1
|
Shi Y, Huang X, Du Z, Tan J. Analysis of single-cell RNA-sequencing data identifies a hypoxic tumor subpopulation associated with poor prognosis in triple-negative breast cancer. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:5793-5812. [PMID: 35603379 DOI: 10.3934/mbe.2022271] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of mammary carcinoma characterized by low expression levels of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Along with the rapid development of the single-cell RNA-sequencing (scRNA-seq) technology, the heterogeneity within the tumor microenvironment (TME) could be studied at a higher resolution level, facilitating an exploration of the mechanisms leading to poor prognosis during tumor progression. In previous studies, hypoxia was considered as an intrinsic characteristic of TME in solid tumors, which would activate downstream signaling pathways associated with angiogenesis and metastasis. Moreover, hypoxia-related genes (HRGs) based risk score models demonstrated nice performance in predicting the prognosis of TNBC patients. However, it is essential to further investigate the heterogeneity within hypoxic TME, such as intercellular communications. In the present study, utilizing single-sample Gene Set Enrichment Analysis (ssGSEA) and cell-cell communication analysis on the scRNA-seq data retrieved from Gene Expression Omnibus (GEO) database with accession number GSM4476488, we identified four tumor subpopulations with diverse functions, particularly a hypoxia-related one. Furthermore, results of cell-cell communication analysis revealed the dominant role of the hypoxic tumor subpopulation in angiogenesis- and metastasis-related signaling pathways as a signal sender. Consequently, regard the TNBC cohorts acquired from The Cancer Genome Atlas (TCGA) and GEO as train set and test set respectively, we constructed a risk score model with reliable capacity for the prediction of overall survival (OS), where ARTN and L1CAM were identified as risk factors promoting angiogenesis and metastasis of tumors. The expression of ARTN and L1CAM were further analyzed through tumor immune estimation resource (TIMER) platform. In conclusion, these two marker genes of the hypoxic tumor subpopulation played vital roles in tumor development, indicating poor prognosis in TNBC patients.
Collapse
Affiliation(s)
- Yi Shi
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
| | - Xiaoqian Huang
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
| | - Zhaolan Du
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
| | - Jianjun Tan
- Department of Biomedical Engineering, Faculty of Environment and Life, Beijing University of Technology, Beijing International Science and Technology Cooperation Base for Intelligent Physiological Measurement and Clinical Transformation, Beijing 100124, China
| |
Collapse
|
2
|
Sonnenberg SB, Rauer J, Göhr C, Gorinski N, Schade SK, Abdel Galil D, Naumenko V, Zeug A, Bischoff SC, Ponimaskin E, Guseva D. The 5-HT 4 receptor interacts with adhesion molecule L1 to modulate morphogenic signaling in neurons. J Cell Sci 2021; 134:jcs.249193. [PMID: 33536244 DOI: 10.1242/jcs.249193] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/19/2021] [Indexed: 11/20/2022] Open
Abstract
Morphological remodeling of dendritic spines is critically involved in memory formation and depends on adhesion molecules. Serotonin receptors are also implicated in this remodeling, though the underlying mechanisms remain enigmatic. Here, we uncovered a signaling pathway involving the adhesion molecule L1CAM (L1) and serotonin receptor 5-HT4 (5-HT4R, encoded by HTR4). Using Förster resonance energy transfer (FRET) imaging, we demonstrated a physical interaction between 5-HT4R and L1, and found that 5-HT4R-L1 heterodimerization facilitates mitogen-activated protein kinase activation in a Gs-dependent manner. We also found that 5-HT4R-L1-mediated signaling is involved in G13-dependent modulation of cofilin-1 activity. In hippocampal neurons in vitro, the 5-HT4R-L1 pathway triggers maturation of dendritic spines. Thus, the 5-HT4R-L1 signaling module represents a previously unknown molecular pathway regulating synaptic remodeling.
Collapse
Affiliation(s)
| | - Jonah Rauer
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Christoph Göhr
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Nataliya Gorinski
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Sophie Kristin Schade
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Dalia Abdel Galil
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Vladimir Naumenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia
| | - André Zeug
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany
| | - Stephan C Bischoff
- Department of Nutritional Medicine, University of Hohenheim, Stuttgart 70599, Germany
| | - Evgeni Ponimaskin
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany .,Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk 630090, Russia.,Institute of Neuroscience, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603950, Russian Federation
| | - Daria Guseva
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover 30625, Germany .,Department of Nutritional Medicine, University of Hohenheim, Stuttgart 70599, Germany
| |
Collapse
|
3
|
Deschepper FM, Zoppi R, Pirro M, Hensbergen PJ, Dall’Olio F, Kotsias M, Gardner RA, Spencer DI, Videira PA. L1CAM as an E-selectin Ligand in Colon Cancer. Int J Mol Sci 2020; 21:ijms21218286. [PMID: 33167483 PMCID: PMC7672641 DOI: 10.3390/ijms21218286] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Metastasis is the main cause of death among colorectal cancer (CRC) patients. E-selectin and its carbohydrate ligands, including sialyl Lewis X (sLeX) antigen, are key players in the binding of circulating tumor cells to the endothelium, which is one of the major events leading to organ invasion. Nevertheless, the identity of the glycoprotein scaffolds presenting these glycans in CRC remains unclear. In this study, we firstly have characterized the glycoengineered cell line SW620 transfected with the fucosyltransferase 6 (FUT6) coding for the α1,3-fucosyltransferase 6 (FUT6), which is the main enzyme responsible for the synthesis of sLeX in CRC. The SW620FUT6 cell line expressed high levels of sLeX antigen and E-selectin ligands. Moreover, it displayed increased migration ability. E-selectin ligand glycoproteins were isolated from the SW620FUT6 cell line, identified by mass spectrometry, and validated by flow cytometry and Western blot (WB). The most prominent E-selectin ligand we identified was the neural cell adhesion molecule L1 (L1CAM). Previous studies have shown association of L1CAM with metastasis in cancer, thus the novel role as E-selectin counter-receptor contributes to understand the molecular mechanism involving L1CAM in metastasis formation.
