. In NDRG1 overexpressing glioma antiangiogenic treatment does not yield a therapeutic response.
Collapse
Affiliation(s)
- Thomas Broggini
- Department of Neurosurgery, Neurochirurgische Klinik - Universitätsmedizin Charite, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Marie Wüstner
- Department of Neurosurgery, Neurochirurgische Klinik - Universitätsmedizin Charite, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology, Universitätsmedizin Charite, Berlin, Germany
| | - Lena Stange
- Department of Neurosurgery, Neurochirurgische Klinik - Universitätsmedizin Charite, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Jonas Blaes
- Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), University of Heidelberg and German Cancer Consortium (DKTK), Germany
| | - Carina Thomé
- Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), University of Heidelberg and German Cancer Consortium (DKTK), Germany
| | - Ulrike Harms
- Department of Neurology, Universitätsmedizin Charite, Berlin, Germany
| | - Susanne Mueller
- Department of Neurology, Universitätsmedizin Charite, Berlin, Germany
| | - Markus Weiler
- Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), University of Heidelberg and German Cancer Consortium (DKTK), Germany
| | - Wolfgang Wick
- Department of Neurooncology, Neurology Clinic and National Center for Tumor Diseases, Clinical Cooperation Unit Neurooncology, German Cancer Research Center (DKFZ), University of Heidelberg and German Cancer Consortium (DKTK), Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Neurochirurgische Klinik - Universitätsmedizin Charite, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Marcus Czabanka
- Department of Neurosurgery, Neurochirurgische Klinik - Universitätsmedizin Charite, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
36
|
Qin TT, Xu GC, Qi JW, Yang GL, Zhang K, Liu HL, Xu LX, Xiang R, Xiao G, Cao H, Wei Y, Zhang QZ, Li LY. Tumour necrosis factor superfamily member 15 (Tnfsf15) facilitates lymphangiogenesis via up-regulation of Vegfr3
gene expression in lymphatic endothelial cells. J Pathol 2015; 237:307-18. [PMID: 26096340 DOI: 10.1002/path.4577] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 06/13/2015] [Accepted: 06/17/2015] [Indexed: 02/06/2023]
Affiliation(s)
- Ting-Ting Qin
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin 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; Tianjin China
| | - Jian-Wei Qi
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Tianjin China
| | - Gui-Li Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
| | - Kun Zhang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin 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; Tianjin China
| | - Li-Xia Xu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research; Nankai University; Tianjin China
| | - Rong Xiang
- School of Medicine; Nankai University; Tianjin China
- Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital; Sichuan University; China
| | - Guozhi Xiao
- Department of Biology; South University of Science and Technology of China; Shenzhen China
| | - Huiling Cao
- Department of Biology; South University of Science and Technology of China; Shenzhen China
| | - Yuquan Wei
- Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital; Sichuan University; China
- State Key Laboratory of Biotherapy, 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; Tianjin China
- Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital; Sichuan University; 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; Tianjin China
- Collaborative Innovation Center for Biotherapy, Nankai University, West China Hospital; Sichuan University; China
| |
Collapse
|
37
|
Böttger A, Islam MS, Chowdhury R, Schofield CJ, Wolf A. The oxygenase Jmjd6--a case study in conflicting assignments. Biochem J 2015; 468:191-202. [PMID: 25997831 DOI: 10.1042/bj20150278] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The Jumonji domain-containing protein 6 (Jmjd6) is a member of the superfamily of non-haem iron(II) and 2-oxoglutarate (2OG)-dependent oxygenases; it plays an important developmental role in higher animals. Jmjd6 was initially assigned a role as the phosphatidylserine receptor responsible for engulfment of apoptotic cells but this now seems unlikely. Jmjd6 has been shown to be a nuclear localized protein with a JmjC domain comprising a distorted double-stranded β-helical structure characteristic of the 2OG-dependent oxygenases. Jmjd6 was subsequently assigned a role in catalysing N-methyl-arginine residue demethylation on the N-terminus of the human histones H3 and H4; however, this function is also subject to conflicting reports. Jmjd6 does catalyse 2OG-dependent C-5 hydroxylation of lysine residues in mRNA splicing-regulatory proteins and histones; there is also accumulating evidence that Jmjd6 plays a role in splicing (potentially in an iron- and oxygen-dependent manner) as well as in other processes regulating gene expression, including transcriptional pause release. Moreover, a link with tumour progression has been suggested. In the present review we look at biochemical, structural and cellular work on Jmjd6, highlighting areas of controversy and consensus.
