1
|
Wang YC, Zhu Y, Meng WT, Zheng Y, Guan XQ, Shao CL, Li XY, Hu D, Wang MZ, Guo HD. Dihydrotanshinone I improves cardiac function by promoting lymphangiogenesis after myocardial ischemia-reperfusion injury. Eur J Pharmacol 2025; 989:177245. [PMID: 39753160 DOI: 10.1016/j.ejphar.2024.177245] [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: 07/11/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
Dihydrotanshinone I (DHT) is an active ingredient derived from Salvia miltiorrhiza. Previous studies have demonstrated that DHT can improve cardiac function in rats with myocardial ischemia-reperfusion injury (IR). However, the mechanism by which DHT improves myocardial injury in rats still requires further research. Lymphangiogenesis can reduce myocardial edema, inflammation, and fibrosis after myocardial infarction in rats, and improve cardiac function. In this study, the changes in cardiac functions, collagen fiber deposition in the infarcted area and the level of relevant indicators of lymphangiogenesis were examined by echocardiography, Masson's trichrome staining, immunohistochemistry and Western blot, respectively. Human lymphatic endothelial cells (HLECs) were transfected with siVE-cadherin and siVEGFR-3, and the effects of DHT on HLEC cell viability, migration and tube formation were detected through CCK8, TUNEL, transwell, wound healing and tube formation assay. We found that in myocardial IR rats treated with DHT, the levels of LYVE-1, PROX1, VEGF-C, VEGFR-3, IGF-1, podoplanin and IGF-1R, which are associated with lymphangiogenesis, were increased, as well as the level of VE-cadherin, which maintains endothelial cell function. DHT reduced the levels of inflammatory factors and myocardial cell apoptosis, thereby improving cardiac function after I/R. To explore the mechanism of DHT promoting lymphangiogenesis, H2O2 and OGD/R injury models of HLECs were constructed to simulate the microenvironment of myocardial IR in vitro. The results proved that DHT could reduce the damage and apoptosis of HLECs. On the other hand, DHT enhanced the expression of VEGFR-3 and VE-cadherin in HLECs, promoted cell migration and tube formation. The effects of DHT on the tube formation and migration of HLECs were significantly decreased after knocking down VEGFR-3 or VE-cadherin. Our research proposed that DHT could improve the heart function after IR through the enhancement of lymphangiogenesis and contributed to the development of the treatment methods for myocardial IR.
Collapse
Affiliation(s)
- Ya-Chao Wang
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Zhu
- Department of Neurological Rehabilitation, The Second Rehabilitation Hospital of Shanghai, Shanghai, China
| | - Wan-Ting Meng
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Zheng
- Jiading Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Xiao-Qi Guan
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chang-le Shao
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiu-Ya Li
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Hu
- Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, 200137, China.
| | - Ming-Zhu Wang
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| | - Hai-Dong Guo
- Academy of Integrated Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
2
|
Li XX, Zhang DC, Wang Y, Wen J, Wang XJ, Cao YL, Jiang R, Li JR, Li YN, Liu HH, Xie WH, Xu ZF, Hu P, Zou K. Cadherin-18 loss in prospermatogonia and spermatogonial stem cells enhances cell adhesion through a compensatory mechanism. Zool Res 2024; 45:1048-1060. [PMID: 39147719 PMCID: PMC11491781 DOI: 10.24272/j.issn.2095-8137.2023.373] [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: 04/01/2024] [Accepted: 04/12/2024] [Indexed: 08/17/2024] Open
Abstract
Extracellular membrane proteins are crucial for mediating cell attachment, recognition, and signal transduction in the testicular microenvironment, particularly germline stem cells. Cadherin 18 (CDH18), a type II classical cadherin, is primarily expressed in the nervous and reproductive systems. Here, we investigated the expression of CDH18 in neonatal porcine prospermatogonia (ProSGs) and murine spermatogonial stem cells (SSCs). Disruption of CDH18 expression did not adversely affect cell morphology, proliferation, self-renewal, or differentiation in cultured porcine ProSGs, but enhanced cell adhesion and prolonged cell maintenance. Transcriptomic analysis indicated that the down-regulation of CDH18 in ProSGs significantly up-regulated genes and signaling pathways associated with cell adhesion. To further elucidate the function of CDH18 in germ cells, Cdh18 knockout mice were generated, which exhibited normal testicular morphology, histology, and spermatogenesis. Transcriptomic analysis showed increased expression of genes associated with adhesion, consistent with the observations in porcine ProSGs. The interaction of CDH18 with β-catenin and JAK2 in both porcine ProSGs and murine SSCs suggested an inhibitory effect on the canonical Wnt and JAK-STAT signaling pathways during CDH18 deficiency. Collectively, these findings highlight the crucial role of CDH18 in regulating cell adhesion in porcine ProSGs and mouse SSCs. Understanding this regulatory mechanism provides significant insights into the testicular niche.
Collapse
Affiliation(s)
- Xiao-Xiao Li
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Dan-Chen Zhang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Yan Wang
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, China
| | - Jian Wen
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Xing-Ju Wang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Yu-Lu Cao
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Ru Jiang
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Jia-Rui Li
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Yi-Nuo Li
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - He-He Liu
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Wen-Hai Xie
- School of Life Sciences and Medicine, Shandong University of Technology, Zibo, Shandong 255000, China
| | - Zheng-Feng Xu
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, China. E-mail:
| | - Ping Hu
- Department of Prenatal Diagnosis, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, Jiangsu 210004, China. E-mail:
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
- Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China. E-mail:
| |
Collapse
|
3
|
Nguyen M, Sullivan J, Shen W. Retinal vascular remodeling in photoreceptor degenerative disease. Exp Eye Res 2023; 234:109566. [PMID: 37423458 DOI: 10.1016/j.exer.2023.109566] [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: 03/01/2023] [Revised: 06/05/2023] [Accepted: 07/03/2023] [Indexed: 07/11/2023]
Abstract
Abnormal vasculature in the retina, specifically tortuous vessels and capillary degeneration, is common in many of the most prevalent retinal degenerative diseases, currently affecting millions of people across the world. However, the formation and development of abnormal vasculature in the context of retinal degenerative diseases are still poorly understood. The FVB/N (rd1) and rd10 mice are well-studied animal models of retinal degenerative diseases, but how photoreceptor degeneration leads to vascular abnormality in the diseases remains to be elucidated. Here, we used advancements in confocal microscopy, immunohistochemistry, and image analysis software to systematically characterize the pathological vasculature in the FVB/N (rd1) and rd10 mice, known as a chronic, rapid and slower retinal degenerative model, respectively. We demonstrated that there was plexus-specific vascular degeneration in the retinal trilaminar vascular network paralleled to photoreceptor degeneration in the diseased retinas. We also quantitatively analyzed the vascular structural architecture in the wild-type and diseased retinas to provide valuable information on vascular remodeling in retinal degenerative disease.
Collapse
Affiliation(s)
- Matthew Nguyen
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - James Sullivan
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Wen Shen
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA.
| |
Collapse
|
4
|
Tonami K, Hayashi T, Uchijima Y, Kanai M, Yura F, Mada J, Sugahara K, Kurihara Y, Kominami Y, Ushijima T, Takubo N, Liu X, Tozawa H, Kanai Y, Tokihiro T, Kurihara H. Coordinated linear and rotational movements of endothelial cells compartmentalized by VE-cadherin drive angiogenic sprouting. iScience 2023; 26:107051. [PMID: 37426350 PMCID: PMC10329149 DOI: 10.1016/j.isci.2023.107051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/22/2023] [Accepted: 06/01/2023] [Indexed: 07/11/2023] Open
Abstract
Angiogenesis is a sequential process to extend new blood vessels from preexisting ones by sprouting and branching. During angiogenesis, endothelial cells (ECs) exhibit inhomogeneous multicellular behaviors referred to as "cell mixing," in which ECs repetitively exchange their relative positions, but the underlying mechanism remains elusive. Here we identified the coordinated linear and rotational movements potentiated by cell-cell contact as drivers of sprouting angiogenesis using in vitro and in silico approaches. VE-cadherin confers the coordinated linear motility that facilitated forward sprout elongation, although it is dispensable for rotational movement, which was synchronous without VE-cadherin. Mathematical modeling recapitulated the EC motility in the two-cell state and angiogenic morphogenesis with the effects of VE-cadherin-knockout. Finally, we found that VE-cadherin-dependent EC compartmentalization potentiated branch elongations, and confirmed this by mathematical simulation. Collectively, we propose a way to understand angiogenesis, based on unique EC behavioral properties that are partially dependent on VE-cadherin function.
Collapse
Affiliation(s)
- Kazuo Tonami
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
| | - Tatsuya Hayashi
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
- Graduate School of Mathematical Science, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8914, Japan
- Faculty of Science and Engineering, Chuo University, 1-13-27 Kasuga, Bunkyo-ku, Tokyo 112-8551, Japan
| | - Yasunobu Uchijima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Masahiro Kanai
- Department of Education and Creation Engineering, Kurume Institute of Technology, 2228-66 Kamitsu-machi, Kurume, Fukuoka 830-0052, Japan
| | - Fumitaka Yura
- Department of Complex and Intelligent Systems, School of Systems Information Science, Future University Hakodate, 116-2 Kamedanakano-cho, Hakodate, Hokkaido 041-8655, Japan
| | - Jun Mada
- College of Industrial Technology, Nihon University, 2-11-1 Shin-ei, Narashino, Chiba 275-8576, Japan
| | - Kei Sugahara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yukiko Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuri Kominami
- Department of Aquatic Bioscience, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1, Yayoi, Bunkyo-ku, Tokyo 113-865, Japan
| | - Toshiyuki Ushijima
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naoko Takubo
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
- Isotope Science Center, The University of Tokyo, 2-11-16, Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Xiaoxiao Liu
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hideto Tozawa
- Department of Chemistry, Graduate School of Science, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yoshimitsu Kanai
- Cell Biology and Anatomy, Graduate School of Medicine, Wakayama Medical University, 811-1 Kimiidera, Wakayama 641-8509, Japan
| | - Tetsuji Tokihiro
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
- Graduate School of Mathematical Science, The University of Tokyo, 3-8-1, Komaba, Meguro-ku, Tokyo 153-8914, Japan
- Department of Mathematical Engineering, Faculty of Engineering, Musashino University, 3-3-3 Ariake, Koto-ku, Tokyo 135-8181, Japan
| | - Hiroki Kurihara
- Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Chiyoda-ku, Tokyo 102-0076, Japan
| |
Collapse
|
5
|
Galdos FX, Xu S, Goodyer WR, Duan L, Huang YV, Lee S, Zhu H, Lee C, Wei N, Lee D, Wu SM. devCellPy is a machine learning-enabled pipeline for automated annotation of complex multilayered single-cell transcriptomic data. Nat Commun 2022; 13:5271. [PMID: 36071107 PMCID: PMC9452519 DOI: 10.1038/s41467-022-33045-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 08/31/2022] [Indexed: 11/09/2022] Open
Abstract
A major informatic challenge in single cell RNA-sequencing analysis is the precise annotation of datasets where cells exhibit complex multilayered identities or transitory states. Here, we present devCellPy a highly accurate and precise machine learning-enabled tool that enables automated prediction of cell types across complex annotation hierarchies. To demonstrate the power of devCellPy, we construct a murine cardiac developmental atlas from published datasets encompassing 104,199 cells from E6.5-E16.5 and train devCellPy to generate a cardiac prediction algorithm. Using this algorithm, we observe a high prediction accuracy (>90%) across multiple layers of annotation and across de novo murine developmental data. Furthermore, we conduct a cross-species prediction of cardiomyocyte subtypes from in vitro-derived human induced pluripotent stem cells and unexpectedly uncover a predominance of left ventricular (LV) identity that we confirmed by an LV-specific TBX5 lineage tracing system. Together, our results show devCellPy to be a useful tool for automated cell prediction across complex cellular hierarchies, species, and experimental systems.
