101
|
Xu JG, Zhu SY, Heng BC, Dissanayaka WL, Zhang CF. TGF-β1-induced differentiation of SHED into functional smooth muscle cells. Stem Cell Res Ther 2017; 8:10. [PMID: 28114966 PMCID: PMC5260045 DOI: 10.1186/s13287-016-0459-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 12/02/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Adequate vascularization is crucial for supplying nutrition and discharging metabolic waste in freshly transplanted tissue-engineered constructs. Obtaining the appropriate building blocks for vascular tissue engineering (i.e. endothelial and mural cells) is a challenging task for tissue neovascularization. Hence, we investigated whether stem cells from human exfoliated deciduous teeth (SHED) could be induced to differentiate into functional vascular smooth muscle cells (vSMCs). METHODS We utilized two cytokines of the TGF-β family, transforming growth factor beta 1 (TGF-β1) and bone morphogenetic protein 4 (BMP4), to induce SHED differentiation into SMCs. Quantitative real-time polymerase chain reaction (RT-qPCR) was used to assess mRNA expression, and protein expression was analyzed using flow cytometry, western blot and immunostaining. Additionally, to examine whether these SHED-derived SMCs possess the same function as primary SMCs, in vitro Matrigel angiogenesis assay, fibrin gel bead assay, and functional contraction study were used here. RESULTS By analyzing the expression of specific markers of SMCs (α-SMA, SM22α, Calponin, and SM-MHC), we confirmed that TGF-β1, and not BMP4, could induce SHED differentiation into SMCs. The differentiation efficiency was relatively high (α-SMA+ 86.1%, SM22α+ 93.9%, Calponin+ 56.8%, and SM-MHC+ 88.2%) as assessed by flow cytometry. In vitro Matrigel angiogenesis assay showed that the vascular structures generated by SHED-derived SMCs and human umbilical vein endothelial cells (HUVECs) were comparable to primary SMCs and HUVECs in terms of vessel stability. Fibrin gel bead assay showed that SHED-derived SMCs had a stronger capacity for promoting vessel formation compared with primary SMCs. Further analyses of protein expression in fibrin gel showed that cultures containing SHED-derived SMCs exhibited higher expression levels of Fibronectin than the primary SMCs group. Additionally, it was also confirmed that SHED-derived SMCs exhibited functional contractility. When SB-431542, a specific inhibitor of ALK5 was administered, TGF-β1 stimulation could not induce SHED into SMCs, indicating that the differentiation of SHED into SMCs is somehow related to the TGF-β1-ALK5 signaling pathway. CONCLUSIONS SHED could be successfully induced into functional SMCs for vascular tissue engineering, and this course could be regulated through the ALK5 signaling pathway. Hence, SHED appear to be a promising candidate cell type for vascular tissue engineering.
Collapse
Affiliation(s)
- Jian Guang Xu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Shao Yue Zhu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Boon Chin Heng
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
| | - Waruna Lakmal Dissanayaka
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
- HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| | - Cheng Fei Zhang
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong China
- HKU Shenzhen Institute of Research and Innovation, Hong Kong, China
| |
Collapse
|
102
|
Kim HK, Choi JS, Lee SW, Joo CK, Joe YA. A Novel Peptide Derived From Tissue-Type Plasminogen Activator Potently Inhibits Angiogenesis and Corneal Neovascularization. J Cell Biochem 2017; 118:1132-1143. [DOI: 10.1002/jcb.25732] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Accepted: 09/09/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Hyun-Kyung Kim
- Cancer Research Institute; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Department of Medical Lifescience; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Cancer Evolution Research Center; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
| | - Jun-Sub Choi
- Department of Ophthalmology and Visual Science; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
| | - Seung Woo Lee
- Cancer Research Institute; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Department of Medical Lifescience; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Cancer Evolution Research Center; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
| | - Choun-Ki Joo
- Department of Ophthalmology and Visual Science; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
| | - Young Ae Joe
- Cancer Research Institute; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Department of Medical Lifescience; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
- Cancer Evolution Research Center; College of Medicine; The Catholic University of Korea; Seoul Republic of Korea
| |
Collapse
|
103
|
Zhang N, Xia Y, Zou Y, Yang W, Zhang J, Zhong Z, Meng F. ATN-161 Peptide Functionalized Reversibly Cross-Linked Polymersomes Mediate Targeted Doxorubicin Delivery into Melanoma-Bearing C57BL/6 Mice. Mol Pharm 2017; 14:2538-2547. [PMID: 28005375 DOI: 10.1021/acs.molpharmaceut.6b00800] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PHSCN peptide (licensed as ATN-161) is an effective α5β1 integrin inhibitor that has advanced to phase II clinical trials to treat solid tumors. Here we developed ATN-161 functionalized self-cross-linkable and intracellularly de-cross-linkable polymersomes (ATN/SCID-Ps) for highly efficient and targeted delivery of doxorubicin hydrochloride (DOX·HCl) into B16F10 melanoma-bearing C57BL/6 mice. ATN/SCID-Ps exhibited a high loading capacity of DOX·HCl. The size of DOX-loaded ATN/SCID-Ps (DOX-ATN/SCID-Ps) decreased from 150 to 88 nm with increasing ATN surface densities from 0 to 100% (mol/mol). DOX-ATN/SCID-Ps were robust with low drug leakage under physiological conditions while quickly releasing DOX with the addition of 10 mM glutathione. MTT assay results displayed that DOX-ATN/SCID-Ps induced ATN density-dependent antitumor activity to α5β1 integrin overexpressing B16F10 melanoma cells, in which 56% ATN-161 was optimal. Flow cytometry and CLSM studies revealed significantly more efficient internalization and cytoplasmic DOX release in B16F10 cells for DOX-ATN/SCID-Ps than for DOX-SCID-Ps (nontargeting control) as well as clinically used pegylated liposomal doxorubicin (DOX-LPs). DOX-ATN/SCID-Ps displayed a long blood circulation time (elimination half-life = 4.13 h) and 4 times higher DOX accumulation in B16F10 bearing C57BL/6 mice than DOX-LPs. Interestingly, DOX-ATN/SCID-Ps exhibited a superior maximum-tolerated dose of over 100 mg DOX·HCl/kg, 10 times higher than DOX-LPs. Remarkably, DOX-ATN/SCID-Ps could significantly inhibit the growth of aggressive B16F10 melanoma with little adverse effects via either multiple or single injection of total dosage of 100 mg DOX·HCl/kg, resulting in greatly improved survival rates as compared to DOX-LPs. ATN/SCID-Ps are appealing nanovehicles for targeted chemotherapy of α5β1 integrin positive solid tumors.