Collapse
Affiliation(s)
- Angelika Böttger
- *Department of Biology II, Ludwig Maximillians University, Munich, Germany
| | - Md Saiful Islam
- †Chemistry Research Laboratory and Oxford Centre for Integrative Systems Biology, University of Oxford, Oxford, UK
| | - Rasheduzzaman Chowdhury
- †Chemistry Research Laboratory and Oxford Centre for Integrative Systems Biology, University of Oxford, Oxford, UK
| | - Christopher J Schofield
- †Chemistry Research Laboratory and Oxford Centre for Integrative Systems Biology, University of Oxford, Oxford, UK
| | - Alexander Wolf
- ‡Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
38
|
Increased cardiac remodeling in cardiac-specific Flt-1 receptor knockout mice with pressure overload. Cell Tissue Res 2015; 362:389-98. [PMID: 26017635 DOI: 10.1007/s00441-015-2209-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/05/2015] [Indexed: 12/14/2022]
Abstract
Vascular endothelial growth factor (VEGF) inhibition has previously been shown to have damaging effects on the heart. Because the role of Flt-1 (a phosphotyrosine kinase receptor for VEGF) in cardiac function and hypertrophy is unclear, we generated mice lacking Flt-1 only in their cardiomyocytes (Flt-1 KO). The hearts from 8- to 10-week-old mice were measured by using echocardiography and histology. No significant differences were seen in fraction shortening, cross-sectional area of cardiomyocytes, and interstitial collagen fraction between littermate controls and KO mice at baseline. To test the hypothesis that Flt-1 is involved in cardiac remodeling, we performed transverse aorta constriction (TAC) by ligating the transverse ascending aorta. Four weeks after TAC, echocardiography of the mice was performed, and the hearts were excised for pathological analysis and Western blotting. No difference in mortality was found between Flt-1 KO mice and controls; however, KO mice showed a greater cardiomyocyte cross-sectional area and interstitial collagen fraction than controls. Western blotting indicated that AKT was activated less in Flt-1 KO hearts after TAC compared with that in control hearts. Thus, Flt-1 deletion in cardiomyocytes increased hypertrophy, fibrosis, and regression of AKT phosphorylation. Our study suggests that Flt-1 plays a critical role in cardiac hypertrophy induced by pressure overload via the activation of AKT, which seems to be cardioprotective.
Collapse
|
39
|
Anton L, Olarerin-George AO, Hogenesch JB, Elovitz MA. Placental expression of miR-517a/b and miR-517c contributes to trophoblast dysfunction and preeclampsia. PLoS One 2015; 10:e0122707. [PMID: 25799546 PMCID: PMC4370750 DOI: 10.1371/journal.pone.0122707] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 02/21/2015] [Indexed: 11/19/2022] Open
Abstract
Preeclampsia is a pregnancy specific hypertensive disease that confers significant maternal and fetal risks. While the exact pathophysiology of preeclampsia is unknown, it is widely accepted that placental dysfunction is mechanistically involved. Recent studies reported aberrant expression of placenta-specific microRNAs (miRNAs) in preeclampsia including miR-517a/b and miR-517c. Using placental biopsies from a preeclampsia case-control study, we found increased expression of miR-517a/b in term and preterm preeclampsia vs controls, while, miR-517c was increased only in preterm preeclampsia vs controls. To determine if miR-517a/b and miR-517c are regulated by hypoxia, we treated first trimester primary extravillous trophoblast cells (EVTs) with a hypoxia mimetic and found both were induced. To test for a mechanistic role in placental function, we overexpressed miR-517a/b or miR-517c in EVTs which resulted in decreased trophoblast invasion. Additionally, we found that miR-517a/b and miR-517c overexpression increased expression of the anti-angiogenic protein, sFLT1. The regulation of sFLT1 is mostly unknown, however, TNFSF15, a cytokine involved in FLT1 splicing, was also increased by miR-517a/b and miR-517c in EVTs. In summary, we demonstrate that miR-517a/b and miR-517c contribute to the development of preeclampsia and suggest that these miRNAs play a critical role in regulating trophoblast and placental function.