Collapse
Affiliation(s)
- Francisco X Galdos
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, USA
| | - Sidra Xu
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - William R Goodyer
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, USA
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, USA
| | - Lauren Duan
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuhsin V Huang
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Soah Lee
- Biopharmaceutical Convergence, School of Pharmacy, Sungkyunkwan University, Suwon, South Korea
| | - Han Zhu
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Palo Alto, USA
| | - Carissa Lee
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Nicholas Wei
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel Lee
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Sean M Wu
- Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Palo Alto, USA.
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Palo Alto, USA.
| |
Collapse
|
6
|
Li H, Chang HM, Li S, Klausen C, Shi Z, Leung PC. Characterization of the roles of amphiregulin and transforming growth factor β1 in microvasculature-like formation in human granulosa-lutein cells. Front Cell Dev Biol 2022; 10:968166. [PMID: 36092732 PMCID: PMC9448859 DOI: 10.3389/fcell.2022.968166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Vascular endothelial-cadherin (VE-cadherin) is an essential component that regulates angiogenesis during corpus luteum formation. Amphiregulin (AREG) and transforming growth factor β1 (TGF-β1) are two intrafollicular factors that possess opposite functions in directing corpus luteum development and progesterone synthesis in human granulosa-lutein (hGL) cells. However, whether AREG or TGF-β1 regulates the VE-cadherin expression and subsequent angiogenesis in the human corpus luteum remains to be elucidated. Results showed that hGL cells cultured on Matrigel spontaneously formed capillary-like and sprout-like microvascular networks. Results of specific inhibitor treatment and small interfering RNA-mediated knockdown revealed that AREG promoteed microvascular-like formation in hGL cells by upregulating the VE-cadherin expression mediated by the epidermal growth factor receptor (EGFR)-extracellular signal-regulated kinase1/2 (ERK1/2) signaling pathway. However, TGF-β1 suppressed microvascular-like formation in hGL cells by downregulating VE-cadherin expression mediated by the activin receptor-like kinase (ALK)5-Sma- and Mad-related protein (SMAD)2/3/4 signaling pathway. Collectively, this study provides important insights into the underlying molecular mechanisms by which TGF-β1 and AREG differentially regulate corpus luteum formation in human ovaries.
Collapse
Affiliation(s)
- Hui Li
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Department of Obstetrics and Gynaecology, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Hsun-Ming Chang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
- *Correspondence: Hsun-Ming Chang, ; Peter C.K. Leung,
| | - Saijiao Li
- Department of Obstetrics and Gynaecology, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Christian Klausen
- Department of Obstetrics and Gynaecology, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Zhendan Shi
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Key Laboratory of Animal Breeding and Reproduction, Institute of Animal Science, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Peter C.K. Leung
- Department of Obstetrics and Gynaecology, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Hsun-Ming Chang, ; Peter C.K. Leung,
| |
Collapse
|
7
|
Yang C, Shi Y, Li X, Guan L, Li H, Lin J. Cadherins and the pathogenesis of epilepsy. Cell Biochem Funct 2022; 40:336-348. [PMID: 35393670 DOI: 10.1002/cbf.3699] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 02/22/2022] [Accepted: 03/12/2022] [Indexed: 12/13/2022]
Abstract
Epilepsy is a nervous system disease caused by abnormal discharge of brain neurons, which is characterized by recurrent seizures. The factors that induce epilepsy include genetic and environmental factors. Genetic factors are important pathogenic factors of epilepsy, such as epilepsy caused by protocadherin-19 (PCDH-19) mutation, which is an X-linked genetic disease. It is more common in female heterozygotes, which are caused by mutations in the PCDH-19 gene. Epilepsy caused by environmental factors is mainly caused by brain injury, which is commonly caused by brain tumors, brain surgery, or trauma to the brain. In addition, the pathogenesis of epilepsy is closely related to abnormalities in some signaling pathways. The Wnt/β-catenin signaling pathway is considered a new target for the treatment of epilepsy. This review summarizes these factors inducing epilepsy and the research hypotheses regarding the pathogenesis of epilepsy. The focus of this review centers on cadherins and the pathogenesis of epilepsy. We analyzed the pathogenesis of epilepsy induced by N-cadherin and PCDH-19 in the cadherin family members. Finally, we expect that in the future, new breakthroughs will be made in the study of the pathogenesis and mechanism of epilepsy at the cellular and molecular levels.
Collapse
Affiliation(s)
- Ciqing Yang
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, China
| | - Yaping Shi
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Xiaoying Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Lihong Guan
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Han Li
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China
| | - Juntang Lin
- Stem Cells & Biotherapy Engineering Research Center of Henan, College of Life Science and Technology, Xinxiang Medical University, Xinxiang, China.,Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang, China
| |
Collapse
|
8
|
Simulated Microgravity Increases the Permeability of HUVEC Monolayer through Up-Regulation of Rap1GAP and Decreased Rap2 Activation. Int J Mol Sci 2022; 23:ijms23020630. [PMID: 35054818 PMCID: PMC8776081 DOI: 10.3390/ijms23020630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 02/01/2023] Open
Abstract
Space microgravity condition has great physiological influence on astronauts’ health. The interaction of endothelial cells, which control vascular permeability and immune responses, is sensitive to mechanical stress. However, whether microgravity has significant effects on the physiological function of the endothelium has not been investigated. In order to address such a question, a clinostat-based culture model with a HUVEC monolayer being inside the culture vessel under the simulated microgravity (SMG) was established. The transmittance of FITC-tagged dextran was used to estimate the change of integrity of the adherens junction of the HUVEC monolayer. Firstly, we found that the permeability of the HUVEC monolayer was largely increased after SMG treatment. To elucidate the mechanism of the increased permeability of the HUVEC monolayer under SMG, the levels of total expression and activated protein levels of Rap1 and Rap2 in HUVEC cells, which regulate the adherens junction of endothelial cells, were detected by WB and GST pull-down after SMG. As the activation of both Rap1 and Rap2 was significantly decreased under SMG, the expression of Rap1GEF1 (C3G) and Rap1GAP in HUVECs, which regulate the activation of them, was further determined. The results indicate that both C3G and Rap1GAP showed a time-dependent increase with the expression of Rap1GAP being dominant at 48 h after SMG. The down-regulation of the expression of junctional proteins, VE-cadherin and β-catenin, in HUVEC cells was also confirmed by WB and immunofluorescence after SMG. To clarify whether up-regulation of Rap1GAP is necessary for the increased permeability of the HUVEC monolayer after SMG, the expression of Rap1GAP was knocked down by Rap1GAP-shRNA, and the change of permeability of the HUVEC monolayer was detected. The results indicate that knock-down of Rap1GAP reduced SMG-induced leaking of the HUVEC monolayer in a time-dependent manner. In total, our results indicate that the Rap1GAP-Rap signal axis was necessary for the increased permeability of the HUVEC monolayer along with the down-regulation of junctional molecules including VE-cadherin and β-catenin.
Collapse
|
9
|
Li F, Xu J, Liu S. Cancer Stem Cells and Neovascularization. Cells 2021; 10:cells10051070. [PMID: 33946480 PMCID: PMC8147173 DOI: 10.3390/cells10051070] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/19/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cells (CSCs) refer to a subpopulation of cancer cells responsible for tumorigenesis, metastasis, and drug resistance. Increasing evidence suggests that CSC-associated tumor neovascularization partially contributes to the failure of cancer treatment. In this review, we discuss the roles of CSCs on tumor-associated angiogenesis via trans-differentiation or forming the capillary-like vasculogenic mimicry, as well as the roles of CSCs on facilitating endothelial cell-involved angiogenesis to support tumor progression and metastasis. Furthermore, we discuss the underlying regulation mechanisms, including the intrinsic signals of CSCs and the extrinsic signals such as cytokines from the tumor microenvironment. Further research is required to identify and verify some novel targets to develop efficient therapeutic approaches for more efficient cancer treatment through interfering CSC-mediated neovascularization.
Collapse
Affiliation(s)
- Fengkai Li
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jiahui Xu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Cancer Institutes, Fudan University, Shanghai 200032, China; (F.L.); (J.X.)
- Key Laboratory of Breast Cancer in Shanghai, The Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
- The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Correspondence: ; Tel.: +86-21-34771023
| |
Collapse
|
10
|
Lei H, Yang T, Mahmood S, Abo-Ismail M, Roy BC, Li C, Plastow GS, Bruce HL. A genome-wide case-control association study of dark cutting in beef cattle. CANADIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1139/cjas-2019-0039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The genetic architecture of dark cutting was investigated with a case-control genome-wide association study on two groups of beef cattle analyzed separately and together (combined group). Groups I (n = 64) and II (n = 150) were genotyped using the 70K GeneSeek Genomic Profiler for Beef Cattle-HD and the 50K Illumina BovineSNP50v2 BeadChip, respectively. Dark cutting was analyzed as a binary trait (case versus control) using logistic regression in an additive model implemented in PLINK version 1.9. Significant loci were not identified when correcting for multiple testing (false discovery rate), suggesting that the trait is not controlled by genes with big effects, or the sample size was not large enough to detect these major genes. Regions harbouring single-nucleotide polymorphisms (SNPs) with a raw p < 0.01 using 1 MB window were analyzed for gene function using the ingenuity pathway analysis. For groups I, II, and the combined group, 449, 301, and 191 SNPs were identified, respectively. Genes identified were involved in pyruvic acid modification and release, 2-deoxyglucose clearance and disposal, sucrose recognition, energy production, and metabolism of carbohydrate. Although detected SNP associations require validation in a large population, results suggested the possibility for marker-assisted or genomic selection of beef cattle to reduce dark cutting.
