151
|
Goel PN, Gude RP. Pentoxifylline regulates the cellular adhesion and its allied receptors to extracellular matrix components in breast cancer cells. Biomed Pharmacother 2014; 68:93-9. [DOI: 10.1016/j.biopha.2013.09.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 09/24/2013] [Indexed: 11/16/2022] Open
|
152
|
Reynolds AL, Kent D, Kennedy BN. Current and Emerging Therapies for Ocular Neovascularisation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 801:797-804. [DOI: 10.1007/978-1-4614-3209-8_100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
153
|
Jahangiri A, Aghi MK, Carbonell WS. β1 integrin: Critical path to antiangiogenic therapy resistance and beyond. Cancer Res 2013; 74:3-7. [PMID: 24327727 DOI: 10.1158/0008-5472.can-13-1742] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Angiogenesis is an important tissue-level program supporting the growth of highly aggressive cancers and early-stage metastases. However, rapid emergence of resistance to antiangiogenic therapies, such as bevacizumab, greatly limits the clinical utility of these promising approaches. The mechanisms of resistance to antiangiogenic therapy remain incompletely understood. The tumor microenvironment has been demonstrated to be a source of broad therapeutic resistance in multiple cancers. Much of the interaction between the cells comprising a tumor and their microenvironment is driven by integrins. Notably, signaling downstream of integrins in tumor cells promotes fundamental programs vital to aggressive cancer biology, including proliferation, growth, invasion, and survival signaling. These functions then can contribute to malignant phenotypes, including metastasis, therapy resistance, epithelial-to-mesenchymal transition, and angiogenesis. Accordingly, we found β1 integrin to be functionally upregulated in tumor specimens from patients after bevacizumab failure and in xenograft models of bevacizumab resistance. Inhibition of β1 in tumor cells with stable gene knockdown or treatment with OS2966, a neutralizing β1 integrin monoclonal antibody, attenuated aggressive tumor phenotypes in vitro and blocked growth of bevacizumab-resistant tumor xenografts in vivo. Thus, β1 integrins promote resistance to antiangiogenic therapy through potentiation of multiple malignant programs facilitated by interactions with the tumor microenvironment. The elucidation of this mechanism creates an outstanding opportunity for improving patient outcomes in cancer.
Collapse
Affiliation(s)
- Arman Jahangiri
- Authors' Affiliations: Department of Neurosurgery, University of California; and OncoSynergy Inc., San Francisco, California
| | | | | |
Collapse
|
154
|
Singh MK, Bhattacharya D, Chaudhuri S, Acharya S, Kumar P, Santra P, Basu AK, Chaudhuri S. T11TS inhibits glioma angiogenesis by modulation of MMPs, TIMPs, with related integrin αv and TGF-β1 expressions. Tumour Biol 2013; 35:2231-46. [PMID: 24242015 DOI: 10.1007/s13277-013-1296-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 10/07/2013] [Indexed: 12/29/2022] Open
Abstract
During glioma development, angiogenesis plays a crucial role in growth and vascularization of primary brain tumors. T11 target structure (T11TS), a bioactive molecule, has been documented as an anti-neoplastic agent in glioma-induced rats and also in human glioma in vitro. This novel molecule induces apoptosis of tumor cells by way of immune potentiation and impairs the glioma cell cycle, but its role in glioma angiogenesis has not been worked out in detail. Matrix metalloproteinases (MMPs) are enzymes promoting tumor angiogenesis by enzymatically remodeling the extracellular matrix and altering surface protein expression such as integrin αv and the matrix-bound proteins like TGF-β1. The present study was formulated to assess the efficacy of T11TS in the modulations of MMP-2 and -9 and their endogenous inhibitors (TIMP-1 and TIMP-2) as well as modulations of integrin αv and TGF-β1 in glioma-induced rats and also on the phenotypic markers of endothelial cells (CD31 and CD34). The parameters used were zymography, western blot, and flow cytometric analyses. It was observed that T11TS administration significantly downregulates the expression of matrix metalloproteinase-2 and -9 along with its ligand integrin αv and upregulates TIMP-1 and TIMP-2. In situ immunofluorescence and FACS results revealed that T11TS administration decreased the expression of the phenotypic markers (CD31/PECAM1, CD34), inhibiting the cell grip and also downregulating TGF-β1 expression (ELISA) from microglia cells in the glioma microenvironment. These results suggest that T11TS suppresses the expression of positive angiogenic growth factors and potentiates the expression of negative regulators in glioma-associated endothelial cells (ECs), resulting in an anti-angiogenic effect on glioma-induced angiogenesis.
Collapse
Affiliation(s)
- Manoj Kumar Singh
- Department of Laboratory Medicine, School of Tropical Medicine, 108, C.R. Avenue, Kolkata, 700073, West Bengal, India
| | | | | | | | | | | | | | | |
Collapse
|
155
|
Onishi M, Kurozumi K, Ichikawa T, Date I. Mechanisms of tumor development and anti-angiogenic therapy in glioblastoma multiforme. Neurol Med Chir (Tokyo) 2013; 53:755-63. [PMID: 24162241 PMCID: PMC4508716 DOI: 10.2176/nmc.ra2013-0200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Despite advances in surgical and medical therapy, glioblastoma multiforme (GBM) remains a fatal disease. There has been no significant increase in survival for patients with this disease over the last 20 years. Tumor vasculature formation and glioma cell invasion along the white matter tracts both play a pivotal role in glioma development. Angiogenesis and invasion are the major factors believed to be responsible for treatment resistance in tumors, and a better understanding of the glioma invasion and angiogenesis mechanisms will lead to the development of potential new treatments. In this review, we focus on the molecular characteristics of angiogenesis and invasion in human malignant glioma. We discuss bevacizumab and cilengitide, which are used to inhibit angiogenesis in GBM.
Collapse
Affiliation(s)
- Manabu Onishi
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
- Address reprint requests to: Manabu Onishi, MD, PhD, Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Okayama, Okayama 700-8558, Japan. e-mail:
| | - Kazuhiko Kurozumi
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
| | - Tomotsugu Ichikawa
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
| | - Isao Date
- Department of Neurological Surgery, Okayama, University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Okayama
| |
Collapse
|
156
|
Boroujerdi A, Welser-Alves JV, Milner R. Extensive vascular remodeling in the spinal cord of pre-symptomatic experimental autoimmune encephalomyelitis mice; increased vessel expression of fibronectin and the α5β1 integrin. Exp Neurol 2013; 250:43-51. [PMID: 24056042 DOI: 10.1016/j.expneurol.2013.09.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Revised: 08/20/2013] [Accepted: 09/10/2013] [Indexed: 10/26/2022]
Abstract
Alterations in vascular structure and function are a central component of demyelinating disease. In addition to blood-brain barrier (BBB) breakdown, which occurs early in the course of disease, recent studies have described angiogenic remodeling, both in multiple sclerosis tissue and in the mouse demyelinating model, experimental autoimmune encephalomyelitis (EAE). As the precise timing of vascular remodeling in demyelinating disease has yet to be fully defined, the purpose of the current study was to define the time-course of these events in the MOG35-55 EAE model. Quantification of endothelial cell proliferation and vessel density revealed that a large part of angiogenic remodeling in cervical spinal cord white matter occurs during the pre-symptomatic phase of EAE. At the height of vascular remodeling, blood vessels in the cervical spinal cord showed strong transient upregulation of fibronectin and the α5β1 integrin. In vitro experiments revealed that α5 integrin inhibition reduced brain endothelial cell proliferation under inflammatory conditions. Interestingly, loss of vascular integrity was evident in all vessels during the first 4-7days post-immunization, but after 14days, was localized predominantly to venules. Taken together, our data demonstrate that extensive vascular remodeling occurs during the pre-symptomatic phase of EAE and point to a potential role for the fibronectin-α5β1 integrin interaction in promoting vascular remodeling during demyelinating disease.
Collapse
Affiliation(s)
- Amin Boroujerdi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
157
|
Zhu B, Xu T, Yuan J, Guo X, Liu D. Transcriptome sequencing reveals differences between primary and secondary hair follicle-derived dermal papilla cells of the Cashmere goat (Capra hircus). PLoS One 2013; 8:e76282. [PMID: 24069460 PMCID: PMC3777969 DOI: 10.1371/journal.pone.0076282] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 08/22/2013] [Indexed: 12/30/2022] Open
Abstract
The dermal papilla is thought to establish the character and control the size of hair follicles. Inner Mongolia Cashmere goats (Capra hircus) have a double coat comprising the primary and secondary hair follicles, which have dramatically different sizes and textures. The Cashmere goat is rapidly becoming a potent model for hair follicle morphogenesis research. In this study, we established two dermal papilla cell lines during the anagen phase of the hair growth cycle from the primary and secondary hair follicles and clarified the similarities and differences in their morphology and growth characteristics. High-throughput transcriptome sequencing was used to identify gene expression differences between the two dermal papilla cell lines. Many of the differentially expressed genes are involved in vascularization, ECM-receptor interaction and Wnt/β-catenin/Lef1 signaling pathways, which intimately associated with hair follicle morphogenesis. These findings provide valuable information for research on postnatal morphogenesis of hair follicles.