Collapse
Affiliation(s)
- Fanny M. Deschepper
- Unidade de Ciências Biomoleculares Aplicadas (UCIBIO), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal; (F.M.D.); (R.Z.)
| | - Roberta Zoppi
- Unidade de Ciências Biomoleculares Aplicadas (UCIBIO), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal; (F.M.D.); (R.Z.)
| | - Martina Pirro
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (M.P.); (P.J.H.)
| | - Paul J. Hensbergen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands; (M.P.); (P.J.H.)
| | - Fabio Dall’Olio
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40138 Bologna, Italy;
| | - Maximillianos Kotsias
- Ludger Ltd., Culham Science Centre, Abingdon, Oxfordshire OX14 3EB, UK; (M.K.); (R.A.G.); (D.I.R.S.)
| | - Richard A. Gardner
- Ludger Ltd., Culham Science Centre, Abingdon, Oxfordshire OX14 3EB, UK; (M.K.); (R.A.G.); (D.I.R.S.)
| | - Daniel I.R. Spencer
- Ludger Ltd., Culham Science Centre, Abingdon, Oxfordshire OX14 3EB, UK; (M.K.); (R.A.G.); (D.I.R.S.)
| | - Paula A. Videira
- Unidade de Ciências Biomoleculares Aplicadas (UCIBIO), Departamento Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal; (F.M.D.); (R.Z.)
- CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), 2829-516 Caparica, Portugal
- Correspondence:
| |
Collapse
|
4
|
Xu YW, Hong CQ, Wu ZY, Peng YH, Ran LQ, Yang SH, Huang BS, Liang XY, Chen HL, Wu JY, Xu XE, Deng JW, Zou HY, Fang WK, Li EM, Xu LY, Xie JJ. Diagnostic and prognostic value of serum L1-cell adhesion molecule in esophageal squamous cell carcinoma. Clin Res Hepatol Gastroenterol 2018; 42:597-603. [PMID: 30219694 DOI: 10.1016/j.clinre.2018.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 06/12/2018] [Accepted: 08/15/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVE L1 cell adhesion molecule (L1CAM) has been found to be dysregulated in several types of human cancers. Here, we aimed to determine the level of soluble L1CAM in serum of patients with esophageal squamous cell carcinoma (ESCC). METHODS Serum levels of L1CAM were determined by an enzyme-linked immunosorbent assay (ELISA) in 191 patients with ESCC and 94 normal controls. Receiver operating characteristics (ROC) was employed to calculate diagnostic accuracy. Cumulative survival time was calculated by the Kaplan-Meier method and analyzed by the logrank test. RESULTS Levels of L1CAM were significantly lower in all ESCC patients than in normal controls (P < 0.001). Detection of serum L1CAM provided a sensitivity of 28.3%, a specificity of 90.4% and an area under the curve (AUC) of 0.644 (95% CI: 0.579-0.710) in diagnosing ESCC. Similar results were observed in the diagnosis of early-stage ESCC (26.2% sensitivity, 90.4% specificity, and an AUC of 0.629). Moreover, decreased level of L1CAM was correlated with depth of tumor invasion (P < 0.05). Kaplan-Meier analysis showed that lower serum L1CAM level was significantly related to shorter overall survival time (P = 0.036) and disease-free survival time (P = 0.021) of ESCC patients. CONCLUSIONS Our study demonstrated that serum L1CAM might serve as a potential biomarker for the diagnosis and prognosis of ESCC.
Collapse
Affiliation(s)
- Yi-Wei Xu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou 515041, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling road, Shantou 515041, PR China
| | - Chao-Qun Hong
- Cancer Research Lab, The Cancer Hospital of Shantou University Medical College, Shantou 515041, PR China
| | - Zhi-Yong Wu
- Department of Surgical Oncology, Shantou Central Hospital, Shantou 515041, PR China
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou 515041, PR China
| | - Li-Qiang Ran
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling road, Shantou 515041, PR China
| | - Shi-Han Yang
- Department of Dermatology and Venereology, Shantou Central Hospital, Shantou 515041, PR China
| | - Bin-Sen Huang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling road, Shantou 515041, PR China
| | - Xiao-Ying Liang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling road, Shantou 515041, PR China
| | - Hai-Lu Chen
- Department of Surgical Oncology, Shantou Central Hospital, Shantou 515041, PR China
| | - Jian-Yi Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling road, Shantou 515041, PR China
| | - Xiu-E Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, PR China
| | - Jian-Wen Deng
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling road, Shantou 515041, PR China
| | - Hai-Ying Zou
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling road, Shantou 515041, PR China
| | - Wang-Kai Fang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling road, Shantou 515041, PR China
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling road, Shantou 515041, PR China
| | - Li-Yan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, PR China.
| | - Jian-Jun Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, No. 22, Xinling road, Shantou 515041, PR China.
| |
Collapse
|
5
|
Fibrin-Based Biomaterial Applications in Tissue Engineering and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:253-261. [DOI: 10.1007/978-981-13-0445-3_16] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
6
|
The Pleiotropic Role of L1CAM in Tumor Vasculature. Int J Mol Sci 2017; 18:ijms18020254. [PMID: 28134764 PMCID: PMC5343790 DOI: 10.3390/ijms18020254] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 01/19/2017] [Accepted: 01/23/2017] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis, the formation of new vessels, is a key step in the development, invasion, and dissemination of solid tumors and, therefore, represents a viable target in the context of antitumor therapy. Indeed, antiangiogenic approaches have given promising results in preclinical models and entered the clinical practice. However, in patients, the results obtained so far with antiangiogenic drugs have not completely fulfilled expectations, especially because their effect has been transient with tumors developing resistance and evasion mechanisms. A better understanding of the mechanisms that underlie tumor vascularization and the functional regulation of cancer vessels is a prerequisite for the development of novel and alternative antiangiogenic treatments. The L1 cell adhesion molecule (L1CAM), a cell surface glycoprotein previously implicated in the development and plasticity of the nervous system, is aberrantly expressed in the vasculature of various cancer types. L1CAM plays multiple pro-angiogenic roles in the endothelial cells of tumor-associated vessels, thus emerging as a potential therapeutic target. In addition, L1CAM prevents the maturation of cancer vasculature and its inhibition promotes vessel normalization, a process that is thought to improve the therapeutic response of tumors to cytotoxic drugs. We here provide an overview on tumor angiogenesis and antiangiogenic therapies and summarize the current knowledge on the biological role of L1CAM in cancer vasculature. Finally, we highlight the clinical implications of targeting L1CAM as a novel antiangiogenic and vessel-normalizing approach.