Collapse
Affiliation(s)
- Huaigang Lei
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Tianfu Yang
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Shahid Mahmood
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Mohammed Abo-Ismail
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Animal Science, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| | - Bimol C. Roy
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Changxi Li
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Agriculture and Agri-Food Canada, Lacombe Research and Development Centre, 6000 C&E Trail, Lacombe, AB T4L 1W1, Canada
| | - Graham S. Plastow
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Heather L. Bruce
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| |
Collapse
|
11
|
Seebach J, Klusmeier N, Schnittler H. Autoregulatory "Multitasking" at Endothelial Cell Junctions by Junction-Associated Intermittent Lamellipodia Controls Barrier Properties. Front Physiol 2021; 11:586921. [PMID: 33488392 PMCID: PMC7815704 DOI: 10.3389/fphys.2020.586921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/30/2020] [Indexed: 01/12/2023] Open
Abstract
Vascular endothelial cell (EC) junctions are key structures controlling tissue homeostasis in physiology. In the last three decades, excellent studies have addressed many aspects of this complex and highly dynamic regulation, including cell signaling, remodeling processes of the proteins of tight junctions, adherens junctions, and gap junctions, the cytoskeleton, and post-transcriptional modifications, transcriptional activation, and gene silencing. In this dynamic process, vascular endothelial cadherin (VE-cadherin) provides the core structure of EC junctions mediating the physical adhesion of cells as well as the control of barrier function and monolayer integrity via remodeling processes, regulation of protein expression and post-translational modifications. In recent years, research teams have documented locally restricted dynamics of EC junctions in which actin-driven protrusions in plasma membranes play a central role. In this regard, our research group showed that the dynamics of VE-cadherin is driven by small (1-5 μm) actin-mediated protrusions in plasma membranes that, due to this specific function, were named "junction-associated intermittent lamellipodia" (JAIL). JAIL form at overlapping, adjacent cells, and exactly at this site new VE-cadherin interactions occur, leading to new VE-cadherin adhesion sites, a process that restores weak or lost VE-cadherin adhesion. Mechanistically, JAIL formation occurs locally restricted (1-5 μm) and underlies autoregulation in which the local VE-cadherin concentration is an important parameter. A decrease in the local concentration of VE-cadherin stimulates JAIL formation, whereas an increase in the concentration of VE-cadherin blocks it. JAIL mediated VE-cadherin remodeling at the subjunctional level have been shown to be of crucial importance in angiogenesis, wound healing, and changes in permeability during inflammation. The concept of subjunctional regulation of EC junctions is strongly supported by permeability assays, which can be employed to quantify actin-driven subjunctional changes. In this brief review, we summarize and discuss the current knowledge and concepts of subjunctional regulation in the endothelium.
Collapse
Affiliation(s)
- Jochen Seebach
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Nadine Klusmeier
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| |
Collapse
|
12
|
Rangel Olguin AG, Rochon PL, Krishnaswamy A. New Optical Tools to Study Neural Circuit Assembly in the Retina. Front Neural Circuits 2020; 14:44. [PMID: 32848633 PMCID: PMC7424070 DOI: 10.3389/fncir.2020.00044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 06/23/2020] [Indexed: 12/17/2022] Open
Abstract
During development, neurons navigate a tangled thicket of thousands of axons and dendrites to synapse with just a few specific targets. This phenomenon termed wiring specificity, is critical to the assembly of neural circuits and the way neurons manage this feat is only now becoming clear. Recent studies in the mouse retina are shedding new insight into this process. They show that specific wiring arises through a series of stages that include: directed axonal and dendritic growth, the formation of neuropil layers, positioning of such layers, and matching of co-laminar synaptic partners. Each stage appears to be directed by a distinct family of recognition molecules, suggesting that the combinatorial expression of such family members might act as a blueprint for retinal connectivity. By reviewing the evidence in support of each stage, and by considering their underlying molecular mechanisms, we attempt to synthesize these results into a wiring model which generates testable predictions for future studies. Finally, we conclude by highlighting new optical methods that could be used to address such predictions and gain further insight into this fundamental process.
Collapse
|
13
|
Yu W, Yang L, Li T, Zhang Y. Cadherin Signaling in Cancer: Its Functions and Role as a Therapeutic Target. Front Oncol 2019; 9:989. [PMID: 31637214 PMCID: PMC6788064 DOI: 10.3389/fonc.2019.00989] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/16/2019] [Indexed: 12/12/2022] Open
Abstract
Cadherin family includes lists of transmembrane glycoproteins which mediate calcium-dependent cell-cell adhesion. Cadherin-mediated adhesion regulates cell growth and differentiation throughout life. Through the establishment of the cadherin-catenin complex, cadherins provide normal cell-cell adhesion and maintain homeostatic tissue architecture. In the process of cell recognition and adhesion, cadherins act as vital participators. As results, the disruption of cadherin signaling has significant implications on tumor formation and progression. Altered cadherin expression plays a vital role in tumorigenesis, tumor progression, angiogenesis, and tumor immune response. Based on ongoing research into the role of cadherin signaling in malignant tumors, cadherins are now being considered as potential targets for cancer therapies. This review will demonstrate the mechanisms of cadherin involvement in tumor progression, and consider the clinical significance of cadherins as therapeutic targets.
Collapse
Affiliation(s)
- Weina Yu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Li Yang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Ting Li
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
14
|
Xiao ZP, Zhao JL, Rong WL, Jiang JW, Li MH. Role of Vascular Endothelial-Cadherin and p120-Catenin in the Formation of Experimental Intracranial Aneurysm in Animals. World Neurosurg 2019; 128:e177-e184. [PMID: 30995547 DOI: 10.1016/j.wneu.2019.04.077] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 04/07/2019] [Accepted: 04/08/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Dysfunction of endothelial cells (ECs) constitutes a critical factor in the formation of intracranial aneurysms (IAs). However, little is known about the response of ECs to hemodynamic insults and its contribution to IA formation. METHODS IAs models were constructed in both adult female New Zealand white rabbits and male Sprague-Dawley rats. Morphologic changes of vessel wall were detected by hematoxylin and eosin staining. Molecular and cellular changes, including p120-catenin (p120ctn) and vascular endothelial-cadherin, in the median sagittal section of the artery bifurcation were analyzed by fluorescent staining. RESULTS Destructive aneurysmal remodeling and the formation of morphologic IAs were observed at the basilar termini of experimental rabbits and the anterior cerebral artery-olfactory artery bifurcation of rats. The expression of p120ctn colocalized with vascular endothelial-cadherin in ECs decreased. Moreover, the expression of p120ctn colocalized with nucleus of ECs increased. These events suggested that p120ctn was transported from the membrane to the nucleus of ECs. CONCLUSIONS The potential mechanism, that IAs are always localizing in the bifurcation apices, may be that the endothelium injury of vessel wall can be induced by different hemodynamic conditions. Hemodynamic changes in artery bifurcation may initiate the formation of IAs.
Collapse
Affiliation(s)
- Zhi-Peng Xiao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China; Department of Neurosurgery, Renji Hospital, School of Medicine of Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Jian-Lan Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China; Department of Neurosurgery, Fudan University Huashan Hospital, Shanghai, P.R. China
| | - Wei-Lin Rong
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Jin-Wen Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China
| | - Mei-Hua Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, P.R. China.
| |
Collapse
|
15
|
Ziegler J, Zalles M, Smith N, Saunders D, Lerner M, Fung KM, Patel M, Wren JD, Lupu F, Battiste J, Towner RA. Targeting ELTD1, an angiogenesis marker for glioblastoma (GBM), also affects VEGFR2: molecular-targeted MRI assessment. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2019; 9:93-109. [PMID: 30911439 PMCID: PMC6420708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/21/2019] [Indexed: 06/09/2023]
Abstract
Glioblastomas (GBM) are deadly brain tumors that currently do not have long-term patient treatments available. GBM overexpress the angiogenesis factor VEGF and its receptor VEGFR2. ETLD1 (epidermal growth factor, latrophilin and seven transmembrane domain-containing protein 1), a G-protein coupled receptor (GPCR) protein, we discovered as a biomarker for high-grade gliomas, is also a novel regulator of angiogenesis. Since it was established that VEGF regulates ELTD1, we wanted to establish if VEGFR2 is also associated with ELTD1, by targeted antibody inhibition. G55 glioma-bearing mice were treated with either anti-ELTD1 or anti-VEGFR2 antibodies. With the use of MRI molecular imaging probes, we were able to detect in vivo levels of either ELTD1 (anti-ELTD1 probe) or VEGFR2 (anti-VEGFR2 probe). Protein expressions were obtained for ELTD1, VEGF or VEGFR2 via immunohistochemistry (IHC). VEGFR2 levels were significantly decreased (P < 0.05) with anti-ELTD1 antibody treatment, and ELTD1 levels were significantly decreased (P < 0.05) with anti-VEGFR2 antibody treatment, both compared to untreated tumors. IHC from mouse tumor tissues established that VEGFR2 and ELTD1 were co-localized. The mouse anti-ELTD1 antibody treatment study indicated that anti-VEGFR2 antibody treatment does not significantly increase survival, decrease tumor volumes, or alter tumor perfusion (measured as relative cerebral blood flow or rCBF). Conversely, anti-ELTD1 antibody treatment was able to significantly increase animal survival (P < 0.01), decrease tumor volumes (P < 0.05), and reduce change in rCBF (P < 0.001), when compared to untreated or IgG-treated tumor bearing mice. Anti-ELTD1 antibody therapy could be beneficial in targeting ELTD1, as well as simultaneously affecting VEGFR2, as a possible GBM treatment.
Collapse
Affiliation(s)
- Jadith Ziegler
- Advanced Magnetic Resonance Center, Oklahoma Medical Research FoundationOklahoma, OK, USA
- Department of Pathology, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
| | - Michelle Zalles
- Advanced Magnetic Resonance Center, Oklahoma Medical Research FoundationOklahoma, OK, USA
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research FoundationOklahoma, OK, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research FoundationOklahoma, OK, USA
| | - Megan Lerner
- Surgery Research Laboratory, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
| | - Maulin Patel
- Cardiovascular Biology, Oklahoma Medical Research FoundationOklahoma, OK, USA
| | - Jonathan D Wren
- Arthritis and Clinical Immunology, Oklahoma Medical Research FoundationOklahoma, OK, USA
| | - Florea Lupu
- Cardiovascular Biology, Oklahoma Medical Research FoundationOklahoma, OK, USA
| | - James Battiste
- Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
- Department of Neurology, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
| | - Rheal A Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research FoundationOklahoma, OK, USA
- Department of Pathology, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences CenterOklahoma, OK, USA
| |
Collapse
|
16
|
Cao J, Schnittler H. Putting VE-cadherin into JAIL for junction remodeling. J Cell Sci 2019; 132:132/1/jcs222893. [DOI: 10.1242/jcs.222893] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT
Junction dynamics of endothelial cells are based on the integration of signal transduction, cytoskeletal remodeling and contraction, which are necessary for the formation and maintenance of monolayer integrity, but also enable repair and regeneration. The VE-cadherin–catenin complex forms the molecular basis of the adherence junctions and cooperates closely with actin filaments. Several groups have recently described small actin-driven protrusions at the cell junctions that are controlled by the Arp2/3 complex, contributing to cell junction regulation. We identified these protrusions as the driving force for VE-cadherin dynamics, as they directly induce new VE-cadherin-mediated adhesion sites, and have accordingly referred to these structures as junction-associated intermittent lamellipodia (JAIL). JAIL extend over only a few microns and thus provide the basis for a subcellular regulation of adhesion. The local (subcellular) VE-cadherin concentration and JAIL formation are directly interdependent, which enables autoregulation. Therefore, this mechanism can contribute a subcellularly regulated adaptation of cell contact dynamics, and is therefore of great importance for monolayer integrity and relative cell migration during wound healing and angiogenesis, as well as for inflammatory responses. In this Review, we discuss the mechanisms and functions underlying these actin-driven protrusions and consider their contribution to the dynamic regulation of endothelial cell junctions.