Collapse
Affiliation(s)
- Lauren Anton
- Maternal and Child Health Research Program, Department of Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| | - Anthony O. Olarerin-George
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Pharmacology and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - John B. Hogenesch
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Pharmacology and the Institute for Translational Medicine and Therapeutics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Michal A. Elovitz
- Maternal and Child Health Research Program, Department of Obstetrics and Gynecology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
40
|
IL12Rβ1ΔTM is a secreted product of il12rb1 that promotes control of extrapulmonary tuberculosis. Infect Immun 2014; 83:560-71. [PMID: 25404030 DOI: 10.1128/iai.01230-13] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
IL12RB1 is a human gene that is important for resistance to Mycobacterium tuberculosis infection. IL12RB1 is expressed by multiple leukocyte lineages, and encodes a type I transmembrane protein (IL12Rβ1) that associates with IL12p40 and promotes the development of host-protective T(H)1 cells. Recently, we observed that il12rb1—the mouse homolog of IL12RB1—is alternatively spliced by leukocytes to produce a second isoform (IL12Rβ1ΔTM) that has biological properties distinct from IL12Rβ1. Although the expression of IL12Rβ1ΔTM is elicited by M. tuberculosis in vivo, and its overexpression enhances IL12p40 responsiveness in vitro, the contribution of IL12Rβ1ΔTM to controlling M. tuberculosis infection has not been tested. Here, we demonstrate that IL12Rβ1ΔTM represents a secreted product of il12rb1 that, when absent from mice, compromises their ability to control M. tuberculosis infection in extrapulmonary organs. Furthermore, elevated M. tuberculosis burdens in IL12Rβ1ΔTM-deficient animals are associated with decreased lymph node cellularity and a decline in TH1 development. Collectively, these data support a model wherein IL12Rβ1ΔTM is a secreted product of il12rb1 that promotes resistance to M. tuberculosis infection by potentiating T(H) cells response to IL-12.
Collapse
|
41
|
Voelkel NF, Gomez-Arroyo J. The Role of Vascular Endothelial Growth Factor in Pulmonary Arterial Hypertension. The Angiogenesis Paradox. Am J Respir Cell Mol Biol 2014; 51:474-84. [DOI: 10.1165/rcmb.2014-0045tr] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
42
|
Death receptor 3 mediates TNFSF15- and TNFα-induced endothelial cell apoptosis. Int J Biochem Cell Biol 2014; 55:109-18. [PMID: 25161149 DOI: 10.1016/j.biocel.2014.08.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/14/2014] [Accepted: 08/17/2014] [Indexed: 01/09/2023]
Abstract
Tumor necrosis factor superfamily 15 (TNFSF15) suppresses angiogenesis by specifically inducing apoptosis in proliferating endothelial cells. Death receptor 3 (DR3), a member of the TNF receptor superfamily (TNFRSF25), has been identified as a receptor for TNFSF15 to activate T cells. It is unclear, however, whether DR3 mediates TNFSF15 activity on endothelial cells. Here we show that siRNA-mediated knockdown of DR3 in an in vivo Matrigel angiogenesis assay, or in adult bovine aortic endothelial (ABAE) cell cultures, leads to resistance of endothelial cells to TNFSF15-induced apoptosis. Interestingly, DR3-depleted cells also exhibited markedly diminished responsiveness to TNFα cytotoxicity, even though DR3 is not a receptor for TNFα. Treatment of the cells with either TNFSF15 siRNA or a TNFSF15-neutralizing antibody, 4-3H, also results in a significant inhibition of TNFα-induced apoptosis. Mechanistically, DR3 siRNA treatment gives rise to an increase of ERK1/2 MAPK activity, and up-regulation of the anti-apoptotic proteins c-FLIP and Bcl-2, thus strengthening apoptosis-resisting potential in the cells. These findings indicate that DR3 mediates TNFSF15-induced endothelial cell apoptosis, and that up-regulation of TNFSF15 expression stimulated by TNFα is partly but significantly responsible for TNFα-induced apoptosis in endothelial cells.