Collapse
|
7
|
Qian J, Oppermann E, Tran A, Imlau U, Qian K, Vogl TJ. Transarterial administration of integrin inhibitor loaded nanoparticles combined with transarterial chemoembolization for treating hepatocellular carcinoma in a rat model. World J Gastroenterol 2016; 22:5042-5049. [PMID: 27275096 PMCID: PMC4886379 DOI: 10.3748/wjg.v22.i21.5042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 02/24/2016] [Accepted: 03/14/2016] [Indexed: 02/06/2023] Open
Abstract
AIM: To compare the effect of transarterial chemoembolization (TACE) plus GRGDSP (Gly-Arg-Gly-Asp-Ser-Pro, integrin-inhibitor) loaded nanoparticles with TACE alone or TACE + GRGDSP in a rat model of liver tumor.
METHODS: Morris hepatoma 3924A tumors were implanted in the livers of 30 ACI rats. The ACI rats were divided randomly into three groups (10 animals each). Tumor volume before treatment (V1) was examined by magnetic resonance imaging (MRI), and then, after laparotomy and placement of a PE-10 catheter into the hepatic artery, the following interventional protocols were performed: TACE (mitomycin C + lipiodol + degradable starch microspheres) + GRGDSP loaded nanoparticles for group A; TACE + GRGDSP for group B (control group 1); TACE alone for group C (control group 2). Tumor volume (V2) was assessed by MRI and the mean ratio of the post-treatment to pretreatment tumor volumes (V2/V1) was calculated. Immunohistochemical analysis was performed to assess the quantification of matrix metalloprotein 9 (MMP-9) and vascular endothelial growth factor (VEGF) positive tumor cells in each treatment group.
RESULTS: The mean tumor growth ratios (V2/V1) were 1.3649 ± 0.1194 in group A, 2.0770 ± 0.1595 in group B, and 3.2148 ± 0.1075 in group C. Compared with groups B and C, group A showed a significant reduction in tumor volume. Lower expression of MMP-9 and VEGF in hepatocellular carcinoma was observed in group A than in groups B and C. The angiogenesis of tumor was evaluated using anti-VEGF antibodies, and the metastasis of tumor was assessed using anti-MMP-9 antibody. MMP-9 and VEGF were expressed in all specimens. The immunoexpression of these proteins was confirmed by the presence of red cytoplasmic staining in tumor cells. Lower expression of MMP-9 and VEGF in hepatocellular carcinoma was observed in group A than in groups B and C.
CONCLUSION: Transarterial administration of integrin inhibitor loaded nanoparticles combined with TACE evidently retards tumor growth and intrahepatic metastases compared with TACE alone or TACE plus integrin inhibitor in an animal model of hepatocellular carcinoma.
Collapse
|
8
|
The promotion of endothelial cell attachment and spreading using FNIII10 fused to VEGF-A165. Biomaterials 2013; 34:5958-68. [DOI: 10.1016/j.biomaterials.2013.04.050] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 04/24/2013] [Indexed: 11/20/2022]
|
9
|
Djogo N, Jakovcevski I, Müller C, Lee HJ, Xu JC, Jakovcevski M, Kügler S, Loers G, Schachner M. Adhesion molecule L1 binds to amyloid beta and reduces Alzheimer's disease pathology in mice. Neurobiol Dis 2013; 56:104-15. [PMID: 23639788 DOI: 10.1016/j.nbd.2013.04.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2012] [Revised: 03/10/2013] [Accepted: 04/09/2013] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common cause of elderly dementia. In an effort to contribute to the potential of molecular approaches to reduce degenerative processes we have tested the possibility that the neural adhesion molecule L1 ameliorates some characteristic cellular and molecular parameters associated with the disease in a mouse model of AD. Three-month-old mice overexpressing mutated forms of amyloid precursor protein and presenilin-1 under the control of a neuron-specific promoter received an injection of adeno-associated virus encoding the neuronal isoform of full-length L1 (AAV-L1) or, as negative control, green fluorescent protein (AAV-GFP) into the hippocampus and occipital cortex. Four months after virus injection, the mice were analyzed for histological and biochemical parameters of AD. AAV-L1 injection decreased the Aβ plaque load, levels of Aβ42, Aβ42/40 ratio and astrogliosis compared with AAV-GFP controls. AAV-L1 injected mice also had increased densities of inhibitory synaptic terminals on pyramidal cells in the hippocampus when compared with AAV-GFP controls. Numbers of microglial cells/macrophages were similar in both groups, but numbers of microglial cells/macrophages per plaque were increased in AAV-L1 injected mice. To probe for a molecular mechanism that may underlie these effects, we analyzed whether L1 would directly and specifically interact with Aβ. In a label-free binding assay, concentration dependent binding of the extracellular domain of L1, but not of the close homolog of L1 to Aβ40 and Aβ42 was seen, with the fibronectin type III homologous repeats 1-3 of L1 mediating this effect. Aggregation of Aβ42 in vitro was reduced in the presence of the extracellular domain of L1. The combined observations indicate that L1, when overexpressed in neurons and glia, reduces several histopathological hallmarks of AD in mice, possibly by reduction of Aβ aggregation. L1 thus appears to be a candidate molecule to ameliorate the pathology of AD, when applied in therapeutically viable treatment schemes.
Collapse
Affiliation(s)
- Nevena Djogo
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Han SW, Jung YK, Lee EJ, Park HR, Kim GW, Jeong JH, Han MS, Choi JY. DICAM inhibits angiogenesis via suppression of AKT and p38 MAP kinase signalling. Cardiovasc Res 2013; 98:73-82. [DOI: 10.1093/cvr/cvt019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
|
11
|
Wang Y, Loers G, Pan HC, Gouveia R, Zhao WJ, Shen YQ, Kleene R, Costa J, Schachner M. Antibody fragments directed against different portions of the human neural cell adhesion molecule L1 act as inhibitors or activators of L1 function. PLoS One 2012; 7:e52404. [PMID: 23272240 PMCID: PMC3525558 DOI: 10.1371/journal.pone.0052404] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 11/16/2012] [Indexed: 02/05/2023] Open
Abstract
The neural cell adhesion molecule L1 plays important roles in neuronal migration and survival, neuritogenesis and synaptogenesis. L1 has also been found in tumors of different origins, with levels of L1 expression correlating positively with the metastatic potential of tumors. To select antibodies targeting the varied functions of L1, we screened the Tomlinson library of recombinant human antibody fragments to identify antibodies binding to recombinant human L1 protein comprising the entire extracellular domain of human L1. We obtained four L1 binding single-chain variable fragment antibodies (scFvs), named I4, I6, I13, and I27 and showed by enzyme-linked immunosorbent assay (ELISA) that scFvs I4 and I6 have high affinity to the immunoglobulin-like (Ig) domains 1-4 of L1, while scFvs I13 and I27 bind strongly to the fibronectin type III homologous (Fn) domains 1-3 of L1. Application of scFvs I4 and I6 to human SK-N-SH neuroblastoma cells reduced proliferation and transmigration of these cells. Treatment of SK-N-SH cells with scFvs I13 and I27 enhanced cell proliferation and migration, neurite outgrowth, and protected against the toxic effects of H(2)O(2) by increasing the ratio of Bcl-2/Bax. In addition, scFvs I4 and I6 inhibited and scFvs I13 and I27 promoted phosphorylation of src and Erk. Our findings indicate that scFvs reacting with the immunoglobulin-like domains 1-4 inhibit L1 functions, whereas scFvs interacting with the fibronectin type III domains 1-3 trigger L1 functions of cultured neuroblastoma cells.