Collapse
Affiliation(s)
- Jiahui Cao
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| |
Collapse
|
17
|
Dynamic of VE-cadherin-mediated spermatid-Sertoli cell contacts in the mouse seminiferous epithelium. Histochem Cell Biol 2018; 150:173-185. [PMID: 29797291 DOI: 10.1007/s00418-018-1682-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2018] [Indexed: 02/04/2023]
Abstract
Spermatids are haploid differentiating cells that, in the meantime they differentiate, translocate along the seminiferous epithelium towards the tubule lumen to be just released as spermatozoa. The success of such a migration depends on dynamic of spermatid-Sertoli cell contacts, the molecular nature of which has not been well defined yet. It was demonstrated that the vascular endothelial cadherin (VEC) is expressed transitorily in the mouse seminiferous epithelium. Here, we evaluated the pattern of VEC expression by immunohistochemistry first in seminiferous tubules at different stages of the epithelial cycle when only unique types of germ cell associations are present. Changes in the pattern of VEC localization according to the step of spermatid differentiation were analysed in detail using testis fragments and spontaneously released germ cells. Utilizing the first wave of spermatogenesis as an in vivo model to have at disposal spermatids at progressive steps of differentiation, we checked for level of looser VEC association with the membrane by performing protein solubilisation under mild detergent conditions and assays through VEC-immunoblotting. Being changes in VEC solubilisation paralleled in changes in phosphotyrosine (pY) content, we evaluated if spermatid VEC undergoes Y658 phosphorylation and if this correlates with VEC solubilisation and spermatid progression in differentiation. Altogether, our study shows a temporally restricted pattern of VEC expression that culminates with the presence of round spermatids to progressively decrease starting from spermatid elongation. Conversely, pY658-VEC signs elongating spermatids; its intracellular polarized compartmentalization suggests a possible involvement of pY658-VEC in the acquisition of spermatid cell polarity.
Collapse
|
18
|
Cioffi M, Gazzerro P, Di Finizio B, Vietri MT, Di Macchia C, Puca GA, Molinari AM. Serum-Soluble E-Cadherin Fragments in Lung Cancer. TUMORI JOURNAL 2018; 85:32-4. [PMID: 10228494 DOI: 10.1177/030089169908500107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Aims and background E-cadherin, also known as uvomorulin or cell-CAM 120/80, is one of the subclasses of cadherins, CA2+-dependent cell adhesion molecules. Several recent studies have suggested that loss of E-cadherin may be associated with tumor progression, such as in lung, gastric, hepatocellular, breast and prostatic carcinoma. Assessment of E-cadherin serum levels in lung cancer showed a relation to histologic type. Methods and study design Using an enzyme immunoassay, we determined E-cadherin serum levels in 79 patients affected with lung cancer (stage l–IV), 9 patients with breast cancer, 23 patients with different benign diseases, and 20 healthy patients. Results At a specificity level of 90%, E-cadherin diagnostic sensitivity was 66.6%, 47.6% and 43.7% in patients affected with squamous cell carcinoma, small cell carcinoma and adenocarcinoma, respectively. Conclusions Preliminary results suggest the use of serum E-cadherin as a prospective tumor marker.
Collapse
Affiliation(s)
- M Cioffi
- Istituto di Patologia Generale e Oncologia, Seconda Università degli Studi di Napoli, Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
19
|
Li W, Chen Z, Chin I, Chen Z, Dai H. The Role of VE-cadherin in Blood-brain Barrier Integrity Under Central Nervous System Pathological Conditions. Curr Neuropharmacol 2018; 16:1375-1384. [PMID: 29473514 PMCID: PMC6251046 DOI: 10.2174/1570159x16666180222164809] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 07/17/2017] [Accepted: 02/22/2018] [Indexed: 12/31/2022] Open
Abstract
The blood-brain barrier (BBB) is a layer between the blood circulation and neural tissue. It plays a pivotal role in maintaining the vulnerable extracellular microenvironment in the neuronal parenchyma. Neuroinflammatory events can result in BBB dysregulation by disturbing adherens junctions (AJs) and tight junctions (TJs). VE-cadherin, as one of the most important components of the vascular system, is specifically responsible for the assembly of AJs and BBB architecture. Here, we present a review, which highlights recently available insights into the relationship between the neuroinflammation and BBB dysregulation. We then explore the specific interaction between VE-cadherin and BBB. Finally, we discuss the changes of VE-cadherin with different neurological diseases from both experimental and clinical studies. An understanding of VE-cadherin in BBB regulation may indicate that VE-cadherin can partially be a biomarker of neuroinflammation disease and lead to novel approaches for abating BBB dysregulation under pathological conditions and the opening of the BBB following central nervous system (CNS) drug delivery.
Collapse
Affiliation(s)
| | | | | | - Zhong Chen
- Address correspondence to these authors at the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, P.R. China; Tel/Fax: +86-571-87783891; E-mail: ; and Depart-ment of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P.R. China; E-mail:
| | - Haibin Dai
- Address correspondence to these authors at the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, P.R. China; Tel/Fax: +86-571-87783891; E-mail: ; and Depart-ment of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, P.R. China; E-mail:
| |
Collapse
|
20
|
Fathi F, Rezabakhsh A, Rahbarghazi R, Rashidi MR. Early-stage detection of VE-cadherin during endothelial differentiation of human mesenchymal stem cells using SPR biosensor. Biosens Bioelectron 2017; 96:358-366. [DOI: 10.1016/j.bios.2017.05.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 05/03/2017] [Accepted: 05/09/2017] [Indexed: 02/08/2023]
|
21
|
Schnittler H. Contraction of endothelial cells: 40 years of research, but the debate still lives. Histochem Cell Biol 2016; 146:651-656. [PMID: 27680546 DOI: 10.1007/s00418-016-1501-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2016] [Indexed: 12/23/2022]
Abstract
Force generation in non-muscle cells is vital for many cellular and tissue functions. Force-generating mechanisms include actomyosin-mediated contraction, actin polymerization that drives plasma membrane protrusions and filopodia as well as kinesin- and dynein-controlled transport of vesicles and organelles along the microtubule cytoskeleton. The actomyosin-mediated contractility and actin remodeling in both epithelium and endothelium were shown to have significant impact on cell migration, shape change and formation and control of intercellular junctions. In endothelium, contraction is supposed to control permeability for fluid and solutes. However, recent studies demonstrated the constitutive appearance of junction-associated intermittent lamellipodia (JAIL) that drive vascular endothelial cadherin (VE-cadherin) dynamics and control endothelial permeability. Since thrombin blocks JAIL formation and thus increases endothelial permeability, the concept of a simple Rho GTPase-controlled contraction, which is supposed to open endothelial junctions, becomes challenged. Furthermore, specific tyrosine phosphorylation sites of VE-cadherin and catenins have been shown to be involved in control of VE-cadherin-mediated cell adhesion. How the causal-mechanistic interdependency between contractility, VE-cadherin and catenin phosphorylation and JAIL-mediated dynamic remodeling of VE-cadherin is regulated is still an open question and needs to be further addressed.
Collapse
Affiliation(s)
- Hans Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany.
| |
Collapse
|
22
|
Seebach J, Cao J, Schnittler HJ. Quantitative dynamics of VE-cadherin at endothelial cell junctions at a glance: basic requirements and current concepts. Discoveries (Craiova) 2016; 4:e63. [PMID: 32309583 PMCID: PMC7159836 DOI: 10.15190/d.2016.10] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Intercellular junctions of the vascular endothelium are dynamic structures that display a high degree of plasticity, which is required to contribute to their regulation of many physiological and pathological processes including monolayer integrity, barrier function, wound healing and angiogenesis. Vascular endothelial cadherin (VE-cadherin) is connected via catenins to the actin cytoskeleton, both of which are key structures in endothelial junction regulation, and thus are the focus of much investigation. Fluorescence-based live cell imaging is the method of choice to study dynamic remodeling in living cells. Although these methods have been successfully applied to many cell types, investigations of endothelial junction dynamics were for a long time limited as they are largely resistant to transfection using many classical protocols. Application of virus-based gene transduction techniques, together with advanced microscopy, now allows both sufficient expression of fluorescence tagged junction-localized proteins in the endothelium and time-lapse recording over long periods. Using highly spatiotemporally resolved fluorescence microscopy it turned out that endothelial junctions display extensive junction heterogeneity at the subcellular level; a fact that largely limits automated quantification by available software. Recent work describes open software tools to quantitatively analyze large amounts of fluorescence-based image data in either single or confluent epithelial and endothelial cells. Based on quantitative VE-cadherin and actin dynamics novel key players, mechanisms and concepts have been suggested that control endothelial junction dynamics. Here we aim to summarize the recent developments in the field.
Collapse
Affiliation(s)
- Jochen Seebach
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| | - Jiahui Cao
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| | - Hans Joachim Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| |
Collapse
|
23
|
Brinkmann BF, Steinbacher T, Hartmann C, Kummer D, Pajonczyk D, Mirzapourshafiyi F, Nakayama M, Weide T, Gerke V, Ebnet K. VE-cadherin interacts with cell polarity protein Pals1 to regulate vascular lumen formation. Mol Biol Cell 2016; 27:2811-21. [PMID: 27466317 PMCID: PMC5025268 DOI: 10.1091/mbc.e16-02-0127] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/19/2016] [Indexed: 02/06/2023] Open
Abstract
Blood vessel tubulogenesis requires the establishment of apicobasal polarity of endothelial cells. A novel interaction is described of the cell adhesion molecule VE-cadherin with the cell polarity protein Pals1. The activity of VE-cadherin in regulation of endothelial lumen formation depends on its interaction with both Pals1 and Par3. Blood vessel tubulogenesis requires the formation of stable cell-to-cell contacts and the establishment of apicobasal polarity of vascular endothelial cells. Cell polarity is regulated by highly conserved cell polarity protein complexes such as the Par3-aPKC-Par6 complex and the CRB3-Pals1-PATJ complex, which are expressed by many different cell types and regulate various aspects of cell polarity. Here we describe a functional interaction of VE-cadherin with the cell polarity protein Pals1. Pals1 directly interacts with VE-cadherin through a membrane-proximal motif in the cytoplasmic domain of VE-cadherin. VE-cadherin clusters Pals1 at cell–cell junctions. Mutating the Pals1-binding motif in VE-cadherin abrogates the ability of VE-cadherin to regulate apicobasal polarity and vascular lumen formation. In a similar way, deletion of the Par3-binding motif at the C-terminus of VE-cadherin impairs apicobasal polarity and vascular lumen formation. Our findings indicate that the biological activity of VE-cadherin in regulating endothelial polarity and vascular lumen formation is mediated through its interaction with the two cell polarity proteins Pals1 and Par3.