Collapse
|
43
|
Heim A, Grimm C, Müller U, Häußler S, Mackeen MM, Merl J, Hauck SM, Kessler BM, Schofield CJ, Wolf A, Böttger A. Jumonji domain containing protein 6 (Jmjd6) modulates splicing and specifically interacts with arginine-serine-rich (RS) domains of SR- and SR-like proteins. Nucleic Acids Res 2014; 42:7833-50. [PMID: 24914048 PMCID: PMC4081092 DOI: 10.1093/nar/gku488] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The Fe(II) and 2-oxoglutarate dependent oxygenase Jmjd6 has been shown to hydroxylate lysine residues in the essential splice factor U2 auxiliary factor 65 kDa subunit (U2AF65) and to act as a modulator of alternative splicing. We describe further evidence for the role of Jmjd6 in the regulation of pre-mRNA processing including interactions of Jmjd6 with multiple arginine–serine-rich (RS)-domains of SR- and SR-related proteins including U2AF65, Luc7-like protein 3 (Luc7L3), SRSF11 and Acinus S′, but not with the bona fide RS-domain of SRSF1. The identified Jmjd6 target proteins are involved in different mRNA processing steps and play roles in exon dependent alternative splicing and exon definition. Moreover, we show that Jmjd6 modifies splicing of a constitutive splice reporter, binds RNA derived from the reporter plasmid and punctually co-localises with nascent RNA. We propose that Jmjd6 exerts its splice modulatory function by interacting with specific SR-related proteins during splicing in a RNA dependent manner.
Collapse
Affiliation(s)
- Astrid Heim
- Department of Biology II, Ludwig Maximilians University, Munich, Großhaderner Strasse 2, 82152 Planegg-Martinsried, Germany
| | - Christina Grimm
- Department of Biology II, Ludwig Maximilians University, Munich, Großhaderner Strasse 2, 82152 Planegg-Martinsried, Germany
| | - Udo Müller
- Department of Biology II, Ludwig Maximilians University, Munich, Großhaderner Strasse 2, 82152 Planegg-Martinsried, Germany
| | - Simon Häußler
- Department of Biology II, Ludwig Maximilians University, Munich, Großhaderner Strasse 2, 82152 Planegg-Martinsried, Germany
| | - Mukram M Mackeen
- Chemistry Research Laboratory and Oxford Centre for Integrative Systems Biology, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK Research Unit Protein Science, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | - Juliane Merl
- Henry Wellcome Building for Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Stefanie M Hauck
- Henry Wellcome Building for Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Benedikt M Kessler
- School of Chemical Science, Faculty of Science and Technology, and Institute of Systems Biology (INBIOSIS) Universiti Kebangsaan Malaysia, 43600 Bangi, Selangor Darul Ehsan, Malaysia
| | - Christopher J Schofield
- Chemistry Research Laboratory and Oxford Centre for Integrative Systems Biology, University of Oxford, 12 Mansfield Road, Oxford OX1 3TA, UK
| | - Alexander Wolf
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstädter Landstrasse 1, 85764 Neuherberg, Germany
| | - Angelika Böttger
- Department of Biology II, Ludwig Maximilians University, Munich, Großhaderner Strasse 2, 82152 Planegg-Martinsried, Germany
| |
Collapse
|