Collapse
Affiliation(s)
- Bing Zhu
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Teng Xu
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Jianlong Yuan
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Xudong Guo
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
- * E-mail: (XG); (DL)
| | - Dongjun Liu
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
- * E-mail: (XG); (DL)
| |
Collapse
|
158
|
Moore SJ, Hayden Gephart MG, Bergen JM, Su YS, Rayburn H, Scott MP, Cochran JR. Engineered knottin peptide enables noninvasive optical imaging of intracranial medulloblastoma. Proc Natl Acad Sci U S A 2013; 110:14598-603. [PMID: 23950221 PMCID: PMC3767496 DOI: 10.1073/pnas.1311333110] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Central nervous system tumors carry grave clinical prognoses due to limited effectiveness of surgical resection, radiation, and chemotherapy. Thus, improved strategies for brain tumor visualization and targeted treatment are critically needed. We demonstrate that mouse cerebellar medulloblastoma (MB) can be targeted and illuminated with a fluorescent, engineered cystine knot (knottin) peptide that binds with high affinity to αvβ3, αvβ5, and α5β1 integrin receptors. This integrin-binding knottin peptide, denoted EETI 2.5F, was evaluated as a molecular imaging probe in both orthotopic and genetic models of MB. Following tail vein injection, fluorescence arising from dye-conjugated EETI 2.5F was localized to the tumor compared with the normal surrounding brain tissue, as measured by optical imaging. The imaging signal intensity correlated with tumor volume. Due to its unique ability to bind to α5β1 integrin, EETI 2.5F showed superior in vivo and ex vivo brain tumor imaging contrast compared with other engineered integrin-binding knottin peptides and with c(RGDfK), a well-studied integrin-binding peptidomimetic. Next, EETI 2.5F was fused to an antibody fragment crystallizable (Fc) domain (EETI 2.5F-Fc) to determine if a larger integrin-binding protein could also target intracranial brain tumors. EETI 2.5F-Fc, conjugated to a fluorescent dye, illuminated MB following i.v. injection and was able to distribute throughout the tumor parenchyma. In contrast, brain tumor imaging signals were not detected in mice injected with EETI 2.5F proteins containing a scrambled integrin-binding sequence, demonstrating the importance of target specificity. These results highlight the potential of using EETI 2.5F and EETI 2.5-Fc as targeted molecular probes for brain tumor imaging.
Collapse
Affiliation(s)
- Sarah J. Moore
- Departments of Bioengineering
- Center for Children’s Brain Tumors
- Stanford Cancer Institute, and
| | - Melanie G. Hayden Gephart
- Departments of Bioengineering
- Neurosurgery
- Developmental Biology
- Center for Children’s Brain Tumors
- Stanford Cancer Institute, and
| | - Jamie M. Bergen
- Departments of Bioengineering
- Center for Children’s Brain Tumors
- Stanford Cancer Institute, and
| | - YouRong S. Su
- Departments of Bioengineering
- Developmental Biology
- Genetics, and
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305
| | - Helen Rayburn
- Departments of Bioengineering
- Developmental Biology
- Genetics, and
- Center for Children’s Brain Tumors
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305
| | - Matthew P. Scott
- Departments of Bioengineering
- Developmental Biology
- Genetics, and
- Center for Children’s Brain Tumors
- Stanford Cancer Institute, and
- Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305
| | - Jennifer R. Cochran
- Departments of Bioengineering
- Chemical Engineering
- Center for Children’s Brain Tumors
- Stanford Cancer Institute, and
| |
Collapse
|
159
|
Marelli UK, Rechenmacher F, Sobahi TRA, Mas-Moruno C, Kessler H. Tumor Targeting via Integrin Ligands. Front Oncol 2013; 3:222. [PMID: 24010121 PMCID: PMC3757457 DOI: 10.3389/fonc.2013.00222] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 08/13/2013] [Indexed: 01/02/2023] Open
Abstract
Selective and targeted delivery of drugs to tumors is a major challenge for an effective cancer therapy and also to overcome the side-effects associated with current treatments. Overexpression of various receptors on tumor cells is a characteristic structural and biochemical aspect of tumors and distinguishes them from physiologically normal cells. This abnormal feature is therefore suitable for selectively directing anticancer molecules to tumors by using ligands that can preferentially recognize such receptors. Several subtypes of integrin receptors that are crucial for cell adhesion, cell signaling, cell viability, and motility have been shown to have an upregulated expression on cancer cells. Thus, ligands that recognize specific integrin subtypes represent excellent candidates to be conjugated to drugs or drug carrier systems and be targeted to tumors. In this regard, integrins recognizing the RGD cell adhesive sequence have been extensively targeted for tumor-specific drug delivery. Here we review key recent examples on the presentation of RGD-based integrin ligands by means of distinct drug-delivery systems, and discuss the prospects of such therapies to specifically target tumor cells.
Collapse
Affiliation(s)
- Udaya Kiran Marelli
- Institute for Advanced Study (IAS) and Center for Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München , Garching , Germany
| | | | | | | | | |
Collapse
|
160
|
Crosstalk between Fibroblast Growth Factor (FGF) Receptor and Integrin through Direct Integrin Binding to FGF and Resulting Integrin-FGF-FGFR Ternary Complex Formation. Med Sci (Basel) 2013. [DOI: 10.3390/medsci1010020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
161
|
Roy DC, Mooney NA, Raeman CH, Dalecki D, Hocking DC. Fibronectin matrix mimetics promote full-thickness wound repair in diabetic mice. Tissue Eng Part A 2013; 19:2517-26. [PMID: 23808793 DOI: 10.1089/ten.tea.2013.0024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
During tissue repair, fibronectin is converted from a soluble, inactive form into biologically active extracellular matrix (ECM) fibrils through a cell-dependent process. ECM fibronectin promotes numerous cell processes that are critical to tissue repair and regulates the assembly of other proteins into the matrix. Nonhealing wounds show reduced levels of ECM fibronectin. To functionally mimic ECM fibronectin, a series of fibronectin matrix mimetics was developed by directly coupling the matricryptic, heparin-binding fragment of the first type III repeat of fibronectin (FNIII1H) to various sequences from the integrin-binding domain (FNIII8-10). The recombinant proteins were produced as glutathione-S-transferase (GST)-tagged fusion proteins for ease of production and purification. Full-thickness, excisional wounds were produced in genetically diabetic mice, and fibronectin matrix mimetics were applied directly to the wounds. A significant enhancement of wound closure was observed by day 9 in response to GST/III1H,8-10 versus GST-treated controls (73.9%±4.1% vs. 58.1%±4.7% closure, respectively). Two weeks after injury, fibronectin matrix mimetic-treated wounds had developed a multi-layered epithelium that completely covered the wound space. Furthermore, significant increases in granulation tissue thickness were observed in response to treatment with GST/III1H,8-10 (4.05±0.93-fold), GST/III1H,8,10 (2.91±0.49-fold), or GST/III1H,8(RGD) (3.55±0.59-fold) compared with GST controls, and was accompanied by dense collagen deposition, the presence of myofibroblasts, and functional vasculature. Thus, the recombinant fibronectin matrix analogs normalized the impairment in repair observed in this chronic wound model and may provide a new approach to accelerate the healing of diabetic wounds.
Collapse
Affiliation(s)
- Daniel C Roy
- 1 Department of Biomedical Engineering, University of Rochester , Rochester, New York
| | | | | | | | | |
Collapse
|
162
|
Modulation of αvβ3- and α5β1-integrin-mediated adhesion by dehydro-β-amino acids containing peptidomimetics. Eur J Med Chem 2013; 66:258-68. [DOI: 10.1016/j.ejmech.2013.05.050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 04/18/2013] [Accepted: 05/22/2013] [Indexed: 02/08/2023]
|
163
|
Galvagni F, Orlandini M, Oliviero S. Role of the AP-1 transcription factor FOSL1 in endothelial cells adhesion and migration. Cell Adh Migr 2013; 7:408-11. [PMID: 24084233 DOI: 10.4161/cam.25894] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Vasculogenesis and angiogenesis, the fundamental processes by which new blood vessels are formed, involve the proliferation, migration, and remodeling of endothelial cells. Dynamic adhesion of endothelial cells to extracellular matrix plays a fundamental role in all these events. Key regulators of endothelial cells adhesion and migration are the αvβ3 and uPA-uPAR complexes. The αvβ3 integrin heterodimer is the receptor for extracellular matrix components such as vitronectin and is overexpressed on the cell surface of angiogenic endothelial cells, but not quiescent cells lining normal vessels. The uPA-uPAR complex contributes to extracellular matrix remodeling by mediating proteolytic activity at the leading edge of migrating cells. We recently reported that the FOSL1 transcription factor of the AP-1 family plays a pivotal role in the regulation of the level of the αvβ3 and uPA-uPAR complexes on the surface of endothelial cells. In this commentary, we review the current knowledge of αv and β3 transcriptional regulation in endothelial cells and discuss the role of FOSL1 in angiogenesis.