Collapse
Affiliation(s)
- Benjamin F Brinkmann
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| | - Tim Steinbacher
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany
| | - Christian Hartmann
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| | - Daniel Kummer
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| | - Denise Pajonczyk
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany
| | - Fatemeh Mirzapourshafiyi
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Masanori Nakayama
- Laboratory for Cell Polarity and Organogenesis, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - Thomas Weide
- Department of Internal Medicine D, Division of Molecular Nephrology, University Hospital Münster, Albert-Schweitzer-Campus 1, University of Münster, 48419 Münster, Germany
| | - Volker Gerke
- Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, 48419 Münster, Germany
| | - Klaus Ebnet
- Institute-Associated Research Group "Cell Adhesion and Cell Polarity,", University of Münster, 48419 Münster, Germany Institute of Medical Biochemistry, Center for Molecular Biology of Inflammation, University of Münster, 48419 Münster, Germany Interdisciplinary Clinical Research Center, University of Münster, 48419 Münster, Germany
| |
Collapse
|
24
|
Root AR, Cao W, Li B, LaPan P, Meade C, Sanford J, Jin M, O'Sullivan C, Cummins E, Lambert M, Sheehan AD, Ma W, Gatto S, Kerns K, Lam K, D'Antona AM, Zhu L, Brady WA, Benard S, King A, He T, Racie L, Arai M, Barrett D, Stochaj W, LaVallie ER, Apgar JR, Svenson K, Mosyak L, Yang Y, Chichili GR, Liu L, Li H, Burke S, Johnson S, Alderson R, Finlay WJJ, Lin L, Olland S, Somers W, Bonvini E, Gerber HP, May C, Moore PA, Tchistiakova L, Bloom L. Development of PF-06671008, a Highly Potent Anti-P-cadherin/Anti-CD3 Bispecific DART Molecule with Extended Half-Life for the Treatment of Cancer. Antibodies (Basel) 2016; 5:E6. [PMID: 31557987 PMCID: PMC6698862 DOI: 10.3390/antib5010006] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/14/2016] [Accepted: 02/17/2016] [Indexed: 01/13/2023] Open
Abstract
Bispecific antibodies offer a promising approach for the treatment of cancer but can be challenging to engineer and manufacture. Here we report the development of PF-06671008, an extended-half-life dual-affinity re-targeting (DART®) bispecific molecule against P-cadherin and CD3 that demonstrates antibody-like properties. Using phage display, we identified anti-P-cadherin single chain Fv (scFv) that were subsequently affinity-optimized to picomolar affinity using stringent phage selection strategies, resulting in low picomolar potency in cytotoxic T lymphocyte (CTL) killing assays in the DART format. The crystal structure of this disulfide-constrained diabody shows that it forms a novel compact structure with the two antigen binding sites separated from each other by approximately 30 Å and facing approximately 90° apart. We show here that introduction of the human Fc domain in PF-06671008 has produced a molecule with an extended half-life (-4.4 days in human FcRn knock-in mice), high stability (Tm1 > 68 °C), high expression (>1 g/L), and robust purification properties (highly pure heterodimer), all with minimal impact on potency. Finally, we demonstrate in vivo anti-tumor efficacy in a human colorectal/human peripheral blood mononuclear cell (PBMC) co-mix xenograft mouse model. These results suggest PF-06671008 is a promising new bispecific for the treatment of patients with solid tumors expressing P-cadherin.
Collapse
Affiliation(s)
- Adam R Root
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Wei Cao
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Bilian Li
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Peter LaPan
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Caryl Meade
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Jocelyn Sanford
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Macy Jin
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Cliona O'Sullivan
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Emma Cummins
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Matthew Lambert
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Alfredo D Sheehan
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Weijun Ma
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Scott Gatto
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Kelvin Kerns
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Khetemenee Lam
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Aaron M D'Antona
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Lily Zhu
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - William A Brady
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Susan Benard
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Amy King
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Tao He
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Lisa Racie
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Maya Arai
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Dianah Barrett
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Wayne Stochaj
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Edward R LaVallie
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - James R Apgar
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Kristine Svenson
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Lidia Mosyak
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Yinhua Yang
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | | | - Liqin Liu
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Hua Li
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Steve Burke
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Syd Johnson
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Ralph Alderson
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - William J J Finlay
- Global Biotherapeutics Technologies, Pfizer Inc., Grange Castle Business Park, Clondalkin, Dublin 22, Ireland.
| | - Laura Lin
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Stéphane Olland
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - William Somers
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Ezio Bonvini
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Hans-Peter Gerber
- Oncology Research Unit, Pfizer Inc., 401 N. Middletown Road, Pearl River, NY 10965, USA.
| | - Chad May
- Oncology Research Unit, Pfizer Inc., 401 N. Middletown Road, Pearl River, NY 10965, USA.
| | - Paul A Moore
- MacroGenics Inc., 9640 Medical Center Drive, Rockville, MD 20850, USA.
| | - Lioudmila Tchistiakova
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| | - Laird Bloom
- Global Biotherapeutics Technologies, Pfizer Inc., 610 Main St., Cambridge, MA 02139, USA.
| |
Collapse
|
25
|
Regulation of Endothelial Adherens Junctions by Tyrosine Phosphorylation. Mediators Inflamm 2015; 2015:272858. [PMID: 26556953 PMCID: PMC4628659 DOI: 10.1155/2015/272858] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 08/16/2015] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells form a semipermeable, regulated barrier that limits the passage of fluid, small molecules, and leukocytes between the bloodstream and the surrounding tissues. The adherens junction, a major mechanism of intercellular adhesion, is comprised of transmembrane cadherins forming homotypic interactions between adjacent cells and associated cytoplasmic catenins linking the cadherins to the cytoskeleton. Inflammatory conditions promote the disassembly of the adherens junction and a loss of intercellular adhesion, creating openings or gaps in the endothelium through which small molecules diffuse and leukocytes transmigrate. Tyrosine kinase signaling has emerged as a central regulator of the inflammatory response, partly through direct phosphorylation and dephosphorylation of the adherens junction components. This review discusses the findings that support and those that argue against a direct effect of cadherin and catenin phosphorylation in the disassembly of the adherens junction. Recent findings indicate a complex interaction between kinases, phosphatases, and the adherens junction components that allow a fine regulation of the endothelial permeability to small molecules, leukocyte migration, and barrier resealing.
Collapse
|
26
|
Seebach J, Taha AA, Lenk J, Lindemann N, Jiang X, Brinkmann K, Bogdan S, Schnittler HJ. The CellBorderTracker, a novel tool to quantitatively analyze spatiotemporal endothelial junction dynamics at the subcellular level. Histochem Cell Biol 2015; 144:517-32. [PMID: 26275669 DOI: 10.1007/s00418-015-1357-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/11/2015] [Indexed: 11/28/2022]
Abstract
Endothelial junctions are dynamic structures organized by multi-protein complexes that control monolayer integrity, homeostasis, inflammation, cell migration and angiogenesis. Newly developed methods for both the genetic manipulation of endothelium and microscopy permit time-lapse recordings of fluorescent proteins over long periods of time. Quantitative data analyses require automated methods. We developed a software package, the CellBorderTracker, allowing quantitative analysis of fluorescent-tagged cell junction protein dynamics in time-lapse sequences. The CellBorderTracker consists of the CellBorderExtractor that segments cells and identifies cell boundaries and mapping tools for data extraction. The tool is illustrated by analyzing fluorescent-tagged VE-cadherin the backbone of adherence junctions in endothelium. VE-cadherin displays high dynamics that is forced by junction-associated intermittent lamellipodia (JAIL) that are actin driven and WASP/ARP2/3 complex controlled. The manual segmentation and the automatic one agree to 90 %, a value that indicates high reliability. Based on segmentations, different maps were generated allowing more detailed data extraction. This includes the quantification of protein distribution pattern, the generation of regions of interest, junction displacements, cell shape changes, migration velocities and the visualization of junction dynamics over many hours. Furthermore, we demonstrate an advanced kymograph, the J-kymograph that steadily follows irregular cell junction dynamics in time-lapse sequences for individual junctions at the subcellular level. By using the CellBorderTracker, we demonstrate that VE-cadherin dynamics is quickly arrested upon thrombin stimulation, a phenomenon that was largely due to transient inhibition of JAIL and display a very heterogeneous subcellular and divers VE-cadherin dynamics during intercellular gap formation and resealing.
Collapse
Affiliation(s)
- Jochen Seebach
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany.
| | - Abdallah Abu Taha
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
| | - Janine Lenk
- Faculty of Medicine Carl Gustav Carus, Fetscherstrasse 74, 01307, Dresden, Germany
| | - Nico Lindemann
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany
| | - Xiaoyi Jiang
- Department of Computer Science, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Klaus Brinkmann
- Institute for Neurobiology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Sven Bogdan
- Institute for Neurobiology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Hans-Joachim Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Vesaliusweg 2-4, 48149, Münster, Germany.
| |
Collapse
|
27
|
Abu Taha A, Schnittler HJ. Dynamics between actin and the VE-cadherin/catenin complex: novel aspects of the ARP2/3 complex in regulation of endothelial junctions. Cell Adh Migr 2015; 8:125-35. [PMID: 24621569 DOI: 10.4161/cam.28243] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Endothelial adherens junctions are critical for physiological and pathological processes such as differentiation, maintenance of entire monolayer integrity, and the remodeling. The endothelial-specific VE-cadherin/catenin complex provides the backbone of adherens junctions and acts in close interaction with actin filaments and actin/myosin-mediated contractility to fulfill the junction demands. The functional connection between the cadherin/catenin complex and actin filaments might be either directly through ?-catenins, or indirectly e.g., via linker proteins such as vinculin, p120ctn, ?-actinin, or EPLIN. However, both junction integrity and dynamic remodeling have to be contemporarily coordinated. The actin-related protein complex ARP2/3 and its activating molecules, such as N-WASP and WAVE, have been shown to regulate the lammellipodia-mediated formation of cell junctions in both epithelium and endothelium. Recent reports now demonstrate a novel aspect of the ARP2/3 complex and the nucleating-promoting factors in the maintenance of endothelial barrier function and junction remodeling of established endothelial cell junctions. Those mechanisms open novel possibilities; not only in fulfilling physiological demands but obtained information may be of critical importance in pathologies such as wound healing, angiogenesis, inflammation, and cell diapedesis.
Collapse
Affiliation(s)
- Abdallah Abu Taha
- Institute of Anatomy & Vascular Biology; WWU-Münster, Vesaliusweg 2-4; Münster, Germany
| | - Hans-J Schnittler
- Institute of Anatomy & Vascular Biology; WWU-Münster, Vesaliusweg 2-4; Münster, Germany
| |
Collapse
|
28
|
Miyamoto Y, Sakane F, Hashimoto K. N-cadherin-based adherens junction regulates the maintenance, proliferation, and differentiation of neural progenitor cells during development. Cell Adh Migr 2015; 9:183-92. [PMID: 25869655 DOI: 10.1080/19336918.2015.1005466] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
This review addresses our current understanding of the regulatory mechanism by which N-cadherin, a classical cadherin, affects neural progenitor cells (NPCs) during development. N-cadherin is responsible for the integrity of adherens junctions (AJs), which develop in the sub-apical region of NPCs in the neural tube and brain cortex. The apical domain, which contains the sub-apical region, is involved in the switching from symmetric proliferative division to asymmetric neurogenic division of NPCs. In addition, N-cadherin-based AJ is deeply involved in the apico-basal polarity of NPCs and the regulation of Wnt-β-catenin, hedgehog (Hh), and Notch signaling. In this review, we discuss the roles of N-cadherin in the maintenance, proliferation, and differentiation of NPCs through components of AJ, β-catenin and αE-catenin.
Collapse
Key Words
- AJ, adherens junction
- EC, extracellular
- Fox, forkhead box
- Frz, frizzled
- GFAP, glial fibrillary acidic protein
- GSK3β, glycogen synthase kinase 3β
- Hes, hairly/enhancer of split
- Hh, hedgehog
- IP, intermediate progenitor
- KO, knockout
- LEF, lymphocyte enhancer factor
- N-cadherin
- NPC, neural progenitor cell
- Par, partition defective complex protein
- Ptc, Pached
- Smo, smoothened
- Sox2, sry (sex determining region Y)-box containing gene 2
- TA cell, transient amplifying cell; ZO-1, Zonula Occludens-1.