Collapse
Affiliation(s)
- Yan Wang
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universität Hamburg, Hamburg, Germany
| | - Hong-Chao Pan
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Ricardo Gouveia
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Wei-Jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Yan-Qin Shen
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie Hamburg, Universität Hamburg, Hamburg, Germany
| | - Julia Costa
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| |
Collapse
|
12
|
Biomimetic hydrogels for controlled biomolecule delivery to augment bone regeneration. Adv Drug Deliv Rev 2012; 64:1078-89. [PMID: 22465487 DOI: 10.1016/j.addr.2012.03.010] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2011] [Revised: 02/12/2012] [Accepted: 03/05/2012] [Indexed: 11/21/2022]
Abstract
The regeneration of large bone defects caused by trauma or disease remains a significant clinical problem. Although osteoinductive growth factors such as bone morphogenetic proteins have entered clinics, transplantation of autologous bone remains the gold standard to treat bone defects. The effective treatment of bone defects by protein therapeutics in humans requires quantities that exceed the physiological doses by several orders of magnitude. This not only results in very high treatment costs but also bears considerable risks for adverse side effects. These issues have motivated the development of biomaterials technologies allowing to better control biomolecule delivery from the solid phase. Here we review recent approaches to immobilize biomolecules by affinity binding or by covalent grafting to biomaterial matrices. We focus on biomaterials concepts that are inspired by extracellular matrix (ECM) biology and in particular the dynamic interaction of growth factors with the ECM. We highlight the value of synthetic, ECM-mimicking matrices for future technologies to study bone biology and develop the next generation of 'smart' implants.
Collapse
|
13
|
Hermann DM, ElAli A. The abluminal endothelial membrane in neurovascular remodeling in health and disease. Sci Signal 2012; 5:re4. [PMID: 22871611 DOI: 10.1126/scisignal.2002886] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
After brain injury, blood-brain barrier (BBB) integrity can be compromised as a consequence of the breakdown of cell-cell interactions in the neurovascular unit, resulting in the loss of the characteristic endothelial luminal-to-abluminal structure. During the process of restoration of the BBB and vascularization, the endothelial cells are continuously reshaped, with both the luminal and abluminal membranes serving as sites of signaling. Here, we focus on the bidirectional signaling processes that are rapidly initiated between endothelial and perivascular cells and occur in certain brain diseases or in response to injury. The goal of these processes is (i) the reemergence of endothelial cell polarity, (ii) the remodeling of extracellular matrix interactions, (iii) the realignment of pericytes and astrocytic endfeet with endothelial cells, and (iv) the restitution of a well-organized and stable BBB. This abluminal membrane exemplifies how the brain vasculature responds to stressors and may represent promising targets for therapeutic interventions of brain diseases.
Collapse
Affiliation(s)
- Dirk M Hermann
- Department of Neurology, University Hospital Essen, Hufelandstrasse 55, D-45122 Essen, Germany.
| | | |
Collapse
|
14
|
Zhu J, He P, Lin L, Jones DR, Marchant RE. Biomimetic poly(ethylene glycol)-based hydrogels as scaffolds for inducing endothelial adhesion and capillary-like network formation. Biomacromolecules 2012; 13:706-13. [PMID: 22296572 PMCID: PMC3310151 DOI: 10.1021/bm201596w] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The extracellular matrix (ECM) is an attractive model for designing synthetic scaffolds with a desirable environment for tissue engineering. Here, we report on the synthesis of ECM-mimetic poly(ethylene glycol) (PEG) hydrogels for inducing endothelial cell (EC) adhesion and capillary-like network formation. A collagen type I-derived peptide GPQGIAGQ (GIA)-containing PEGDA (GIA-PEGDA) was synthesized with the collagenase-sensitive GIA sequence attached in the middle of the PEGDA chain, which was then copolymerized with RGD capped-PEG monoacrylate (RGD-PEGMA) to form biomimetic hydrogels. The hydrogels degraded in vitro with the rate dependent on the concentration of collagenase and also supported the adhesion of human umbilical vein ECs (HUVECs). Biomimetic RGD/GIA-PEGDA hydrogels with incorporation of 1% RGD-PEGDA into GIA-PEGDA hydrogels induced capillary-like organization when HUVECs were seeded on the hydrogel surface, while RGD/PEGDA and GIA-PEGDA hydrogels did not. These results indicate that both cell adhesion and biodegradability of scaffolds play important roles in the formation of capillary-like networks.
Collapse
Affiliation(s)
- Junmin Zhu
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Ping He
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Lin Lin
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Derek R. Jones
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| | - Roger E. Marchant
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106
| |
Collapse
|
15
|
Peng LH, Tsang SY, Tabata Y, Gao JQ. Genetically-manipulated adult stem cells as therapeutic agents and gene delivery vehicle for wound repair and regeneration. J Control Release 2011; 157:321-30. [PMID: 21893122 DOI: 10.1016/j.jconrel.2011.08.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 08/10/2011] [Indexed: 02/06/2023]
Abstract
Wound therapy remains a clinical challenge and much effort has been focused on the development of novel therapeutic approaches for wound management. New knowledge about the way in which signals control wound cellular and molecular behavior has promoted the topical application of multipotent stem cells and bioactive molecules to injured tissue, for skin regeneration with less scar formation. However, limited clinical success indicates that the effective delivery of polypeptides and therapeutic cells, with controlled releasing profile, is a major challenge which is yet to be overcome. Recently, a technique in which the genetically-manipulated stem cells were used both as the therapeutic agents and the vehicle for gene delivery for wound treatment - a method which serves to provide regenerative cells and bioactive genes within an optimal environment of regulatory molecular expression for wound sites - has emerged as a promising strategy for wound regenerative therapy. In this article, the roles of adult stem cells - as the therapeutics and the vehicles in these advanced biomimetic drug delivery systems for wound regeneration medicine - are scrutinized to indicate their mechanisms, characteristics, broad applicability and future lines of investigation.