Collapse
Affiliation(s)
- Federico Galvagni
- Dipartimento di Biotecnologie; Chimica e Farmacia Università di Siena; via A.Moro; Siena, Italy
| | - Maurizio Orlandini
- Dipartimento di Biotecnologie; Chimica e Farmacia Università di Siena; via A.Moro; Siena, Italy
| | - Salvatore Oliviero
- Dipartimento di Biotecnologie; Chimica e Farmacia Università di Siena; via A.Moro; Siena, Italy; HUGEF; Torino, Italy
| |
Collapse
|
164
|
Abstract
Integrins are transmembrane receptors that mediate cell adhesion to neighboring cells and to the extracellular matrix. Here, the various modes in which integrin-mediated adhesion regulates intracellular signaling pathways impinging on cell survival, proliferation, and differentiation are considered. Subsequently, evidence that integrins also control crucial signaling cascades in cancer cells is discussed. Lastly, the important role of integrin signaling in tumor cells as well as in stromal cells that support cancer growth, metastasis, and therapy resistance indicates that integrin signaling may be an attractive target for (combined) cancer therapy strategies. Current approaches to target integrins in this context are reviewed.
Collapse
|
165
|
Kocatürk B, Versteeg HH. Tissue factor-integrin interactions in cancer and thrombosis: every Jack has his Jill. J Thromb Haemost 2013; 11 Suppl 1:285-93. [PMID: 23809132 DOI: 10.1111/jth.12222] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Tissue factor (TF) is a 47 kDa membrane protein that initiates coagulation by binding to FVII(a) and FX(a) and is a risk factor for thrombosis in many disease states. In addition to its coagulant activity, TF also influences cancer progression by triggering signaling effects via a group of G-protein coupled receptors named protease-activated receptors (PARs). TF localizes to cytoskeletal structures in migrating cells, influences cytoskeleton reorganization and promotes migration. Recently, integrins, important mediators of cell motility, have emerged as important binding partners for TF and influence both TF coagulant and PAR-2-dependent signaling functions. Direct binding of TF to integrins also impacts processes such as cell migration and signaling independent of PAR-2. A recently discovered alternatively spliced, soluble TF isoform also ligates integrins to augment angiogenesis, thus fuelling cancer progression. To date, the literature describes a complex interplay between different integrin subunits and distinct TF isoforms, but our understanding of TF-integrin bidirectional regulation remains clouded. In this review, we aim to summarize the existing knowledge on integrin-TF interaction and speculate on its relevance to physiology and pathology.
Collapse
Affiliation(s)
- B Kocatürk
- The Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | | |
Collapse
|
166
|
Mondal G, Barui S, Chaudhuri A. The relationship between the cyclic-RGDfK ligand and αvβ3 integrin receptor. Biomaterials 2013; 34:6249-60. [PMID: 23702147 DOI: 10.1016/j.biomaterials.2013.04.065] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Accepted: 04/30/2013] [Indexed: 11/18/2022]
Abstract
Ever since the finding that αvβ3 integrin receptors are over expressed on the endothelial cell surfaces of tumor vasculatures relative to normal resting vasculatures was disclosed in 1994, αvβ3 integrin receptor selective systems are finding increasing applications both for targeting anti-cancer drugs/genes selectively to tumor vasculatures and for imaging growing tumors. Among the cyclic peptide based integrin antagonists identified through both phage display and structure-activity studies, mainly αvβ3 integrin selective cyclic peptide c(RGDfK-) has found most widespread exploitations for targeting chemotherapeutic drugs/genes to both tumor and tumor vasculatures in anti-angiogenic cancer therapy. Herein we show that a lipopeptide containing widely acclaimed αvβ3 integrin receptor selective cyclic RGDfK ligand in its head-group area can effectively deliver genes into both the endothelial and tumor cells via all the three widely used integrin receptors namely αvβ3, αvβ5 & α5β1 integrins. We demonstrate that intravenous administration of the electrostatic complex of the cationic liposomes of an amphiphiles with cyclic RGDfK head-group and the anti-cancer p53 gene leads to significant tumor growth inhibition in a syngeneic mouse tumor model presumably through inducing apoptosis of tumor neovasculatures. The findings delineated herein provide experimental evidence that cyclic-RGDfK-ligand may not be that highly selective for αvβ3 integrin receptor as is popularly believed.
Collapse
Affiliation(s)
- Goutam Mondal
- Division of Lipid Science and Technology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | | | | |
Collapse
|
167
|
Carbonell WS, DeLay M, Jahangiri A, Park CC, Aghi MK. β1 integrin targeting potentiates antiangiogenic therapy and inhibits the growth of bevacizumab-resistant glioblastoma. Cancer Res 2013; 73:3145-54. [PMID: 23644530 DOI: 10.1158/0008-5472.can-13-0011] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Antiangiogenic therapies like bevacizumab offer promise for cancer treatment, but acquired resistance, which often includes an aggressive mesenchymal phenotype, can limit the use of these agents. Upregulation of β1 integrin (ITGB1) occurs in some bevacizumab-resistant glioblastomas (BRG) whereby, mediating tumor-microenvironment interactions, we hypothesized that it may mediate a mesenchymal-type resistance to antiangiogenic therapy. Immunostaining analyses of β1 integrin and its downstream effector kinase FAK revealed upregulation in 75% and 86% of BRGs, respectively, compared with pretreatment paired specimens. Furthermore, flow cytometry revealed eight-fold more β1 integrin in primary BRG cells compared with cells from bevacizumab-naïve glioblastomas (BNG). Fluorescence recovery after photobleaching of cells engineered to express a β1-GFP fusion protein indicated that the mobile β1 integrin fraction was doubled, and half-life of β1 integrin turnover in focal adhesions was reduced markedly in BRG cells compared with bevacizumab-responsive glioblastoma multiforme cells. Hypoxia, which was increased with acquisition of bevacizumab resistance, was associated with increased β1 integrin expression in cultured BNG cells. BRGs displayed an aggressive mesenchymal-like phenotype in vitro. We found that growth of BRG xenograft tumors was attenuated by the β1 antibody, OS2966, allowing a 20-fold dose reduction of bevacizumab per cycle in this model. Intracranial delivery of OS2966 through osmotic pumps over 28 days increased tumor cell apoptosis, decreased tumor cell invasiveness, and blunted the mesenchymal morphology of tumor cells. We concluded that β1 integrin upregulation in BRGs likely reflects an onset of hypoxia caused by antiangiogenic therapy, and that β1 inhibition is well tolerated in vivo as a tractable strategy to disrupt resistance to this therapy.
Collapse
Affiliation(s)
- W Shawn Carbonell
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA 94143, USA.
| | | | | | | | | |
Collapse
|
168
|
Jin M, Xiao R, Wang J, Liu X, Liu Y, Xue Z, Lv L, Zheng Y, Li Q. Low concentrations of the recombinant toxin protein rLj-RGD3 suppress TNF-α-induced human renal carcinoma cell invasion. Acta Biochim Biophys Sin (Shanghai) 2013; 45:377-82. [PMID: 23435195 DOI: 10.1093/abbs/gmt015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
A hallmark of renal cell carcinoma (RCC) invasion is the degradation of the extracellular matrix (ECM) by the local production of gelatinase enzymes. Matrix metalloproteinase-9 (MMP-9)-induced cancer cell invasion is one of the pivotal steps in cancer metastasis. It has been reported that tumor necrosis factor-α (TNF-α), a regulator of MMP-9, can induce invasion in human renal carcinoma cells. Previous work in our laboratory has shown that rLj-RGD3, a recombinant RGD (Arg-Gly-Asp)-toxin protein from the buccal gland secretion of Lampetra japonica, possesses anti-tumor activity. In this study, we demonstrated that rLj-RGD3 suppressed TNF-α-induced MMP-9 secretion in 786-0 cells (human renal carcinoma cells). To investigate the regulatory effect of rLj-RGD3 on TNF-α-induced MMP-9 secretion, we pre-treated cells with rLj-RGD3. Interestingly, rLj-RGD3 had no significant effect on the constitutive secretion of MMPs. However, low concentrations of rLj-RGD3 decreased TNF-α-induced MMP-9 secretion. Functional studies revealed that rLj-RGD3 induced apoptosis and significantly inhibited the proliferation, migration, and invasion of 786-0 cells. Furthermore, the actin architecture in cells pre-treated with rLj-RGD3 was aggregated and disorganized. Our findings suggest that rLj-RGD3 may be used as a potential drug in renal cancer therapy.
Collapse
Affiliation(s)
- Minli Jin
- College of Life Sciences, Liaoning Normal University, Dalian 116021, China
| | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Moore SJ, Leung CL, Norton HK, Cochran JR. Engineering agatoxin, a cystine-knot peptide from spider venom, as a molecular probe for in vivo tumor imaging. PLoS One 2013; 8:e60498. [PMID: 23573262 PMCID: PMC3616073 DOI: 10.1371/journal.pone.0060498] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 02/26/2013] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Cystine-knot miniproteins, also known as knottins, have shown great potential as molecular scaffolds for the development of targeted therapeutics and diagnostic agents. For this purpose, previous protein engineering efforts have focused on knottins based on the Ecballium elaterium trypsin inhibitor (EETI) from squash seeds, the Agouti-related protein (AgRP) neuropeptide from mammals, or the Kalata B1 uterotonic peptide from plants. Here, we demonstrate that Agatoxin (AgTx), an ion channel inhibitor found in spider venom, can be used as a molecular scaffold to engineer knottins that bind with high-affinity to a tumor-associated integrin receptor. METHODOLOGY/PRINCIPAL FINDINGS We used a rational loop-grafting approach to engineer AgTx variants that bound to αvβ3 integrin with affinities in the low nM range. We showed that a disulfide-constrained loop from AgRP, a structurally-related knottin, can be substituted into AgTx to confer its high affinity binding properties. In parallel, we identified amino acid mutations required for efficient in vitro folding of engineered integrin-binding AgTx variants. Molecular imaging was used to evaluate in vivo tumor targeting and biodistribution of an engineered AgTx knottin compared to integrin-binding knottins based on AgRP and EETI. Knottin peptides were chemically synthesized and conjugated to a near-infrared fluorescent dye. Integrin-binding AgTx, AgRP, and EETI knottins all generated high tumor imaging contrast in U87MG glioblastoma xenograft models. Interestingly, EETI-based knottins generated significantly lower non-specific kidney imaging signals compared to AgTx and AgRP-based knottins. CONCLUSIONS/SIGNIFICANCE In this study, we demonstrate that AgTx, a knottin from spider venom, can be engineered to bind with high affinity to a tumor-associated receptor target. This work validates AgTx as a viable molecular scaffold for protein engineering, and further demonstrates the promise of using tumor-targeting knottins as probes for in vivo molecular imaging.