- TCF, T-cell factor
- aPKC, atypical protein kinase C
- adherens junction
- apico-basal polarity
- iPSC, induced pluripotent stem cell
- neural progenitor cells
- ngn2, neurogenin 2
- shRNA, short hairpin RNA
- β-catenin
Collapse
Affiliation(s)
- Yasunori Miyamoto
- a The Graduate School of Humanities and Sciences; Ochanomizu University ; Tokyo , Japan
| | | | | |
Collapse
|
29
|
Abstract
During brain development, billions of neurons organize into highly specific circuits. To form specific circuits, neurons must build the appropriate types of synapses with appropriate types of synaptic partners while avoiding incorrect partners in a dense cellular environment. Defining the cellular and molecular rules that govern specific circuit formation has significant scientific and clinical relevance because fine scale connectivity defects are thought to underlie many cognitive and psychiatric disorders. Organizing specific neural circuits is an enormously complicated developmental process that requires the concerted action of many molecules, neural activity, and temporal events. This review focuses on one class of molecules postulated to play an important role in target selection and specific synapse formation: the classic cadherins. Cadherins have a well-established role in epithelial cell adhesion, and although it has long been appreciated that most cadherins are expressed in the brain, their role in synaptic specificity is just beginning to be unraveled. Here, we review past and present studies implicating cadherins as active participants in the formation, function, and dysfunction of specific neural circuits and pose some of the major remaining questions.
Collapse
Affiliation(s)
- Raunak Basu
- a Department of Neurobiology and Anatomy ; University of Utah ; Salt Lake City , UT USA
| | | | | |
Collapse
|
30
|
Whole exome sequencing in females with autism implicates novel and candidate genes. Int J Mol Sci 2015; 16:1312-35. [PMID: 25574603 PMCID: PMC4307305 DOI: 10.3390/ijms16011312] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 12/31/2014] [Indexed: 01/17/2023] Open
Abstract
Classical autism or autistic disorder belongs to a group of genetically heterogeneous conditions known as Autism Spectrum Disorders (ASD). Heritability is estimated as high as 90% for ASD with a recently reported compilation of 629 clinically relevant candidate and known genes. We chose to undertake a descriptive next generation whole exome sequencing case study of 30 well-characterized Caucasian females with autism (average age, 7.7 ± 2.6 years; age range, 5 to 16 years) from multiplex families. Genomic DNA was used for whole exome sequencing via paired-end next generation sequencing approach and X chromosome inactivation status. The list of putative disease causing genes was developed from primary selection criteria using machine learning-derived classification score and other predictive parameters (GERP2, PolyPhen2, and SIFT). We narrowed the variant list to 10 to 20 genes and screened for biological significance including neural development, function and known neurological disorders. Seventy-eight genes identified met selection criteria ranging from 1 to 9 filtered variants per female. Five females presented with functional variants of X-linked genes (IL1RAPL1, PIR, GABRQ, GPRASP2, SYTL4) with cadherin, protocadherin and ankyrin repeat gene families most commonly altered (e.g., CDH6, FAT2, PCDH8, CTNNA3, ANKRD11). Other genes related to neurogenesis and neuronal migration (e.g., SEMA3F, MIDN), were also identified.
Collapse
|
31
|
|
32
|
Wright RD, Cooper D. Glycobiology of leukocyte trafficking in inflammation. Glycobiology 2014; 24:1242-51. [DOI: 10.1093/glycob/cwu101] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
33
|
Bravi L, Dejana E, Lampugnani MG. VE-cadherin at a glance. Cell Tissue Res 2014; 355:515-22. [PMID: 24643676 DOI: 10.1007/s00441-014-1843-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 02/04/2014] [Indexed: 10/25/2022]
Abstract
Although being a monolayer the vascular endothelium controls fundamental vessel functions such as permeability, leukocyte extravasation and angiogenesis. The endothelial selective transmembrane constituent of adherens junctions, Vascular Endothelial- (VE-) cadherin plays a crucial role in the regulation of such activities. The signaling pathways controlled by VE-cadherin as well as the ones that regulate VE-cadherin activity start to be elucidated. This delineates a complex network of molecular and functional interactions that can be altered in pathologies.
Collapse
Affiliation(s)
- Luca Bravi
- FIRC Institute of Molecular Oncology (IFOM) Fondazione, Via Adamello 16, 20139, Milan, Italy
| | | | | |
Collapse
|
34
|
Abu Taha A, Taha M, Seebach J, Schnittler HJ. ARP2/3-mediated junction-associated lamellipodia control VE-cadherin-based cell junction dynamics and maintain monolayer integrity. Mol Biol Cell 2013; 25:245-56. [PMID: 24227887 PMCID: PMC3890345 DOI: 10.1091/mbc.e13-07-0404] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The ARP2/3 complex controls junction-associated intermittent lamellipodia (JAIL), which trigger VE-cadherin adhesion and dynamics. JAIL formation maintains paraendothelial barrier function under physiological conditions and depends on the local VE-cadherin concentration. Maintenance and remodeling of endothelial cell junctions critically depend on the VE-cadherin/catenin complex and its interaction with the actin filament cytoskeleton. Here we demonstrate that local lack of vascular endothelial (VE)-cadherin at established cell junctions causes actin-driven and actin-related protein 2/3 complex (ARP2/3)–controlled lamellipodia to appear intermittently at those sites. Lamellipodia overlap the VE-cadherin–free adjacent plasma membranes and facilitate formation of new VE-cadherin adhesion sites, which quickly move into the junctions, driving VE-cadherin dynamics and remodeling. Inhibition of the ARP2/3 complex by expression of the N-WASP (V)CA domain or application of two ARP2/3 inhibitors, CK-548 and CK-666, blocks VE-cadherin dynamics and causes intercellular gaps. Furthermore, expression of carboxy-terminal–truncated VE-cadherin increases the number of ARP2/3-controlled lamellipodia, whereas overexpression of wild-type VE-cadherin largely blocks it and decreases cell motility. The data demonstrate a functional interrelationship between VE-cadherin–mediated cell adhesion and actin-driven, ARP2/3-controlled formation of new VE-cadherin adhesion sites via intermittently appearing lamellipodia at established cell junctions. This coordinated mechanism controls VE-cadherin dynamics and cell motility and maintains monolayer integrity, thus potentially being relevant in disease and angiogenesis.
Collapse
Affiliation(s)
- Abdallah Abu Taha
- Institute of Anatomy and Vascular Biology, WWU-Münster, 48149 Münster, Germany
| | | | | | | |
Collapse
|
35
|
Paolinelli R, Corada M, Ferrarini L, Devraj K, Artus C, Czupalla CJ, Rudini N, Maddaluno L, Papa E, Engelhardt B, Couraud PO, Liebner S, Dejana E. Wnt activation of immortalized brain endothelial cells as a tool for generating a standardized model of the blood brain barrier in vitro. PLoS One 2013; 8:e70233. [PMID: 23940549 PMCID: PMC3734070 DOI: 10.1371/journal.pone.0070233] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 06/18/2013] [Indexed: 02/02/2023] Open
Abstract
Reproducing the characteristics and the functional responses of the blood–brain barrier (BBB) in vitro represents an important task for the research community, and would be a critical biotechnological breakthrough. Pharmaceutical and biotechnology industries provide strong demand for inexpensive and easy-to-handle in vitro BBB models to screen novel drug candidates. Recently, it was shown that canonical Wnt signaling is responsible for the induction of the BBB properties in the neonatal brain microvasculature in vivo. In the present study, following on from earlier observations, we have developed a novel model of the BBB in vitro that may be suitable for large scale screening assays. This model is based on immortalized endothelial cell lines derived from murine and human brain, with no need for co-culture with astrocytes. To maintain the BBB endothelial cell properties, the cell lines are cultured in the presence of Wnt3a or drugs that stabilize β-catenin, or they are infected with a transcriptionally active form of β-catenin. Upon these treatments, the cell lines maintain expression of BBB-specific markers, which results in elevated transendothelial electrical resistance and reduced cell permeability. Importantly, these properties are retained for several passages in culture, and they can be reproduced and maintained in different laboratories over time. We conclude that the brain-derived endothelial cell lines that we have investigated gain their specialized characteristics upon activation of the canonical Wnt pathway. This model may be thus suitable to test the BBB permeability to chemicals or large molecular weight proteins, transmigration of inflammatory cells, treatments with cytokines, and genetic manipulation.
Collapse
Affiliation(s)
| | - Monica Corada
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Luca Ferrarini
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Kavi Devraj
- Institute of Neurology (Edinger Institute), Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Cédric Artus
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Cathrin J. Czupalla
- Institute of Neurology (Edinger Institute), Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Noemi Rudini
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Luigi Maddaluno
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | - Eleanna Papa
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | | | - Pierre Olivier Couraud
- Inserm, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Stefan Liebner
- Institute of Neurology (Edinger Institute), Johann Wolfgang Goethe University, Frankfurt, Germany
| | - Elisabetta Dejana
- IFOM-FIRC Institute of Molecular Oncology Foundation, Milan, Italy
- Department of Biosciences, University of Milan, Milan, Italy
- * E-mail:
| |
Collapse
|
36
|
Law AYS, Wong CKC. Stanniocalcin-1 and -2 promote angiogenic sprouting in HUVECs via VEGF/VEGFR2 and angiopoietin signaling pathways. Mol Cell Endocrinol 2013; 374:73-81. [PMID: 23664860 DOI: 10.1016/j.mce.2013.04.024] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 03/27/2013] [Accepted: 04/26/2013] [Indexed: 12/15/2022]
Abstract
The members of stanniocalcins (STCs: STC-1 and STC-2) family are known to be involved in tumor progression and metastasis. Although current evidences suggest the involvement of STCs in vascular biology, the functional roles of STCs in angiogenesis have not yet been elucidated. The objective of this study was to decipher the roles of STCs in angiogenesis of human umbilical vascular endothelial cells (HUVECs). We prepared STC1 or STC2 lentiviral particles to transduce the cells to reveal their effects on the processes of cell proliferation, migration and tube formation. The stimulatory effects of STCs on these processes were demonstrated, supporting the notion of STCs in angiogenesis. To dissect the molecular components involved, STC1 or STC2 transduction led to significant increases in the expression levels of cell cycle regulators (i.e. cyclin-D and phospho-retinoblastoma), matrix metalloproteinase (MMP)-2 but a decrease of tissue inhibitors of metalloproteases (TIMP)-1. The expression levels of the cell adhesion/junctional proteins vimentin and VE-cadherin, were significantly induced. Moreover the transduction induced both mRNA and protein levels of eNOS, VEGF and VEGFR2 (KDR mRNA and pKDR), highlighting the stimulatory effects of STCs on VEGF-signaling pathway. Furthermore STC2 transduction but not STC1, activated angiopoietin (Ang)-2 pathway. Taken together, STC1 and STC2 play positive roles in angiogenic sprouting. The action of STC1 was mediated via VEGF/VEGFR2 pathway while STC2 were mediated via VEGF/VEGFR2 and Ang-2 pathways.
Collapse
Affiliation(s)
- Alice Y S Law
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | | |
Collapse
|
37
|
Paulson AF, Prasad MS, Thuringer AH, Manzerra P. Regulation of cadherin expression in nervous system development. Cell Adh Migr 2013; 8:19-28. [PMID: 24526207 DOI: 10.4161/cam.27839] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This review addresses our current understanding of the regulatory mechanisms for classical cadherin expression during development of the vertebrate nervous system. The complexity of the spatial and temporal expression patterns is linked to morphogenic and functional roles in the developing nervous system. While the regulatory networks controlling cadherin expression are not well understood, it is likely that the multiple signaling pathways active in the development of particular domains also regulate the specific cadherins expressed at that time and location. With the growing understanding of the broader roles of cadherins in cell-cell adhesion and non-adhesion processes, it is important to understand both the upstream regulation of cadherin expression and the downstream effects of specific cadherins within their cellular context.