Collapse
Affiliation(s)
- Li-Hua Peng
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, PR China
| | | | | | | |
Collapse
|
16
|
Novosel EC, Kleinhans C, Kluger PJ. Vascularization is the key challenge in tissue engineering. Adv Drug Deliv Rev 2011; 63:300-11. [PMID: 21396416 DOI: 10.1016/j.addr.2011.03.004] [Citation(s) in RCA: 683] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 02/09/2011] [Accepted: 03/02/2011] [Indexed: 12/11/2022]
Abstract
The main limitation in engineering in vitro tissues is the lack of a sufficient blood vessel system - the vascularization. In vivo almost all tissues are supplied by these endothelial cell coated tubular networks. Current strategies to create vascularized tissues are discussed in this review. The first strategy is based on the endothelial cells and their ability to form new vessels known as neoangiogenesis. Herein prevascularization techniques are compared to approaches in which biomolecules, such as growth factors, cytokines, peptides and proteins as well as cells are applied to generate new vessels. The second strategy is focused on scaffold-based techniques. Naturally-derived scaffolds, which contain vessels, are distinguished from synthetically manufactured matrices. Advantages and pitfalls of the approaches to create vascularized tissues in vitro are outlined and feasible future strategies are discussed.
Collapse
|
17
|
Ong LL, Li W, Oldigs JK, Kaminski A, Gerstmayer B, Piechaczek C, Wagner W, Li RK, Ma N, Steinhoff G. Hypoxic/normoxic preconditioning increases endothelial differentiation potential of human bone marrow CD133+ cells. Tissue Eng Part C Methods 2011; 16:1069-81. [PMID: 20073989 DOI: 10.1089/ten.tec.2009.0641] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
CD133+ cells are hemangioblasts that have capacity to generate into both hematopoietic and endothelial cells (ECs). Hypoxia/normoxia has shown to be the regulator of the balance between stemness and differentiation. In this study we performed Agilent's whole human genome oligo microarray analysis and examined the differentiation potential of the bone-marrow-derived CD133+ cells after hypoxic/normoxic preconditioning of CD133+ cells. Results showed that there was no significant increase in erythroid colony forming unit (CFU-E) and CFU-granulocyte, erythrocyte, monocyte, and megakaryocyte formation with cells treated under hypoxia/normoxia. However, a significant increment of EC forming unit at 24 h (143.2 +/- 8.0%) compared to 0 h (100 +/- 11.4%) was observed in CFU-EC analysis. Reverse transcription-polymerase chain reaction and immunostaining analysis showed that the differentiated cells diminished hematopoietic stem cell surface markers and acquired the gene markers and functional phenotype of ECs. The transcriptome profile revealed a cluster of 232 downregulated and 498 upregulated genes in cells treated for 24 h under hypoxia. The upregulated genes include angiogenic genes, angiogenic growth factor genes, angiogenic cytokine and chemokine genes, as well as angiogenic-positive regulatory genes, including FGFBP1, PDGFB, CCL15, CXCL12, CXCL6, IL-6, PTN, EREG, ERBB2, EDG5, FGF3, FHF2, GDF15, JUN, L1CAM, NRG1, NGFR, and PDGFB. On the other hand, angiogenesis inhibitors and related genes, including IL12A, MLLT7, STAB1, and TIMP2, are downregulated. Taken together, hypoxic/normoxic preconditioning may lead to the differentiation of CD133+ cells toward endothelial lineage, which may improve the current clinical trial studies.
Collapse
Affiliation(s)
- Lee-Lee Ong
- Department of Cardiac Surgery, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Jung JP, Moyano JV, Collier JH. Multifactorial optimization of endothelial cell growth using modular synthetic extracellular matrices. Integr Biol (Camb) 2011; 3:185-96. [PMID: 21249249 DOI: 10.1039/c0ib00112k] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Extracellular matrices (ECMs) are complex materials, containing at least dozens of different macromolecules that are assembled together, thus complicating their optimization towards applications in 3D cell culture or tissue engineering. The natural complexity of ECMs has limited cell-matrix investigations predominantly to experiments where only one matrix component is adjusted at a time, making it difficult to uncover interactions between different matrix components or to efficiently determine optimal matrix compositions for specific desired biological responses. Here we have developed modular synthetic ECMs based on peptide self-assembly whose incorporation of multiple different peptide ligands can be adjusted. The peptides can co-assemble in a wide range of combinations to form hydrogels of uniform morphology and consistent mechanical properties, but with precisely varied mixtures of peptide ligands. The modularity of this system in turn enabled multi-factorial experimental designs for investigating interactions between these ligands and for determining a multi-peptide matrix formulation that maximized endothelial cell growth. In cultures of HUVECs, we observed a previously unknown antagonistic interaction between the laminin-derived peptide YIGSR and RGDS-mediated cell attachment and growth. We also identified an optimized combination of self-assembled peptides bearing the ligands RGDS and IKVAV that led to endothelial cell growth equivalent to that on native full-length fibronectin. Both of these findings would have been challenging to uncover using more traditional one-factor-at-a-time analyses.
Collapse
Affiliation(s)
- Jangwook P Jung
- Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Mail code 5032, Chicago, IL 60637, USA
| | | | | |
Collapse
|
19
|
Linking L1CAM-mediated signaling to NF-κB activation. Trends Mol Med 2010; 17:178-87. [PMID: 21195665 DOI: 10.1016/j.molmed.2010.11.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2010] [Revised: 11/19/2010] [Accepted: 11/19/2010] [Indexed: 01/13/2023]
Abstract
The cell adhesion molecule L1 (L1CAM) was originally identified as a neural adhesion molecule essential for neurite outgrowth and axon guidance. Many studies have now shown that L1CAM is overexpressed in human carcinomas and associated with poor prognosis. So far, L1CAM-mediated cellular signaling has been largely attributed to an association with growth factor receptors, referred to as L1CAM-'assisted' signaling. New data demonstrate that L1CAM can signal via two additional mechanisms: 'forward' signaling via regulated intramembrane proteolysis and 'reverse' signaling via the activation of the transcription factor nuclear factor (NF)-κB. Taken together, these findings lead to a new understanding of L1CAM downstream signaling that is fundamental for the development of anti-L1CAM antibody-mediated therapeutics in human tumor cells.