Collapse
Affiliation(s)
- Sarah J. Moore
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
| | - Cheuk Lun Leung
- Department of Chemical Engineering, Stanford University, Stanford, California, United States of America
| | - Heidi K. Norton
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
| | - Jennifer R. Cochran
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
- Department of Chemical Engineering, Stanford University, Stanford, California, United States of America
- Stanford Cancer Institute and Bio-X Program, Stanford, California, United States of America
| |
Collapse
|
170
|
Endothelialization of chitosan porous conduits via immobilization of a recombinant fibronectin fragment (rhFNIII7-10). Acta Biomater 2013; 9:5643-52. [PMID: 23117145 DOI: 10.1016/j.actbio.2012.10.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/17/2012] [Accepted: 10/23/2012] [Indexed: 12/22/2022]
Abstract
The present study aimed to develop a pre-endothelialized chitosan (CH) porous hollowed scaffold for application in spinal cord regenerative therapies. CH conduits with different degrees of acetylation (DA; 4% and 15%) were prepared, characterized (microstructure, porosity and water uptake) and functionalized with a recombinant fragment of human fibronectin (rhFNIII(7-10)). Immobilized rhFNIII(7-10) was characterized in terms of amount ((125)I-radiolabelling), exposure of cell-binding domains (immunofluorescence) and ability to mediate endothelial cell (EC) adhesion and cytoskeletal rearrangement. Functionalized conduits revealed a linear increase in immobilized rhFNIII(7-10) with rhFNIII(7-10) concentration, and, for the same concentration, higher amounts of rhFNIII(7-10) on DA 4% compared with DA 15%. Moreover, rhFNIII(7-10) concentrations as low as 5 and 20μg ml(-1) in the coupling reaction were shown to provide DA 4% and 15% scaffolds, respectively, with levels of exposed cell-binding domains exceeding those observed on the control (DA 4% scaffolds incubated in a 20μg ml(-1) human fibronectin solution). These grafting conditions proved to be effective in mediating EC adhesion/cytoskeletal organization on CH with DA 4% and 15%, without affecting the endothelial angiogenic potential. rhFNIII(7-10) grafting to CH could be a strategy of particular interest in tissue engineering applications requiring the use of endothelialized porous matrices with tunable degradation rates.
Collapse
|
171
|
Tigges U, Boroujerdi A, Welser-Alves JV, Milner R. TNF-α promotes cerebral pericyte remodeling in vitro, via a switch from α1 to α2 integrins. J Neuroinflammation 2013; 10:33. [PMID: 23448258 PMCID: PMC3616978 DOI: 10.1186/1742-2094-10-33] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 02/15/2013] [Indexed: 01/07/2023] Open
Abstract
Background There is increasing evidence to suggest that pericytes play a crucial role in regulating the remodeling state of blood vessels. As cerebral pericytes are embedded within the extracellular matrix (ECM) of the vascular basal lamina, it is important to understand how individual ECM components influence pericyte remodeling behavior, and how cytokines regulate these events. Methods The influence of different vascular ECM substrates on cerebral pericyte behavior was examined in assays of cell adhesion, migration, and proliferation. Pericyte expression of integrin receptors was examined by flow cytometry. The influence of cytokines on pericyte functions and integrin expression was also examined, and the role of specific integrins in mediating these effects was defined by function-blocking antibodies. Expression of pericyte integrins within remodeling cerebral blood vessels was analyzed using dual immunofluorescence (IF) of brain sections derived from the animal model of multiple sclerosis, experimental autoimmune encephalomyelitis (EAE). Results Fibronectin and collagen I promoted pericyte proliferation and migration, but heparan sulfate proteoglycan (HSPG) had an inhibitory influence on pericyte behavior. Flow cytometry showed that cerebral pericytes express high levels of α5 integrin, and lower levels of α1, α2, and α6 integrins. The pro-inflammatory cytokine tumor necrosis factor (TNF)-α strongly promoted pericyte proliferation and migration, and concomitantly induced a switch in pericyte integrins, from α1 to α2 integrin, the opposite to the switch seen when pericytes differentiated. Inhibition studies showed that α2 integrin mediates pericyte adhesion to collagens, and significantly, function blockade of α2 integrin abrogated the pro-modeling influence of TNF-α. Dual-IF on brain tissue with the pericyte marker NG2 showed that while α1 integrin was expressed by pericytes in both stable and remodeling vessels, pericyte expression of α2 integrin was strongly induced in remodeling vessels in EAE brain. Conclusions Our results suggest a model in which ECM constituents exert an important influence on pericyte remodeling status. In this model, HSPG restricts pericyte remodeling in stable vessels, but during inflammation, TNF-α triggers a switch in pericyte integrins from α1 to α2, thereby stimulating pericyte proliferation and migration on collagen. These results thus define a fundamental molecular mechanism in which TNF-α stimulates pericyte remodeling in an α2 integrin-dependent manner.
Collapse
Affiliation(s)
- Ulrich Tigges
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla CA, 92037, USA
| | | | | | | |
Collapse
|
172
|
Bai XW, Li T, Qi JM. Clinical significance of expression of integrin α5β1 in gastric carcinoma. Shijie Huaren Xiaohua Zazhi 2013; 21:478-483. [DOI: 10.11569/wcjd.v21.i6.478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the expression of integrin α5β1 in gastric carcinoma and to analyze its relationship with the development and prognosis of gastric carcinoma.
METHODS: The expression of integrin α5β1 protein and mRNA was analyzed using immunohistochemistry and in situ hybridization (ISH) in 80 cases of gastric carcinoma and 50 cases of normal stomach tissue. The relationship between integrin α5β1 expression and clinicopathological parameters of gastric carcinoma was analyzed.
RESULTS: The positive rate of integrin α5β1 protein expression in gastric carcinoma was significantly higher than that in normal stomach tissue (60% vs 22%, P < 0.05). The positive rate of integrin α5β1 mRNA expression in gastric carcinoma was also significantly higher than that in normal stomach tissue (61.25% vs 24%, P < 0.05). The expression of integrin α5β1 in gastric carcinoma was closely associated with tumor differentiation, TNM stage, lymph node metastasis, and depth of invasion (all P < 0.05), but not with age, gender or pathological type (all P > 0.05).
CONCLUSION: High expression of integrin α5β1 has a close relation with the growth, invasion and metastasis of gastric carcinoma.
Collapse
|
173
|
SCUBE3 regulation of early lung cancer angiogenesis and metastatic progression. Clin Exp Metastasis 2013; 30:741-52. [PMID: 23420440 DOI: 10.1007/s10585-013-9575-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 02/01/2013] [Indexed: 12/12/2022]
Abstract
Signal peptide-CUB-EGF-like domain-containing protein 3 (SCUBE3) is strongly expressed in extremely invasive lung carcinoma. We showed in our previous study that SCUBE3 triggers the transforming growth factor-β pathway and subsequently promotes tumor angiogenesis and the epithelial-mesenchymal transition (EMT). However, the role of SCUBE3 in early tumor expansion hasn't been fully demonstrated in vivo. The present study used dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) to temporally assess tumor angiogenesis in SCUBE3-knockdown and control non-small-cell lung carcinoma (NSCLC) cancer cells in the early tumor stage (weeks 1-3). We further evaluated the metastatic potential of the SCUBE3-knockdown and control tumor cells using a circulating tumor cell (CTC) assay. The differences in gene expression profile between these cell lines were determined using microarray analysis. The results show that SCUBE3 knockdown was associated with lower vascular permeability in the tumor and effectively inhibited the metastatic potential of NSCLC, as evidenced by the decreased CTCs in the mice bearing SCUBE3-knockdown tumors. Microarray analysis revealed that several genes involved in angiogenesis and EMT were down-regulated in SCUBE3-knockdown tumors, including matrix metalloproteinases (MMPs) 2, 9, and 14, (MMP-2, MMP-9, and MMP-14, respectively), fibronectin (FN-1), lysyl oxidase (LOX), hairy/enhancer-of-split related with YRPW motif protein 1 (HEY1), early growth response protein 1 (EGR1), and interleukin 8 (IL-8). Together these data suggest that SCUBE3 is a potential target for pharmacological intervention. The findings of the present study also show that differences in vascular permeability precede the CTCs detection, indicating that DCE-MRI may be a sensitive biomarker for assessing tumor invasiveness.