Collapse
Affiliation(s)
- Alicia F Paulson
- Division of Basic Biomedical Sciences; Sanford School of Medicine of The University of South Dakota; Vermillion, SD USA
| | - Maneeshi S Prasad
- Department of Molecular Biosciences; Northwestern University; Evanston, IL USA
| | | | - Pasquale Manzerra
- Division of Basic Biomedical Sciences; Sanford School of Medicine of The University of South Dakota; Vermillion, SD USA
| |
Collapse
|
38
|
Roussoulières A, McGregor B, Chalabreysse L, Cerutti C, Garnier JL, Boissonnat P, Bastien O, Scoazec JY, Thivolet-Bejui F, Sebbag L, L. McGregor J. Expression of VE-Cadherin in Peritubular Endothelial Cells during Acute Rejection after Human Renal Transplantation. J Biomed Biotechnol 2012; 2007:41705. [PMID: 17710242 PMCID: PMC1940055 DOI: 10.1155/2007/41705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2006] [Accepted: 05/10/2007] [Indexed: 12/11/2022] Open
Abstract
Genes involved in acute rejection (AR) after organ transplantation remain to be further elucidated. In a previous work we have demonstrated the under-expression of VE-Cadherin by endothelial cells (EC) in AR following murine and human heart transplantation. Serial sections from 15 human kidney Banff-graded transplant biopsies were examined for the presence of VE-Cadherin and CD34 staining by immunohistochemistry (no AR (n = 5), AR grade IA (n = 5), or AR grade IIA (n = 5)).
Quantification of peritubular EC staining were evaluated and results were expressed by the percentage of stained cells per surface analysed. There was no difference in CD34 staining between the 3 groups. VE-Cadherin expression was significantly reduced in AR Grade IIA when compared to no AR (P = .01) and to AR grade IA (P = .02). This study demonstrates a reduced VE-Cadherin expression by EC in AR after renal transplantation. The down-regulation of VE-Cadherin may strongly participate in human AR.
Collapse
Affiliation(s)
- Ana Roussoulières
- INSERM U331/EA 3740, Faculté de Médecine RTH Laennec, 8 rue Guillaume Paradin, 69003 Lyon, France
- Department of Cardiac Transplantation, Hôpital Cardiologique Louis Pradel, 28 avenue du Doyen Lépine, 69003 Lyon, France
- *Ana Roussoulières:
| | - Brigitte McGregor
- Department of Pathology, Hôpital Edouard Herriot, 5 place d'Arsonval, 69003 Lyon, France
| | - Lara Chalabreysse
- Department of Pathology, Hôpital Cardiologique Louis Pradel, 28 avenue du 4 Doyen Lépine, 69003 Lyon, France
| | - Catherine Cerutti
- INSERM U331/EA 3740, Faculté de Médecine RTH Laennec, 8 rue Guillaume Paradin, 69003 Lyon, France
| | - Jeanne-Luce Garnier
- Department of Nephrology, Hôpital Edouard Herriot, 5 place d'Arsonval, 69003 Lyon, France
| | - Pascale Boissonnat
- Department of Cardiac Transplantation, Hôpital Cardiologique Louis Pradel, 28 avenue du Doyen Lépine, 69003 Lyon, France
| | - Olivier Bastien
- Department of Cardiac Transplantation, Hôpital Cardiologique Louis Pradel, 28 avenue du Doyen Lépine, 69003 Lyon, France
| | - Jean-Yves Scoazec
- Department of Pathology, Hôpital Edouard Herriot, 5 place d'Arsonval, 69003 Lyon, France
| | - Françoise Thivolet-Bejui
- Department of Pathology, Hôpital Cardiologique Louis Pradel, 28 avenue du 4 Doyen Lépine, 69003 Lyon, France
| | - Laurent Sebbag
- Department of Cardiac Transplantation, Hôpital Cardiologique Louis Pradel, 28 avenue du Doyen Lépine, 69003 Lyon, France
| | - John L. McGregor
- INSERM U331/EA 3740, Faculté de Médecine RTH Laennec, 8 rue Guillaume Paradin, 69003 Lyon, France
- Centre for Cardiovascular Biology & Medicine, King's College London, Strand WC2R 2LS, UK
| |
Collapse
|
39
|
Abstract
Cadherins are Ca(2+)-dependent cell-cell adhesion molecules that play critical roles in animal morphogenesis. Various cadherin-related molecules have also been identified, which show diverse functions, not only for the regulation of cell adhesion but also for that of cell proliferation and planar cell polarity. During the past decade, understanding of the roles of these molecules in the nervous system has significantly progressed. They are important not only for the development of the nervous system but also for its functions and, in turn, for neural disorders. In this review, we discuss the roles of cadherins and related molecules in neural development and function in the vertebrate brain.
Collapse
Affiliation(s)
- Shinji Hirano
- Department of Neurobiology and Anatomy, Kochi Medical School, Okoh-cho Kohasu, Nankoku-City 783–8505, Japan.
| | | |
Collapse
|
40
|
Structural and functional characterization of two alternative splicing variants of mouse Endothelial Cell-Specific Chemotaxis Regulator (ECSCR). Int J Mol Sci 2012; 13:4920-4936. [PMID: 22606020 PMCID: PMC3344256 DOI: 10.3390/ijms13044920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/19/2012] [Accepted: 03/20/2012] [Indexed: 01/29/2023] Open
Abstract
Endothelial cells (ECs) that line the lumen of blood vessels are important players in blood vessel formation, and EC migration is a key component of the angiogenic process. Thus, identification of genes that are specifically or preferentially expressed in vascular ECs and in-depth understanding of their biological functions may lead to discovery of new therapeutic targets. We have previously reported molecular characterization of human endothelial cell-specific molecule 2 (ECSM2)/endothelial cell-specific chemotaxis regulator (ECSCR). In the present study, we cloned two mouse full-length cDNAs by RT-PCR, which encode two putative ECSCR isoform precursors with considerable homology to the human ECSCR. Nucleotide sequence and exon-intron junction analyses suggested that they are alternative splicing variants (ECSCR isoform-1 and -2), differing from each other in the first and second exons. Quantitative RT-PCR results revealed that isoform-2 is the predominant form, which was most abundant in heart, lung, and muscles, and moderately abundant in uterus and testis. In contrast, the expression of isoform-1 seemed to be more enriched in testis. To further explore their potential cellular functions, we expressed GFP- and FLAG-tagged ECSCR isoforms, respectively, in an ECSCR deficient cell line (HEK293). Interestingly, the actual sizes of either ECSCR-GFP or -FLAG fusion proteins detected by immunoblotting are much larger than their predicted sizes, suggesting that both isoforms are glycoproteins. Fluorescence microscopy revealed that both ECSCR isoforms are localized at the cell surface, which is consistent with the structural prediction. Finally, we performed cell migration assays using mouse endothelial MS1 cells overexpressing GFP alone, isoform-1-GFP, and isoform-2-GFP, respectively. Our results showed that both isoforms significantly inhibited vascular epidermal growth factor (VEGF)-induced cell migration. Taken together, we have provided several lines of experimental evidence that two mouse ECSCR splicing variants/isoform precursors exist. They are differentially expressed in a variety of tissue types and likely involved in modulation of vascular EC migration. We have also defined the gene structure of mouse ECSCR using bioinformatics tools, which provides new information towards a better understanding of alternative splicing of ECSCR.
Collapse
|
41
|
Brasch J, Harrison OJ, Ahlsen G, Carnally SM, Henderson RM, Honig B, Shapiro L. Structure and binding mechanism of vascular endothelial cadherin: a divergent classical cadherin. J Mol Biol 2011; 408:57-73. [PMID: 21269602 PMCID: PMC3084036 DOI: 10.1016/j.jmb.2011.01.031] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 01/11/2011] [Accepted: 01/13/2011] [Indexed: 10/18/2022]
Abstract
Vascular endothelial cadherin (VE-cadherin), a divergent member of the type II classical cadherin family of cell adhesion proteins, mediates homophilic adhesion in the vascular endothelium. Previous investigations with a bacterially produced protein suggested that VE-cadherin forms cell surface trimers that bind between apposed cells to form hexamers. Here we report studies of mammalian-produced VE-cadherin ectodomains suggesting that, like other classical cadherins, VE-cadherin forms adhesive trans dimers between monomers located on opposing cell surfaces. Trimerization of the bacterially produced protein appears to be an artifact that arises from a lack of glycosylation. We also present the 2.1-Å-resolution crystal structure of the VE-cadherin EC1-2 adhesive region, which reveals homodimerization via the strand-swap mechanism common to classical cadherins. In common with type II cadherins, strand-swap binding involves two tryptophan anchor residues, but the adhesive interface resembles type I cadherins in that VE-cadherin does not form a large nonswapped hydrophobic surface. Thus, VE-cadherin is an outlier among classical cadherins, with characteristics of both type I and type II subfamilies.
Collapse
Affiliation(s)
- Julia Brasch
- Department of Biochemistry and Molecular Biophysics, Columbia University, 635 West 165 Street, New York, NY 10033, USA
| | - Oliver J. Harrison
- Department of Biochemistry and Molecular Biophysics, Columbia University, 635 West 165 Street, New York, NY 10033, USA
- Howard Hughes Medical Institute, Columbia University, 1130 St Nicholas Avenue, New York, NY 10032, USA
| | - Goran Ahlsen
- Department of Biochemistry and Molecular Biophysics, Columbia University, 635 West 165 Street, New York, NY 10033, USA
| | - Stewart M. Carnally
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Robert M. Henderson
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, UK
| | - Barry Honig
- Department of Biochemistry and Molecular Biophysics, Columbia University, 635 West 165 Street, New York, NY 10033, USA
- Howard Hughes Medical Institute, Columbia University, 1130 St Nicholas Avenue, New York, NY 10032, USA
- Center for Computational Biology and Bioinformatics, Columbia University, 1130 St Nicholas Avenue, New York, NY 10032, USA
| | - Lawrence Shapiro
- Department of Biochemistry and Molecular Biophysics, Columbia University, 635 West 165 Street, New York, NY 10033, USA
- Edward S. Harkness Eye Institute, Columbia University in the City of New York, New York, USA
| |
Collapse
|
42
|
Kher R, Sha EC, Escobar MR, Andreoli EM, Wang P, Xu WM, Wandinger-Ness A, Bacallao RL. Ectopic expression of cadherin 8 is sufficient to cause cyst formation in a novel 3D collagen matrix renal tubule culture. Am J Physiol Cell Physiol 2011; 301:C99-C105. [PMID: 21389276 DOI: 10.1152/ajpcell.00151.2010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
While a variety of genetic mutations have been shown to be associated with renal cyst formation, mechanisms of renal cyst formation are largely unknown. In prior communications we described alterations in E-cadherin assembly in cultured cystic epithelial cells (Charron AJ, Nakamura S, Bacallao R, Wandinger-Ness A. J Cell Biol 149: 111-124, 2000). Using the same cell line we assayed cadherin expression by RT-PCR using primer pairs that anneal to highly conserved sequences of cadherin genes but flank informative regions of cadherins. Using this approach we found that autosomal dominant polycystic kidney disease (ADPKD) cells express cadherin 8, a neuronal cadherin with limited expression in the kidney. Immunohistochemistry confirmed cadherin 8 expression in cystic epithelia. To test the functional significance of cadherin 8 expression in renal epithelial cells, we adapted a three-dimensional collagen culture method in which HK-2 cells form tubule structures and microinjected adenovirus into the matrix space surrounding tubule structures. Adenovirus expressing cadherin 8 under the control of a tet promoter caused cyst structures to grow out of the tubules when coinjected with adenovirus expressing a tet transactivator. Microinjection of single adenovirus expressing either tet transactivator or cadherin 8 failed to cause cyst formation. When doxycycline was added to the culture, following coinjection of adenovirus, there was a dose-response reduction in cadherin 8 expression and cyst formation. Similarly, HK-2 cells transfected with Flag-tagged cadherin 8 form cysts in addition to tubular structures. HK-2 cells transfected with Flag-tagged N-cadherin do not form cysts. These data suggest that ectopic expression of cadherin 8 in renal epithelial cells is sufficient to cause the morphogenic pattern of cyst formation.