Collapse
|
20
|
Li Y, Galileo DS. Soluble L1CAM promotes breast cancer cell adhesion and migration in vitro, but not invasion. Cancer Cell Int 2010; 10:34. [PMID: 20840789 PMCID: PMC2949617 DOI: 10.1186/1475-2867-10-34] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2010] [Accepted: 09/15/2010] [Indexed: 11/17/2022] Open
Abstract
Background Neural recognition molecule L1CAM, which is a key protein involved in early nervous system development, is known to be abnormally expressed and shed in several types of cancers where it participates in metastasis and progression. The distinction of L1CAM presence in cancerous vs. normal tissues has suggested it to be a new target for cancer treatment. Our current study focused on the potential role of soluble L1CAM in breast cancer cell adhesion to extracellular matrix proteins, migration, and invasion. Results We found L1 expression levels were correlated with breast cancer stage of progression in established data sets of clinical samples, and also were high in more metastatic breast cancer cell lines MDA-MB-231 and MDA-MB-435, but low in less migratory MDA-MB-468 cells. Proteolysis of L1 into its soluble form (sL1) was detected in cell culture medium from all three above cell lines, and can be induced by PMA activation. Over-expression of the L1 ectodomain in MDA-MB-468 cells by using a lentiviral vector greatly increased the amount of sL1 released by those cells. Concomitantly, cell adhesion to extracellular matrix and cell transmigration ability were significantly promoted, while cell invasion ability through Matrigel™ remained unaffected. On the other hand, attenuating L1 expression in MDA-MB-231 cells by using a shRNA lentiviral vector resulted in reduced cell-matrix adhesion and transmigration. Similar effects were also shown by monoclonal antibody blocking of the L1 extracellular region. Moreover, sL1 in conditioned cell culture medium induced a directional migration of MDA-MB-468 cells, which could be neutralized by antibody treatment. Conclusions Our data provides new evidence for the function of L1CAM and its soluble form in promoting cancer cell adhesion to ECM and cell migration. Thus, L1CAM is validated further to be a potential early diagnostic marker in breast cancer progression and a target for breast cancer therapy.
Collapse
Affiliation(s)
- Yupei Li
- Department of Biological Sciences, University of Delaware, Wolf Hall, Newark, DE 19716 USA.
| | | |
Collapse
|
21
|
Lühmann T, Hänseler P, Grant B, Hall H. The induction of cell alignment by covalently immobilized gradients of the 6th Ig-like domain of cell adhesion molecule L1 in 3D-fibrin matrices. Biomaterials 2009; 30:4503-12. [DOI: 10.1016/j.biomaterials.2009.05.041] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 05/18/2009] [Indexed: 11/30/2022]
|
22
|
Danhier F, Vroman B, Lecouturier N, Crokart N, Pourcelle V, Freichels H, Jérôme C, Marchand-Brynaert J, Feron O, Préat V. Targeting of tumor endothelium by RGD-grafted PLGA-nanoparticles loaded with paclitaxel. J Control Release 2009; 140:166-73. [PMID: 19699245 DOI: 10.1016/j.jconrel.2009.08.011] [Citation(s) in RCA: 241] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2009] [Revised: 07/03/2009] [Accepted: 08/05/2009] [Indexed: 11/24/2022]
Abstract
Paclitaxel (PTX)-loaded PEGylated PLGA-based nanoparticles (NP) have been previously described as more effective in vitro and in vivo than taxol. The aim of this study was to test the hypothesis that our PEGylated PLGA-based nanoparticles grafted with the RGD peptide or RGD-peptidomimetic (RGDp) would target the tumor endothelium and would further enhance the anti-tumor efficacy of PTX. The ligands were grafted on the PEG chain of PCL-b-PEG included in the nanoparticles. We observed in vitro that RGD-grafted nanoparticles were more associated to human umbilical vein endothelial cells (HUVEC) by binding to alpha(v)beta(3) integrin than non-targeted nanoparticles. Doxorubicin was also used to confirm the findings observed for PTX. In vivo, we demonstrated the targeting of RGD and RGDp-grafted nanoparticles to tumor vessels as well as the effective retardation of TLT tumor growth and prolonged survival times of mice treated by PTX-loaded RGD-nanoparticles when compared to non-targeted nanoparticles. Hence, the targeting of anti-cancer drug to tumor endothelium by RGD-labeled NP is a promising approach.
Collapse
Affiliation(s)
- Fabienne Danhier
- Université Catholique de Louvain, Unité de Pharmacie Galénique, Avenue Mounier 73-20, 1200 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lu C, Pelech S, Zhang H, Bond J, Spach K, Noubade R, Blankenhorn EP, Teuscher C. Pertussis toxin induces angiogenesis in brain microvascular endothelial cells. J Neurosci Res 2009; 86:2624-40. [PMID: 18500752 DOI: 10.1002/jnr.21716] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pertussis toxin (PTX) is an ancillary adjuvant used to elicit experimental allergic encephalomyelitis (EAE), the principal autoimmune model of multiple sclerosis. One mechanism whereby PTX potentiates EAE is to increase blood-brain barrier (BBB) permeability. To elucidate further the mechanism of action of PTX on the BBB, we investigated the genomic and proteomic responses of isolated mouse brain endothelial cells (MBEC) following intoxication. Among approximately 14,000 mouse genes tracked by cDNA microarray, 34 showed altered expression in response to PTX. More than one-third of these genes have roles in angiogenesis. Accordingly, we show that intoxication of MBEC induces tube formation in vitro and angiogenesis in vivo. The global effect of PTX on signaling protein levels and phosphorylation in MBEC was investigated by using Kinex antibody microarrays. In total, 113 of 372 pan-specific and 58 of 258 phospho-site-specific antibodies revealed changes >or=25% following intoxication. Increased STAT1 Tyr-701 and Ser-727 phosphorylation; reduced phosphorylation of the activating phospho-sites in Erk1, Erk2, and MAPKAPK2; and decreased phosphorylation of arrestin beta1 Ser-412 and Hsp27 Ser-82 were confirmed by Kinetworks multi-immunoblotting. The importance of signal transduction pathways on PTX-induced MBEC tube formation was evaluated pharmacologically. Inhibition of phospholipase C, MEK1, and p38 MAP kinase had little effect, whereas inhibition of cAMP-dependent protein kinase, protein kinase C, and phosphatidylinositol 3-kinase partially blocked tube formation. Taken together, these findings are consistent with the concept that PTX may lead to increased BBB permeability by altering endothelial plasticity and angiogenesis.