Collapse
|
174
|
Rapraeger AC, Ell BJ, Roy M, Li X, Morrison OR, Thomas GM, Beauvais DM. Vascular endothelial-cadherin stimulates syndecan-1-coupled insulin-like growth factor-1 receptor and cross-talk between αVβ3 integrin and vascular endothelial growth factor receptor 2 at the onset of endothelial cell dissemination during angiogenesis. FEBS J 2013; 280:2194-206. [PMID: 23331867 DOI: 10.1111/febs.12134] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 01/05/2013] [Accepted: 01/11/2013] [Indexed: 12/23/2022]
Abstract
Vascular endothelial growth factor (VEGF)-stimulated angiogenesis depends on a cross-talk mechanism involving VEGF receptor 2 (VEGFR2), vascular endothelial (VE)-cadherin and the αVβ3 integrin. Because we have shown that αVβ3 integrin activation is dependent on its incorporation, along with the insulin-like growth factor-1 receptor (IGF1R) kinase, into a ternary receptor complex organized by the matrix receptor syndecan-1 (Sdc1), we questioned the role of this core complex in VEGF-stimulated angiogenesis. We find that the Sdc1-coupled ternary receptor complex is required for VEGF signalling and for stimulation of vascular endothelial cell migration by vascular endothelial cadherin (VE-cadherin) engagement. VE-cadherin binding to Fc/VE-cadherin extracellular domain chimera activates Sdc1-coupled IGF1R and αvβ3 integrin; this depends on VEGFR2 and c-Src activated by the cadherin. Blocking homotypic VE-cadherin engagement disrupts VEGF-stimulated cell migration, which is restored by clustering the cadherin in the absence of cell-cell adhesion. This cadherin-dependent stimulation requires VEGFR2 and IGF1R and is blocked by synstatin (SSTN)(92-119), a peptide that competitively disrupts the Sdc1-coupled ternary complex and prevents the αVβ3 integrin activation required for VEGFR2 activation. VEGFR2-stimulated angiogenesis in the mouse aortic ring explant assay is disrupted by SSTN, although only early in the process, suggesting that IGF1R coupling to Sdc1 and αVβ3 integrin comprises a core activation mechanism activated by VE-cadherin that is necessary for VEGFR2 and integrin activation in the initial stages of endothelial cell dissemination during angiogenesis.
Collapse
Affiliation(s)
- Alan C Rapraeger
- Department of Human Oncology, University of Wisconsin-Madison, WI 53705, USA.
| | | | | | | | | | | | | |
Collapse
|
175
|
Shih CH, Chiang TB, Wang WJ. Inhibition of integrins αv/α5-dependent functions in melanoma cells by an ECD-disintegrin acurhagin-C. Matrix Biol 2013; 32:152-9. [PMID: 23333557 DOI: 10.1016/j.matbio.2013.01.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 12/23/2012] [Accepted: 01/02/2013] [Indexed: 11/19/2022]
Abstract
Acurhagin-C, a Glu-Cys-Asp (ECD)-disintegrin from Agkistrodon acutus venom, has been reported as an endothelial apoptosis inducer, previously. Here we further evaluate its potential applications in cancer therapy. In vitro assays indicated that acurhagin-C not only may influence the cell viability at higher concentration, but also can potently and dose-dependently decrease cell proliferation in murine B16-F10 melanoma. Otherwise, it also had a dose-dependent inhibition on B16-F10 cell adhesion to extracellular matrices, collagen VI, gelatin B and fibronectin, as well as disturbed transendothelial migration of B16-F10 cell. Morphological study found that acurhagin-C dramatically affected B16-F10 cell adhesion to immobilized fibronectin, leading to the formation of multicellular aggregates with rounded shape. Detected by flow cytometry, acurhagin-C was able to induce B16-F10 cell apoptosis and alter cell cycle distribution through its interactions with integrins αv/α5, and thereafter initiation the apoptotic pathways of caspase-8/-9. Furthermore, acurhagin-C could synergistically enhance the anti-proliferative activity of methotrexate in B16-F10 cells and human melanoma SK-MEL-1 cells, without diminishing the growth of human epidermal melanocytes. Taken together, acurhagin-C proved to be a potent inhibitor of integrin-based functions in melanoma cells by activating the complex apoptotic pathways.
Collapse
Affiliation(s)
- Chun-Ho Shih
- Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan, Taiwan
| | | | | |
Collapse
|
176
|
Schaffner F, Ray AM, Dontenwill M. Integrin α5β1, the Fibronectin Receptor, as a Pertinent Therapeutic Target in Solid Tumors. Cancers (Basel) 2013; 5:27-47. [PMID: 24216697 PMCID: PMC3730317 DOI: 10.3390/cancers5010027] [Citation(s) in RCA: 157] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/09/2013] [Accepted: 01/11/2013] [Indexed: 12/11/2022] Open
Abstract
Integrins are transmembrane heterodimeric proteins sensing the cell microenvironment and modulating numerous signalling pathways. Changes in integrin expression between normal and tumoral cells support involvement of specific integrins in tumor progression and aggressiveness. This review highlights the current knowledge about α5β1 integrin, also called the fibronectin receptor, in solid tumors. We summarize data showing that α5β1 integrin is a pertinent therapeutic target expressed by tumoral neovessels and tumoral cells. Although mainly evaluated in preclinical models, α5β1 integrin merits interest in particular in colon, breast, ovarian, lung and brain tumors where its overexpression is associated with a poor prognosis for patients. Specific α5β1 integrin antagonists will be listed that may represent new potential therapeutic agents to fight defined subpopulations of particularly aggressive tumors.
Collapse
Affiliation(s)
- Florence Schaffner
- UMR 7213 CNRS, Laboratoire de Biophotonique et Pharmacologie, Tumoral Signaling and Therapeutic Targets, Université de Strasbourg, Faculté de Pharmacie, 67401 Illkirch, France.
| | | | | |
Collapse
|
177
|
Newman AC, Chou W, Welch-Reardon KM, Fong AH, Popson SA, Phan DT, Sandoval DR, Nguyen DP, Gershon PD, Hughes CCW. Analysis of stromal cell secretomes reveals a critical role for stromal cell-derived hepatocyte growth factor and fibronectin in angiogenesis. Arterioscler Thromb Vasc Biol 2013; 33:513-22. [PMID: 23288153 DOI: 10.1161/atvbaha.112.300782] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Angiogenesis requires tightly coordinated crosstalk between endothelial cells (ECs) and stromal cells, such as fibroblasts and smooth muscle cells. The specific molecular mechanisms moderating this process are still poorly understood. METHODS AND RESULTS Stromal cell-derived factors are essential for EC sprouting and lumen formation. We therefore compared the abilities of 2 primary fibroblast isolates and a primary smooth muscle cell isolate to promote in vitro angiogenesis, and analyzed their secretomes using a combination of nano liquid chromatography-mass spectrometry/mass spectrometry, quantitative PCR, and ELISA. Each isolate exhibited a different level of angiogenic ability. Using quantitative MS, we then compared the secretomes of a fibroblast isolate exhibiting low angiogenic activity, a fibroblast isolate exhibiting high angiogenic activity, and human umbilical vein ECs. High angiogenic fibroblast supernatants exhibited an overabundance of proteins associated with extracellular matrix constituents compared with low angiogenic fibroblasts or ECs. Finally, small interfering RNA technology and purified protein were used to confirm a role for stromal cell-derived hepatocyte growth factor and fibronectin in inducing EC sprouting. CONCLUSIONS Differences in stromal cell ability to induce angiogenesis are a result of differences in the secreted proteomes of both extracellular matrix proteins and proangiogenic growth factors.
Collapse
Affiliation(s)
- Andrew C Newman
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA 92697, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
178
|
Goswami S. Importance of integrin receptors in the field of pharmaceutical & medical science. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/abc.2013.32028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
179
|
Adil M, Belur L, Pearce TR, Levine RM, Tisdale AW, Sorenson BS, McIvor RS, Kokkoli E. PR_b functionalized stealth liposomes for targeted delivery to metastatic colon cancer. Biomater Sci 2013; 1:393-401. [DOI: 10.1039/c2bm00128d] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
180
|
Xu S, Olenyuk BZ, Okamoto CT, Hamm-Alvarez SF. Targeting receptor-mediated endocytotic pathways with nanoparticles: rationale and advances. Adv Drug Deliv Rev 2013; 65:121-38. [PMID: 23026636 PMCID: PMC3565049 DOI: 10.1016/j.addr.2012.09.041] [Citation(s) in RCA: 324] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 09/13/2012] [Accepted: 09/20/2012] [Indexed: 12/22/2022]
Abstract
Targeting of drugs and their carrier systems by using receptor-mediated endocytotic pathways was in its nascent stages 25 years ago. In the intervening years, an explosion of knowledge focused on design and synthesis of nanoparticulate delivery systems as well as elucidation of the cellular complexity of what was previously-termed receptor-mediated endocytosis has now created a situation when it has become possible to design and test the feasibility of delivery of highly specific nanoparticle drug carriers to specific cells and tissue. This review outlines the mechanisms governing the major modes of receptor-mediated endocytosis used in drug delivery and highlights recent approaches using these as targets for in vivo drug delivery of nanoparticles. The review also discusses some of the inherent complexity associated with the simple shift from a ligand-drug conjugate versus a ligand-nanoparticle conjugate, in terms of ligand valency and its relationship to the mode of receptor-mediated internalization.