Collapse
Affiliation(s)
- Rajesh Kher
- Division of Nephrology, Richard L. Roudebush Department of Veterans Affairs Medical Center and Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Rank A, Nieuwland R, Liebhardt S, Iberer M, Grützner S, Toth B, Pihusch R. Apheresis platelet concentrates contain platelet-derived and endothelial cell-derived microparticles. Vox Sang 2011; 100:179-86. [DOI: 10.1111/j.1423-0410.2010.01385.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
44
|
Zhang CC, Yan Z, Zhang Q, Kuszpit K, Zasadny K, Qiu M, Painter CL, Wong A, Kraynov E, Arango ME, Mehta PP, Popoff I, Casperson GF, Los G, Bender S, Anderes K, Christensen JG, VanArsdale T. PF-03732010: a fully human monoclonal antibody against P-cadherin with antitumor and antimetastatic activity. Clin Cancer Res 2010; 16:5177-88. [PMID: 20829331 DOI: 10.1158/1078-0432.ccr-10-1343] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE P-cadherin is a membrane glycoprotein that functionally mediates tumor cell adhesion, proliferation, and invasiveness. We characterized the biological properties of PF-03732010, a human monoclonal antibody against P-cadherin, in cell-based assays and tumor models. EXPERIMENTAL DESIGN The affinity, selectivity, and cellular inhibitory activity of PF-03732010 were tested in vitro. Multiple orthotopic and metastatic tumor models were used for assessing the antitumor and antimetastatic activities of PF-03732010. Treatment-associated pharmacodynamic changes were also investigated. RESULTS PF-03732010 selectively inhibits P-cadherin-mediated cell adhesion and aggregation in vitro. In the P-cadherin-overexpressing tumor models, including MDA-MB-231-CDH3, 4T1-CDH3, MDA-MB-435HAL-CDH3, HCT116, H1650, PC3M-CDH3, and DU145, PF-03732010 inhibited the growth of primary tumors and metastatic progression, as determined by bioluminescence imaging. Computed tomography imaging, H&E stain, and quantitative PCR analysis confirmed the antimetastatic activity of PF-03732010. In contrast, PF-03732010 did not show antitumor and antimetastatic efficacy in the counterpart tumor models exhibiting low P-cadherin expression. Mechanistic studies via immunofluorescence, immunohistochemical analyses, and 3'-[(18)F]fluoro-3'-deoxythymidine-positron emission tomography imaging revealed that PF-03732010 suppressed P-cadherin levels, caused degradation of membrane β-catenin, and concurrently suppressed cytoplasmic vimentin, resulting in diminished metastatic capacity. Changes in the levels of Ki67, caspase-3, and 3'-[(18)F]fluoro-3'-deoxythymidine tracer uptake also indicated antiproliferative activity and increased apoptosis in the tested xenografts. CONCLUSIONS These findings suggest that interrupting the P-cadherin signaling pathway may be a novel therapeutic approach for cancer therapy. PF-03732010 is presently undergoing evaluation in Phase 1 clinical trials.
Collapse
Affiliation(s)
- Cathy C Zhang
- Translational Research Group in Oncology Research Unit, Pfizer Global Research and Development, La Jolla Laboratories, San Diego, California 92121, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
The soluble fragment of VE-cadherin inhibits angiogenesis by reducing endothelial cell proliferation and tube capillary formation. Cancer Gene Ther 2010; 17:700-7. [PMID: 20559333 DOI: 10.1038/cgt.2010.26] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Vascular endothelial-specific cadherin (VE-cadherin) is an endothelial cell-specific adhesion molecule, localized at cell-cell contact sites. It is involved in physiological and pathological angiogenesis. In this study, we showed that in vitro a soluble N-terminal fragment of VE-cadherin (EC1-3) corresponding to cadherin 1-3 ectodomains inhibited vascular endothelial growth factor-stimulated endothelial cell proliferation and capillary tube structure formation in the matrigel model. In vivo, EC1-3 was tested in a murine colon cancer model. EC1-3-expressing colon cancer C51 cells were subcutaneously grafted into nude mice, and tumor growth and angiogenesis were evaluated. At day 33, the mean volume of the tumors developed was reduced (510±104 versus 990±120 mm(3) for control). Similarly, injection of EC1-3 virus-producing cells into established C51 tumors resulted in an inhibition by 33% of tumor growth. Immunohistological staining of vessels on tumor sections showed a significantly reduced intratumoral angiogenesis. Furthermore, EC1-3 did not induce vessel injury in the lung, liver, spleen, heart and brain in the mice. These results suggest that the soluble N-terminal fragment of VE-cadherin EC1-3 could exert an antitumoral effect by targeting tumor angiogenesis, which included blocking endothelial cell proliferation and capillary tube formation with no obvious toxicity on normal organs.
Collapse
|
46
|
Association of genetic variants with chronic kidney disease in Japanese individuals with or without hypertension or diabetes mellitus. Exp Ther Med 2010; 1:137-145. [PMID: 23136606 DOI: 10.3892/etm_00000023] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Accepted: 09/08/2009] [Indexed: 11/05/2022] Open
Abstract
Hypertension and diabetes mellitus are important risk factors for chronic kidney disease (CKD). The purpose of the present study was to identify genetic variants that confer susceptibility to CKD in individuals with or without hypertension or diabetes mellitus, thereby contributing to the personalized prevention of CKD in such individuals separately. The study population comprised 5835 unrelated Japanese individuals, including 1763 subjects with CKD and 4072 controls. The 150 polymorphisms were selected by genome-wide association studies of ischemic stroke and myocardial infarction with the use of the GeneChip Human Mapping 500K Array Set (Affymetrix). The genotypes for these polymorphisms were determined by a method that combines polymerase chain reaction and sequence-specific oligonucleotide probes with suspension array technology. The χ(2) test, multivariable logistic regression analysis with adjustment for covariates, as well as a stepwise forward selection procedure revealed that two different polymorphisms were significantly (P<0.005) associated with the prevalence of CKD in individuals with or without hypertension or diabetes mellitus: the A→G (Lys625Arg) polymorphism of CDH4 (rs6142884) in individuals without diabetes mellitus, and the C→T polymorphism of PTPRN2 (rs1638021) in individuals with hypertension and diabetes mellitus. No polymorphism was significantly associated with CKD in individuals with or without hypertension, in those with diabetes mellitus, or in those without hypertension or diabetes mellitus. Stratification of subjects based on hypertension or diabetes mellitus may thus be fundamental to achieving the personalized prevention of CKD with the use of genetic information.
Collapse
|
47
|
Caveda L, Corada M, Padura IM, Maschio AD, Breviario F, Lampugnani MG, Dejana E. Structural Characteristics and Functional Role of Endothelial Cell to Cell Junctions. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/10623329409024630] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
48
|
Dong Z, Nör JE. Transcriptional targeting of tumor endothelial cells for gene therapy. Adv Drug Deliv Rev 2009; 61:542-53. [PMID: 19393703 PMCID: PMC2727054 DOI: 10.1016/j.addr.2009.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2008] [Accepted: 02/05/2009] [Indexed: 12/21/2022]
Abstract
It is well known that angiogenesis plays a critical role in the pathobiology of tumors. Recent clinical trials have shown that inhibition of angiogenesis can be an effective therapeutic strategy for patients with cancer. However, one of the outstanding issues in anti-angiogenic treatment for cancer is the development of toxicities related to off-target effects of drugs. Transcriptional targeting of tumor endothelial cells involves the use of specific promoters for selective expression of therapeutic genes in the endothelial cells lining the blood vessels of tumors. Recently, several genes that are expressed specifically in tumor-associated endothelial cells have been identified and characterized. These discoveries have enhanced the prospectus of transcriptionally targeting tumor endothelial cells for cancer gene therapy. In this manuscript, we review the promoters, vectors, and therapeutic genes that have been used for transcriptional targeting of tumor endothelial cells, and discuss the prospects of such approaches for cancer gene therapy.
Collapse
Affiliation(s)
- Zhihong Dong
- Angiogenesis Research Laboratory, Department of Restorative Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Jacques E. Nör
- Angiogenesis Research Laboratory, Department of Restorative Sciences, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan College of Engineering, Ann Arbor, MI 48109, USA
- University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109, USA
| |
Collapse
|
49
|
Herwig MC, Müller KM, Müller AM. Endothelial VE-cadherin expression in human lungs. Pathol Res Pract 2008; 204:725-30. [PMID: 18639387 DOI: 10.1016/j.prp.2008.04.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Accepted: 04/15/2008] [Indexed: 11/26/2022]
Abstract
Cell adhesion molecule vascular endothelial cadherin (VE-cadherin) is the major component of endothelial adherence junctions, maintaining endothelial cell integrity. Studies dealing with constitutive VE-cadherin expression patterns in different pulmonary vessel types (arteries, arterioles, capillaries, venules, veins) or with the influence of physiological factors such as age or sex on VE-cadherin expression have not been published yet. Knowledge of constitutive resp. varying expression patterns not only fundamentally contribute to understanding the role of VE-cadherin in the pathogenesis of pulmonary diseases but also help to develop therapies based on immunotargeting. Hence, endothelial VE-cadherin expression was studied in regular lung tissue. Fifty-eight specimens of regular lung tissue (30 females, 28 males between 1 month and 75 years old) were immunohistochemically stained with an antibody against VE-cadherin. There was strong endothelial expression of VE-cadherin in arteries, arterioles, and capillaries but almost no expression in veins and venules. Neither age nor sex had any influence on the expression pattern or staining intensity. There is a vessel type-specific expression pattern for VE-cadherin in regular human lung tissue, which is not influenced by age or sex. Further studies will have to prove whether this is influenced by pathological conditions, e.g., ARDS.
Collapse
Affiliation(s)
- Martina C Herwig
- Department of Pathology, University of Bonn, Sigmund-Freud-Strasse 25, 53127 Bonn, Germany
| | | | | |
Collapse
|
50
|
Zhou R, Skalli O. Identification of cadherin-11 down-regulation as a common response of astrocytoma cells to Transforming Growth Factor-α. Differentiation 2008. [DOI: 10.1111/j.1432-0436.2000.660402.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|