Collapse
Affiliation(s)
- Changming Lu
- Department of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Friedli A, Fischer E, Novak-Hofer I, Cohrs S, Ballmer-Hofer K, Schubiger PA, Schibli R, Grünberg J. The soluble form of the cancer-associated L1 cell adhesion molecule is a pro-angiogenic factor. Int J Biochem Cell Biol 2009; 41:1572-80. [PMID: 19401151 DOI: 10.1016/j.biocel.2009.01.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 01/09/2009] [Accepted: 01/12/2009] [Indexed: 01/29/2023]
Abstract
A soluble form of the L1 cell adhesion molecule (sL1) is released from various tumor cells and can be found in serum and ascites fluid of uterine and ovarian carcinoma patients. sL1 is a ligand for several Arg-Gly-Asp (RGD)-binding integrins and can be deposited in the extracellular matrix. In this study we describe a novel function of this physiologically relevant form of L1 as a pro-angiogenic factor. We demonstrated that the anti-L1 monoclonal antibody (mAb) chCE7 binds near or to the sixth Ig-like domain of human L1 which contains a single RGD sequence. mAb chCE7 inhibited the RGD-dependent adhesion of ovarian carcinoma cells to sL1 and reversed the sL1-induced proliferation, matrigel invasion and tube formation of bovine aortic endothelial (BAE) cells. A combination of sL1 with vascular endothelial growth factor-A (VEGF-A(165)), which is an important angiogenic inducer in tumors, strongly potentiated VEGF receptor-2 tyrosine phosphorylation in BAE cells. Chick chorioallantoic membrane (CAM) assays revealed the pro-angiogenic potency of sL1 in vivo which could be abolished by chCE7. These results indicate an important role of released L1 in tumor angiogenesis and represent a novel function of antibody chCE7 in tumor therapy.
Collapse
Affiliation(s)
- Alexandra Friedli
- Center for Radiopharmaceutical Science ETH-PSI-USZ, Paul Scherrer Institute, Villigen, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
The extracellular matrix (ECM) acts both as a physical scaffold for cells and as a repository for growth factors. Moreover, ECM structure and physical-chemical properties convey precise information to cells that profoundly influences their biology by interactions with cell surface receptors termed integrins. During angiogenesis, the perivascular ECM plays a critical role in determining the proliferative, invasive and survival responses of the local vascular cells to the angiogenic growth factors. Dynamic changes in both the ECM and the local vascular cells act in concert to regulate new blood vessel growth. The digestion of ECM components by proteolysis is critical for the invasive capacity of endothelial cells, but also creates ECM fragments, which antagonize the mechanosensory function of integrins, and can be apoptogenic. Here, we discuss the roles of integrins in modulating cellular responses to a changing ECM, in particular the regulation of survival and invasion among invasive endothelial cells.
Collapse
|
26
|
Sieminski AL, Semino CE, Gong H, Kamm RD. Primary sequence of ionic self-assembling peptide gels affects endothelial cell adhesion and capillary morphogenesis. J Biomed Mater Res A 2008; 87:494-504. [DOI: 10.1002/jbm.a.31785] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
27
|
The role of adhesion molecules, alpha v beta 3, alpha v beta 5 and their ligands in the tumor cell and endothelial cell adhesion. Eur J Cancer Prev 2008; 16:517-27. [PMID: 18090124 DOI: 10.1097/cej.0b013e3280145c00] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Tumor metastasis is a complex process involving the interaction between tumor cells and endothelial cells in which some adhesion molecules play an important role. It was our aim to investigate the role of the adhesion molecules, alpha v beta 3 and alpha v beta 5 and their ligands, developmental endothelial locus-1 (Del-1) and L1, in tumor cell adhesion to endothelial cells in vitro. In this study, the expression and regulation of alpha v beta 3, alpha v beta 5 and intercellular adhesion molecule -1 on liver sinusoidal endothelial cells and liver cancer endothelial cells (T3A) were analyzed by real-time PCR and fluorescent-activated cell sorter. The expression and regulation of the integrin ligands, Del-1 and L1, in six tumor cell lines were analyzed by real-time PCR and western blot. We found the expressions of alpha v beta 3 and alpha v beta 5 were higher on T3A than that on liver sinusoidal endothelial cells, whereas expression of intercellular adhesion molecule-1 was lower on T3A than that on liver sinusoidal endothelial cells. After 24 h hypoxia, the expressions of alpha v beta 3 and alpha v beta 5 were upregulated on T3A and liver sinusoidal endothelial cells; the expression of intercellular adhesion molecule-1 was increased on liver sinusoidal endothelial cells, but remained unchanged on T3A. Del-1 and L1 expression levels were obviously diverse in various tumor cell lines and differentially modulated after 12 h hypoxia. The adhesion of tumor cells with Del-1 and L1 expression was higher in T3A than that in liver sinusoidal endothelial cells, and was significantly increased under hypoxic conditions. Interestingly, the tumor cell adherence could be inhibited by antibodies against alpha v beta 5 and alpha v beta 5, but not by an antibody against intercellular adhesion molecule-1. The adhesion of tumor cells without Del-1 and L1 expression was also higher on T3A than that on liver sinusoidal endothelial cells, but the adhesion could not be inhibited by antibodies against alpha v beta 5, alpha v beta 5 or intercellular adhesion molecule-1, suggesting that other receptors are involved. In conclusion, alpha v beta 5, alpha v beta 5 and their ligands Del-1 and L1 play an important role in the process of tumor cells moving from the original place.