Collapse
Affiliation(s)
- Shi Xu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, USA 90033
| | - Bogdan Z. Olenyuk
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, USA 90033
| | - Curtis T. Okamoto
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, USA 90033
| | - Sarah F. Hamm-Alvarez
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA, USA 90033
| |
Collapse
|
181
|
Zhao G, Rodriguez BL. Molecular targeting of liposomal nanoparticles to tumor microenvironment. Int J Nanomedicine 2012; 8:61-71. [PMID: 23293520 PMCID: PMC3534304 DOI: 10.2147/ijn.s37859] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Liposomes are biodegradable and can be used to deliver drugs at a much higher concentration in tumor tissues than in normal tissues. Both passive and active drug delivery by liposomal nanoparticles can significantly reduce the toxic side effects of anticancer drugs and enhance the therapeutic efficacy of the drugs delivered. Active liposomal targeting to tumors is achieved by recognizing specific tumor receptors through tumor-specific ligands or antibodies coupled onto the surface of the liposomes, or by stimulus-sensitive drug carriers such as acid-triggered release or enzyme-triggered drug release. Tumors are often composed of tumor cells and nontumor cells, which include endothelial cells, pericytes, fibroblasts, stromal, mesenchymal cells, innate, and adaptive immune cells. These nontumor cells thus form the tumor microenvironment, which could be targeted and modified so that it is unfavorable for tumor cells to grow. In this review, we briefly summarized articles that had taken advantage of liposomal nanoparticles as a carrier to deliver anticancer drugs to the tumor microenvironment, and how they overcame obstacles such as nonspecific uptake, interaction with components in blood, and toxicity. Special attention is devoted to the liposomal targeting of anticancer drugs to the endothelium of tumor neovasculature, tumor associated macrophages, fibroblasts, and pericytes within the tumor microenvironment.
Collapse
Affiliation(s)
- Gang Zhao
- Institute of Materia Medica, Shandong Academy of Medical Science, Shandong, China
| | | |
Collapse
|
182
|
A randomized phase II study of cilengitide (EMD 121974) in patients with metastatic melanoma. Melanoma Res 2012; 22:294-301. [PMID: 22668797 DOI: 10.1097/cmr.0b013e32835312e4] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Cilengitide (EMD 121974) is a selective inhibitor of integrins αvβ3 and αvβ5. The αvβ3 promotes the proliferation of tumor-associated endothelial cells and potentially the survival of melanoma cells. We conducted a randomized phase II trial in patients with metastatic melanoma to evaluate the clinical efficacy of cilengitide. Patients with stage IV or unresectable stage III melanoma who were either chemonaive or who had previously received one systemic therapy were enrolled. Patients were randomly assigned to either 500 or 2000 mg of cilengitide administered intravenously twice weekly. The primary aim of this study was to determine the progression-free survival rate at 8 weeks. Tumor samples and blood samples were collected for pharmacodynamic and pharmacokinetic studies. Twenty-nine patients were enrolled, of whom 26 were treated (14 at 500 mg and 12 at 2000 mg). Among those treated, only three were progression free at 8 weeks: two in the 500 mg arm and one in the 2000 mg arm. One patient in the 2000 mg arm showed a prolonged partial response after an initial 28% enlargement of her target lesions. The treatment was well tolerated without clinically significant adverse events. The sole responder and one of two patients with stable disease had no αvβ3 expression at baseline. Overall, αvβ3 expression was decreased by day 8 of the treatment (P=0.05). Cilengitide was well tolerated by patients in both the treatment arms but had minimal clinical efficacy as a single-agent therapy for metastatic melanoma, and the efficacy was not related to baseline αvβ3 expression.
Collapse
|
183
|
Martin S, Janouskova H, Dontenwill M. Integrins and p53 pathways in glioblastoma resistance to temozolomide. Front Oncol 2012; 2:157. [PMID: 23120745 PMCID: PMC3484330 DOI: 10.3389/fonc.2012.00157] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2012] [Accepted: 10/16/2012] [Indexed: 01/08/2023] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor. Surgical resection, postoperative radiotherapy plus concomitant and adjuvant chemotherapy with temozolomide (TMZ) is the standard of care for newly diagnosed glioblastoma. In the past decade, efforts have been made to decipher genomic and core pathway alterations to identify clinically relevant glioblastoma subtypes. Based on these studies and more academic explorations, new potential therapeutic targets were found and several targeting agents were developed. Such molecules should hopefully overcome the resistance of glioblastoma to the current therapy. One of the hallmarks of glioblastoma subtypes was the enrichment of extracellular matrix/invasion-related genes. Integrins, which are cell adhesion molecules important in glioma cell migration/invasion and angiogenesis were one of those genes. Integrins seem to be pertinent therapeutic targets and antagonists recently reached the clinic. Although the p53 pathway appears often altered in glioblastoma, conflicting results can be found in the literature about the clinically relevant impact of the p53 status in the resistance to TMZ. Here, we will summarize the current knowledge on (1) integrin expression, (2) p53 status, and (3) relationship between integrins and p53 to discuss their potential impact on the resistance of glioblastoma to temozolomide.
Collapse
Affiliation(s)
- Sophie Martin
- Laboratory of Biophotonics and Pharmacology, UMR 7213 CNRS, Faculté de Pharmacie, Université de Strasbourg Illkirch, France
| | | | | |
Collapse
|
184
|
Kachgal S, Mace KA, Boudreau NJ. The dual roles of homeobox genes in vascularization and wound healing. Cell Adh Migr 2012; 6:457-70. [PMID: 23076135 PMCID: PMC3547888 DOI: 10.4161/cam.22164] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Homeobox genes represent a family of highly conserved transcription factors originally discovered to regulate organ patterning during development. More recently, several homeobox genes were shown to affect processes in adult tissue, including angiogenesis and wound healing. Whereas a subset of members of the Hox-family of homeobox genes activate growth and migration to promote angiogenesis or wound healing, other Hox genes function to restore or maintain quiescent, differentiated tissue function. Pathological tissue remodeling is linked to differential expression of activating or stabilizing Hox genes and dysregulation of Hox expression can contribute to disease progression. Studies aimed at understanding the role and regulation of Hox genes have provided insight into how these potent morphoregulatory genes can be applied to enhance tissue engineering or limit cancer progression.
Collapse
Affiliation(s)
- Suraj Kachgal
- Surgical Research Laboratory, Department of Surgery, University of California, San Francisco, San Francisco, CA USA
| | | | | |
Collapse
|
185
|
|
186
|
Besse B, Tsao LC, Chao DT, Fang Y, Soria JC, Almokadem S, Belani CP. Phase Ib safety and pharmacokinetic study of volociximab, an anti-α5β1 integrin antibody, in combination with carboplatin and paclitaxel in advanced non-small-cell lung cancer. Ann Oncol 2012; 24:90-6. [PMID: 22904239 DOI: 10.1093/annonc/mds281] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND This phase Ib study evaluated volociximab, an anti-α5β1 integrin antibody, in combination with carboplatin (Eli Lilly and Co., Indianapolis, IN) and paclitaxel (Taxol) in advanced, untreated non-small-cell lung cancer (NSCLC). PATIENTS AND METHODS Three cohorts were treated with volociximab (10, 20, or 30 mg/kg) for up to six 3-week cycles in combination with carboplatin-paclitaxel chemotherapy and continued as maintenance therapy for patients with stable disease (SD) or better. Dose-limiting toxic effects, adverse events (AEs), pharmacokinetics, and anti-volociximab antibodies were assessed. RESULTS A maximum tolerated dose was not reached up to the maximum planned dose of 30 mg/kg. In 29 patients who received volociximab, the most common grade≥3 AEs were neutropenia (24%), hyponatremia (17%), and fatigue (10%). Three patients experienced volociximab-related serious AEs. No hemorrhages were observed. Of 33 patients enrolled, 8 (24%) achieved a partial response and 17 (52%) had SD. The median progression-free survival was 6.3 months (95% confidence interval 5.5-8.1). Levels of potential biomarkers of angiogenesis or metastasis were reduced following six cycles of treatment. CONCLUSIONS Volociximab combined with carboplatin and paclitaxel was generally well-tolerated and showed preliminary evidence of efficacy in advanced NSCLC.
Collapse
Affiliation(s)
- B Besse
- Cancer Medicine/Thoracic Unit, Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | |
Collapse
|
187
|
Pearce TR, Shroff K, Kokkoli E. Peptide targeted lipid nanoparticles for anticancer drug delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2012; 24:3803-22, 3710. [PMID: 22674563 DOI: 10.1002/adma.201200832] [Citation(s) in RCA: 138] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2012] [Indexed: 05/21/2023]
Abstract
Encapsulating anticancer drugs in nanoparticles has proven to be an effective mechanism to alter the pharmacokinetic and pharmacodynamic profiles of the drugs, leading to clinically useful cancer therapeutics like Doxil and DaunoXome. Underdeveloped tumor vasculature and lymphatics allow these first-generation nanoparticles to passively accumulate within the tumor, but work to create the next-generation nanoparticles that actively participate in the tumor targeting process is underway. Lipid nanoparticles functionalized with targeting peptides are among the most often studied. The goal of this article is to review the recently published literature of targeted nanoparticles to highlight successful designs that improved in vivo tumor therapy, and to discuss the current challenges of designing these nanoparticles for effective in vivo performance.
Collapse
Affiliation(s)
- Timothy R Pearce
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | |
Collapse
|
188
|
Hung WY, Huang KH, Wu CW, Chi CW, Kao HL, Li AFY, Yin PH, Lee HC. Mitochondrial dysfunction promotes cell migration via reactive oxygen species-enhanced β5-integrin expression in human gastric cancer SC-M1 cells. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1820:1102-1110. [PMID: 22561002 DOI: 10.1016/j.bbagen.2012.04.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 03/19/2012] [Accepted: 04/18/2012] [Indexed: 02/06/2023]
Abstract
BACKGROUND Mitochondrial dysfunction has been shown to promote cancer cell migration. However, molecular mechanism by which mitochondrial dysfunction enhances gastric cancer (GC) cell migration remains unclear. METHODS Mitochondria specific inhibitors, oligomycin and antimycin A, were used to induce mitochondrial dysfunction and to enhance cell migration of human gastric cancer SC-M1 cells. Antioxidant N-acetylcysteine (NAC) was used for evaluating the effect of reactive oxygen species (ROS). Protein expressions of epithelial-to-mesenchymal transition (EMT) markers and the cell-extracellular matrix (ECM) adhesion molecules, the integrin family, were analyzed. A migratory subpopulation of SC-M1 cells (SC-M1-3rd) was selected using a transwell assay for examining the association of mitochondrial bioenergetic function, intracellular ROS content and β5-integrin expression. Clinicopathologic characteristics of β5-integrin expression were analyzed in GC specimens by immunohistochemical staining. RESULTS Treatments with mitochondrial inhibitors elevated mitochondria-generated ROS and cell migration of SC-M1 cells. The protein expression of β5-integrin and cell surface expression of αvβ5-integrin were upregulated, and which were suppressed by NAC. Pretreatments with NAC and anti-αvβ5-integrin neutralizing antibody respectively prevented the mitochondrial dysfunction-induced cell migration. The selected migratory SC-M1-3rd cells showed impaired mitochondrial function, higher mitochondria-generated ROS, and increased β5-integrin expression. The migration ability was also repressed by anti-αvβ5-integrin neutralizing antibody. In clinical specimens, GCs with higher β5-integrin protein expression had more aggressive behavior. In conclusion, mitochondrial dysfunction may lead to GC progression by enhancing migration through mitochondria-generated ROS mediated β5-integrin expression. GENERAL SIGNIFICANCE These results support the role of mitochondrial dysfunction in GC progression.
Collapse
Affiliation(s)
- Wen-Yi Hung
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, and Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
189
|
Li L, Welser-Alves J, van der Flier A, Boroujerdi A, Hynes RO, Milner R. An angiogenic role for the α5β1 integrin in promoting endothelial cell proliferation during cerebral hypoxia. Exp Neurol 2012; 237:46-54. [PMID: 22721769 DOI: 10.1016/j.expneurol.2012.06.005] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 06/01/2012] [Accepted: 06/09/2012] [Indexed: 12/11/2022]
Abstract
Fibronectin is a critical regulator of vascular modelling, both in development and in the adult. In the hypoxic adult central nervous system (CNS), fibronectin is induced on angiogenic vessels, and endothelial cells show strong induction of the two fibronectin receptors α5β1 and αvβ3 integrins. In a previous study, we found that the αvβ3 integrin is dispensable for hypoxic-induced cerebral angiogenesis, but a role for the endothelial α5β1 integrin was suggested. To directly investigate the role of endothelial α5 integrin in cerebral angiogenesis, wild-type mice and mice lacking α5 integrin expression in endothelial cells (α5-EC-KO) were subject to hypoxia (8% O(2)) for 0, 2, 4, 7 or 14 days. Quantification of cerebral vessel density and endothelial-specific proteins claudin-5 and Glut-1 revealed that α5-EC-KO mice displayed an attenuated angiogenic response, which correlated with delayed endothelial proliferation. α5-EC-KO mice showed no defect in the ability to organize a cerebrovascular fibronectin matrix, and no compensatory increase in vascular αvβ3 integrin expression. Consistent with these findings, primary α5KO brain endothelial cells (BEC) in culture exhibited delayed growth and proliferation. Taken together, these studies demonstrate an important angiogenic role for the α5β1 integrin in promoting BEC proliferation in response to cerebral hypoxia.
Collapse
Affiliation(s)
- Longxuan Li
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
190
|
Spiegel J, Mas-Moruno C, Kessler H, Lubell WD. Cyclic Aza-peptide Integrin Ligand Synthesis and Biological Activity. J Org Chem 2012; 77:5271-8. [DOI: 10.1021/jo300311q] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Jochen Spiegel
- Department Chemie, Institute
for Advanced Study, Technische Universität München, Lichtenbergstrasse 4, 85747 Garching, Germany
- Département de Chimie, Université de Montréal, C.P. 6128, Succursale
Centre Ville, Montréal, Québec H3C 3J7, Canada
| | - Carlos Mas-Moruno
- Department Chemie, Institute
for Advanced Study, Technische Universität München, Lichtenbergstrasse 4, 85747 Garching, Germany
| | - Horst Kessler
- Department Chemie, Institute
for Advanced Study, Technische Universität München, Lichtenbergstrasse 4, 85747 Garching, Germany
- Chemistry Department, Faculty
of Science, King Abdulaziz University,
P.O. Box 80203, Jeddah 21589, Saudi Arabia
| | - William D. Lubell
- Département de Chimie, Université de Montréal, C.P. 6128, Succursale
Centre Ville, Montréal, Québec H3C 3J7, Canada
| |
Collapse
|
191
|
DeVolder R, Kong HJ. Hydrogels for in vivo
-like three-dimensional cellular studies. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2012; 4:351-65. [DOI: 10.1002/wsbm.1174] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
192
|
Wu SD, Ma YS, Fang Y, Liu LL, Fu D, Shen XZ. Role of the microenvironment in hepatocellular carcinoma development and progression. Cancer Treat Rev 2012; 38:218-25. [DOI: 10.1016/j.ctrv.2011.06.010] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Revised: 06/22/2011] [Accepted: 06/27/2011] [Indexed: 02/07/2023]
|
193
|
Delouvrié B, Al-Kadhimi K, Arnould JC, Barry ST, Cross DAE, Didelot M, Gavine PR, Germain H, Harris CS, Hughes AM, Jude DA, Kendrew J, Lambert-van der Brempt C, Lohmann JJ, Ménard M, Mortlock AA, Pass M, Rooney C, Vautier M, Vincent JL, Warin N. Structure-activity relationship of a series of non peptidic RGD integrin antagonists targeting α5β1: part 1. Bioorg Med Chem Lett 2012; 22:4111-6. [PMID: 22575869 DOI: 10.1016/j.bmcl.2012.04.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 04/10/2012] [Accepted: 04/11/2012] [Indexed: 11/28/2022]
Abstract
Potent antagonists of the integrin α(5)β(1), which are RGD mimetics built from tyrosine are described. This letter describes the optimization of in vitro potency obtained by variation of two parts of the molecule, the basic group and the linker between the basic group and the phenyl central core.
Collapse
Affiliation(s)
- Bénédicte Delouvrié
- AstraZeneca, Centre de Recherches, Z.I. La Pompelle, B.P. 1050, Chemin de Vrilly, 51689 Reims, Cedex 2, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Mikuła-Pietrasik J, Kuczmarska A, Kucińska M, Murias M, Wierzchowski M, Winckiewicz M, Staniszewski R, Bręborowicz A, Książek K. Resveratrol and its synthetic derivatives exert opposite effects on mesothelial cell-dependent angiogenesis via modulating secretion of VEGF and IL-8/CXCL8. Angiogenesis 2012; 15:361-76. [PMID: 22451299 PMCID: PMC3409370 DOI: 10.1007/s10456-012-9266-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 03/13/2012] [Indexed: 11/19/2022]
Abstract
We examined the effect of resveratrol (RVT) and its two derivatives (3,3′,4,4′-tetrahydroxy-trans-stilbene and 3,3′,4,4′,5,5′-hexahydroxy-trans-stilbene) on human peritoneal mesothelial cell (HPMC)-dependent angiogenesis in vitro. To this end, angiogenic activity of endothelial cells (HUVEC, HMVEC, and HMEC-1) was monitored upon their exposure to conditioned medium (CM) from young and senescent HPMCs treated with stilbenes or to stilbenes themselves. Results showed that proliferation and migration of endothelial cells were inhibited in response to indirect (HPMC-dependent) or direct RVT activity. This effect was associated with decreased secretion of VEGF and IL-8/CXCL8 by HPMCs treated with RVT, which confirmed the experiments with recombinant forms of these angiogenic agents. Angiogenic activity of endothelial cells treated with CM from HPMCs exposed to RVT analogues was more effective. Improved migration was particularly evident in cells exposed to CM from senescent HPMCs. Upon direct treatment, RVT derivatives stimulated proliferation (but not migration) of HUVECs, and failed to affect the behaviour of HMVEC and HMEC-1 cells. These compounds stimulated production of VEGF and IL-8/CXCL8 by HPMCs. Studies with neutralizing antibodies against angiogenic factors revealed that augmented angiogenic reactions of endothelial cells exposed to CM from HPMC treated with RVT analogues were related to enhanced production of VEGF and IL-8/CXCL8. Collectively, these findings indicate that RVT and its synthetic analogues divergently alter the secretion of the angiogenic factors by HPMCs, and thus modulate HPMC-dependent angiogenic responses in the opposite directions. This may have implications for the attempts of practical employment of the stilbenes for treatment of pathologies proceeding with abnormal vascularisation of the peritoneal tissue.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Pathophysiology, Poznań University of Medical Sciences, Święcickiego 6 Str., 60-781, Poznań, Poland
| | | | | | | | | | | | | | | | | |
Collapse
|
195
|
Schiefner A, Gebauer M, Skerra A. Extra-domain B in oncofetal fibronectin structurally promotes fibrillar head-to-tail dimerization of extracellular matrix protein. J Biol Chem 2012; 287:17578-17588. [PMID: 22442152 DOI: 10.1074/jbc.m111.303131] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The type III extra-domain B (ED-B) is specifically spliced into fibronectin (Fn) during embryogenesis and neoangiogenesis, including many cancers. The x-ray structure of the recombinant four-domain fragment Fn(III)7B89 reveals a tightly associated, extended head-to-tail dimer, which is stabilized via pair-wise shape and charge complementarity. A tendency toward ED-B-dependent dimer formation in solution was supported by size exclusion chromatography and analytical ultracentrifugation. When amending the model with the known three-dimensional structure of the Fn(III)10 domain, its RGD loop as well as the adhesion synergy region in Fn(III)9-10 become displayed on the same face of the dimer; this should allow simultaneous binding of at least two integrins and, thus, receptor clustering on the cell surface and intracellular signaling. Insertion of ED-B appears to stabilize overall head-to-tail dimerization of two separate Fn chains, which, together with alternating homodimer formation via disulfide bridges at the C-terminal Fn tail, should lead to the known macromolecular fibril formation.