Collapse
|
28
|
Experimental study on transarterial administration of GRGDSP combined with transarterial chemoembolization in rats with hepatic carcinoma. Cardiovasc Intervent Radiol 2007; 31:377-82. [PMID: 18058171 DOI: 10.1007/s00270-007-9233-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2006] [Revised: 01/30/2007] [Accepted: 10/16/2007] [Indexed: 12/19/2022]
Abstract
PURPOSE To evaluate the effects of transarterial administration of an integrin antagonist, GRGDSP (Gly-Arg-Gly-Asp-Ser-Pro), combined with transarterial chemoembolization (TACE) to treat hepatic carcinoma in rats. METHODS Walker-256 tumor was implanted beneath the liver capsule in 26 Wistar rats. Animal subjects were assigned to groups based on which treatment was injected into the hepatic artery: group A, GRGDSP + TACE; group B, TACE alone; and group C, normal saline. Magnetic resonance imaging (MRI), tumor pathology, and immunohistochemistry were performed to assess each treatment. RESULTS The ratios of the post-treatment to pretreatment tumor volumes (V2/V1) in groups A, B, and C were 4.42 +/- 0.48, 6.98 +/- 1.09, and 13.00 +/- 1.68, respectively. The metastatic potential of the tumors was assessed by tumor cell nest counts, which were 5.00 +/- 1.25, 6.63 +/- 1.60, and 7.22 +/- 1.92 in groups A, B, and C, respectively. Microvessel density (MVD) was quantified by measuring von Willebrand factor density values, which were 0.18 +/- 0.02, 0.22 +/- 0.02, and 0.23 +/- 0.02 in groups A, B, and C, respectively. CONCLUSIONS Transarterial infusion of GRGDSP combined with TACE noticeably inhibited the growth of hepatic carcinoma and intrahepatic metastases in rats.
Collapse
|
29
|
Choi DS, Lee JM, Park GW, Lim HW, Bang JY, Kim YK, Kwon KH, Kwon HJ, Kim KP, Gho YS. Proteomic analysis of microvesicles derived from human colorectal cancer cells. J Proteome Res 2007; 6:4646-55. [PMID: 17956143 DOI: 10.1021/pr070192y] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Microvesicles (MV) are membrane vesicles secreted from the plasma and endosomal membrane compartment by various cell types such as hematopoietic, epithelial, and tumor cells. Actively growing tumor cells shed MV, and the rate of shedding increases in malignant tumors. Although recent progress in this area has revealed that tumor-derived MV play multiple roles in tumor growth and metastasis via immune escape, tumor invasion, and angiogenesis, the mechanism of vesicle formation and the biological roles of tumor-derived MV are not understood. Here, we report the first global proteomic analysis of highly purified MV from human colorectal cancer cells. Using 1D SDS gel electrophoresis and nano-LC-MS/MS analyses, we identified a total of 547 microvesicular proteins from three independent experiments with high confidence; 416 proteins were identified at least in two trials, including 181 as yet unreported proteins. We identified 49 proteins involved in the biogenesis of MV, including annexins, ADP-ribosylation factors, and Rab proteins. We also identified 28 proteins that may function in tumorigenesis via promotion of migration, invasion, and growth of tumor cells, immune modulation, metastasis, and angiogenesis. Taken together with previously reported results, our observations suggest that tumor-derived MV may act as communicasomes, that is, extracellular organelles that play diverse roles in intercellular communication. This information will help elucidate the biogenesis and functions of tumor-derived MV, and aid in the development of effective vaccines for various cancers, including colorectal cancer.
Collapse
Affiliation(s)
- Dong-Sic Choi
- Department of Life Science and Division of Molecular and Life Sciences, Pohang University of Science and Technology, Pohang 790-784, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Reidy M, Zihlmann P, Hubbell JA, Hall H. Activation of cell-survival transcription factor NFkappaB in L1Ig6-stimulated endothelial cells. J Biomed Mater Res A 2007; 77:542-50. [PMID: 16493660 DOI: 10.1002/jbm.a.30590] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Ligation of the integrin alpha(v)beta(3) in endothelial cells has been shown to be important for their survival. Such ligation induces signalling events merging into the Raf-Ras-ERK cascade that eventually induces activation of nuclear factor kappa B (NFkappaB), leading to its phosphorylation and nuclear translocation and thus inhibiting apoptosis. Here, the recombinant sixth immunoglobulin-like domain of cell adhesion molecules L1 (L1Ig6), a ligand for integrin alpha(v)beta(3), was explored as a component of vascular implant surfaces to initiate the NFkappaB-cell survival pathway. This supposition was supported. Specifically, NFkappaB-p65 was expressed in human umbilical vein endothelial cells (HUVECs) and when stimulated on L1Ig6, the phosphorylated form was found in the nucleus in over 60% of the cells. NFkappaB was not translocated into the nucleus on a number of other extracellular matrix substrates examined or when fibroblasts were cultured on L1Ig6. NFkappaB phosphorylation and nuclear translocation could be inhibited by blocking ligation of alpha(v)beta(3) by L1Ig6 with an antibody recognizing alpha(v)beta(3), with a cyclic RGD peptide, and with soluble L1Ig6. Moreover, blocking of alpha(v)beta(3) interaction with L1Ig6 was correlated with induction of apoptosis. Thus, these experiments demonstrate that L1Ig6 may be useful as alpha(v)beta(3) ligand for the induction of endothelial survival pathways mediated by NFkappaB-p65.
Collapse
Affiliation(s)
- Martin Reidy
- Institute for Biomedical Engineering and Department of Materials, ETH Zurich, Zürich, Switzerland
| | | | | | | |
Collapse
|
31
|
Zhou BBS, Fridman JS, Liu X, Friedman SM, Newton RC, Scherle PA. ADAM proteases, ErbB pathways and cancer. Expert Opin Investig Drugs 2005; 14:591-606. [PMID: 16004590 DOI: 10.1517/13543784.14.6.591] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A disintegrin and metalloproteases (ADAMs) are zinc-dependent trans-membrane metalloproteases that shed the extracellular domains of membrane-bound growth factors, cytokines and receptors. Key functions of ADAMs have emerged in ErbB signalling pathways as being sheddases for multiple ErbB ligands. As the ErbB pathway is a validated target for anti-cancer drugs, the upstream activators of ErbB ligands, their sheddases, now enter the spotlight as new drug targets in the ErbB pathway. ADAMs are involved not only in tumour cell proliferation but also in angiogenesis and metastasis. Therefore, strategies targeting ADAMs might be an important complement to existing anti-ErbB approaches.
Collapse
Affiliation(s)
- Bin-Bing S Zhou
- Drug Discovery Biology, Incyte Corporation, Experimental Station, Route 141 & Henry Clay Road, Building 400, Wilmington, DE 19880, USA.
| | | | | | | | | | | |
Collapse
|