Collapse
Affiliation(s)
- André Schiefner
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, 85350 Freising-Weihenstephan, Germany
| | - Michaela Gebauer
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, 85350 Freising-Weihenstephan, Germany
| | - Arne Skerra
- Munich Center for Integrated Protein Science (CIPS-M) and Lehrstuhl für Biologische Chemie, Technische Universität München, 85350 Freising-Weihenstephan, Germany.
| |
Collapse
|
196
|
Minea R, Helchowski C, Rubino B, Brodmann K, Swenson S, Markland F. Development of a chimeric recombinant disintegrin as a cost-effective anti-cancer agent with promising translational potential. Toxicon 2012; 59:472-86. [PMID: 21354198 PMCID: PMC3130806 DOI: 10.1016/j.toxicon.2011.02.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2010] [Revised: 02/15/2011] [Accepted: 02/16/2011] [Indexed: 12/11/2022]
Abstract
Vicrostatin (VCN) is a chimeric recombinant disintegrin generated in Origami B (DE3) Escherichia coli as a genetic fusion between the C-terminal tail of a viperid disintegrin echistatin and crotalid disintegrin contortrostatin (CN). The therapeutic modulation of multiple integrin pathways via soluble disintegrins was previously shown by us and others to elicit potent anti-angiogenic and anti-metastatic effects in several animal cancer models. Despite these favorable attributes, these polypeptides are notoriously difficult to produce recombinantly in significant quantity due to their structure which requires the correct pairing of multiple disulfide bonds for biological activity. In this report, we show that VCN can be reliably produced in large amounts (yields in excess of 200 mg of active purified disintegrin per liter of bacterial culture) in Origami B (DE3), an E. coli expression strain engineered to support the folding of disulfide-rich heterologous proteins directly in its oxidative cytoplasmic compartment. VCN retains the integrin binding specificity of both parental molecules it was derived from, but with a different binding affinity profile. While competing for the same integrin receptors that are preferentially upregulated in the tumor microenvironment, VCN exerts a potent inhibitory effect on endothelial cell (EC) migration and tube formation in a dose-dependent manner, by forcing these cells to undergo significant actin cytoskeleton reorganization when exposed to this agent in vitro. Moreover, VCN has a direct effect on breast cancer cells inhibiting their in vitro motility. In an effort to address our main goal of developing a clinically relevant delivery method for recombinant disintegrins, VCN was efficiently packaged in liposomes (LVCN) and evaluated in vivo in an animal breast cancer model. Our data demonstrate that LVCN is well tolerated, its intravenous administration inducing a significant delay in tumor growth and an increase in animal survival, results that can be partially explained by potent tumor apoptotic effects.
Collapse
Affiliation(s)
- Radu Minea
- Department of Biochemistry and Molecular Biology, University of Southern California, Keck School of Medicine, 1303 N Mission Road, CRL-106, Los Angeles, CA 90033, USA
| | | | | | | | | | | |
Collapse
|
197
|
Prager GW, Poettler M, Unseld M, Zielinski CC. Angiogenesis in cancer: Anti-VEGF escape mechanisms. Transl Lung Cancer Res 2012; 1:14-25. [PMID: 25806151 PMCID: PMC4367591 DOI: 10.3978/j.issn.2218-6751.2011.11.02] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 11/29/2011] [Indexed: 12/21/2022]
Abstract
It is now widely accepted that tumor-angiogenesis plays a crucial role in tumor growth, tumor propagation and metastasis formation. Among several angiogenic activators, the vascular endothelial growth factor (VEGF) and its receptors represent one of the major inducers of tumor angiogenesis. Thus, this system has become the focus of therapeutic interventions, which led to the approval of the anti-VEGF blocking antibody bevacizumab and the VEGFR-2 pathway inhibitors pazopanib, sorafenib and sunitinib. However, not every cancer patient benefits from such treatment or finally becomes resistant to anti-VEGF approaches; others are suffering from adverse effects. Thus, there is an urgent need for a better understanding of VEGF-independent mechanisms leading to angiogenesis in cancer. This review focuses on anti-VEGF escape mechanisms of tumor cells and its microenvironment.
Collapse
Affiliation(s)
- Gerald W Prager
- Medical University of Vienna, Comprehensive Cancer Center Vienna, Department of Medicine I, Austria
| | - Marina Poettler
- Medical University of Vienna, Comprehensive Cancer Center Vienna, Department of Medicine I, Austria
| | - Matthias Unseld
- Medical University of Vienna, Comprehensive Cancer Center Vienna, Department of Medicine I, Austria
| | - Christoph C Zielinski
- Medical University of Vienna, Comprehensive Cancer Center Vienna, Department of Medicine I, Austria
| |
Collapse
|
198
|
Mousa SA. Comparative Pharmacodynamic Assessment of the Antiangiogenesis Activity of Heparin and Low-Molecular-Weight Heparin Fractions. Clin Appl Thromb Hemost 2012; 19:48-54. [DOI: 10.1177/1076029611436194] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Shaker A. Mousa
- The Pharmaceutical Research Institute at Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
199
|
Shroff K, Pearce TR, Kokkoli E. Enhanced integrin mediated signaling and cell cycle progression on fibronectin mimetic peptide amphiphile monolayers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2012; 28:1858-1865. [PMID: 22149259 DOI: 10.1021/la203322t] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
In recent years, a variety of biomimetic constructs have emerged which mimic the bioactive sequences found in the natural extracellular matrix (ECM) proteins such as fibronectin (FN) that promote cell adhesion as well as proliferation on artificially functionalized interfaces. Much interest lies in investigating the ability of the ECM mimetic materials in regulating a number of vital cell functions including differentiation, gene expression, migration, and proliferation. A peptide amphiphile PR_b containing both the cell adhesive GRGDSP and synergistic PHSRN peptide sequences was developed in our group that was shown to support enhanced cell proliferation and ECM FN secretion as compared to GRGDSP and FN functionalized interfaces. In this study, we have investigated the binding affinity of the PR_b peptide ligand with the FN cell surface receptor, the α(5)β(1) integrin. We compared PR_b functionalized surfaces with FN and BSA coated surfaces and GRGDSP functionalized surfaces in terms of promoting intracellular signaling cascades that are essential for enhanced cellular activity. Specifically, we studied the phosphorylation of focal adhesion kinase (FAK) at tyrosine residues Y397 and Y576 and the formation of cyclin D1, both of which are intracellular markers of integrin mediated attachment of cells, signaling pathways, and progression of cell cycle. FAK and cyclin D1 encourage enhanced cell proliferation, differentiation, and gene expression. Our results show that the PR_b peptide ligand has a specific and strong binding affinity for the α(5)β(1) integrin with a dissociation constant of 76.3 ± 6.3 nM. The PR_b peptide ligands supported enhanced FAK phosphorylation activity and increased cyclin D1 formation as compared to the widely used GRGDSP ligand, the native protein FN (positive control), and BSA nonadhesive surfaces (negative control). These results encourage the use of the FN mimetic PR_b peptide in functionalizing biomaterials for potential tissue engineering and therapeutic applications.
Collapse
Affiliation(s)
- Kamlesh Shroff
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | | | | |
Collapse
|
200
|
Angiogenic response of endothelial cells to fibronectin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 749:131-51. [PMID: 22695843 DOI: 10.1007/978-1-4614-3381-1_10] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|