Collapse
Affiliation(s)
- Ning Zhang
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Yifeng Xia
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Yan Zou
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Weijing Yang
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Jian Zhang
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University , Suzhou, 215123, P. R. China
| |
Collapse
|
104
|
Montassar F, Darche M, Blaizot A, Augustin S, Conart JB, Millet A, Elayeb M, Sahel JA, Réaux-Le Goazigo A, Sennlaub F, Marrakchi N, Messadi E, Guillonneau X. Lebecetin, a C-type lectin, inhibits choroidal and retinal neovascularization. FASEB J 2016; 31:1107-1119. [PMID: 27974593 DOI: 10.1096/fj.201600351r] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 11/28/2016] [Indexed: 12/14/2022]
Abstract
Angiogenesis is a cause of visual impairment and blindness in the wet form of age-related macular degeneration and in ischemic retinopathies. Current therapies include use of anti-VEGF agents to reduce choroidal neovascularization (CNV) and edema. These treatments are effective in most cases, but spontaneous or acquired resistance to anti-VEGF and possible adverse effects of long-term VEGF inhibition in the retina and choroid highlight a need for additional alternative therapies. Integrins αvβ3 and αvβ5, which regulate endothelial cell proliferation and stabilization, have been implicated in ocular angiogenesis. Lebecetin (LCT) is a 30-kDa heterodimeric C-type lectin that is isolated from Macrovipera lebetina venom and interacts with α5β1- and αv-containing integrins. We previously showed that LCT inhibits human brain microvascular endothelial cell adhesion, migration, proliferation, and tubulogenesis. To evaluate the inhibitory effect of LCT on ocular angiogenesis, we cultured aortic and choroidal explants in the presence of LCT and analyzed the effect of LCT on CNV in the mouse CNV model and on retinal neovascularization in the oxygen-induced retinopathy model. Our data demonstrate that a single injection of LCT efficiently reduced CNV and retinal neovascularization in these models.-Montassar, F., Darche, M., Blaizot, A., Augustin, S., Conart, J.-B., Millet, A., Elayeb, M., Sahel, J.-A., Réaux-Le Goazigo, A., Sennlaub, F., Marrakchi, N., Messadi, E., Guillonneau, X. Lebecetin, a C-type lectin, inhibits choroidal and retinal neovascularization.
Collapse
Affiliation(s)
- Fadoua Montassar
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, France.,Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Institut Pasteur de Tunis, Tunis, Tunisia.,Université de Carthage, Faculté des Sciences de Bizerte, Bizerte, Tunisia
| | - Marie Darche
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, France
| | - Amandine Blaizot
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, France
| | - Sébastien Augustin
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, France
| | - Jean-Baptiste Conart
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, France
| | - Aurélie Millet
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Mohamed Elayeb
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Institut Pasteur de Tunis, Tunis, Tunisia
| | - José-Alain Sahel
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, France
| | - Annabelle Réaux-Le Goazigo
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, France
| | - Florian Sennlaub
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, France
| | - Naziha Marrakchi
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Erij Messadi
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Xavier Guillonneau
- Sorbonne Universités, Université Pierre et Marie Curie, INSERM, Centre National de la Recherche Scientifique, Institut de la Vision, Paris, France;
| |
Collapse
|
105
|
Bianconi D, Unseld M, Prager GW. Integrins in the Spotlight of Cancer. Int J Mol Sci 2016; 17:ijms17122037. [PMID: 27929432 PMCID: PMC5187837 DOI: 10.3390/ijms17122037] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 11/17/2016] [Accepted: 11/28/2016] [Indexed: 02/07/2023] Open
Abstract
Integrins are heterodimeric cell surface receptors that bind to different extracellular ligands depending on their composition and regulate all processes which enable multicellular life. In cancer, integrins trigger and play key roles in all the features that were once described as the Hallmarks of Cancer. In this review, we will discuss the contribution of integrins to these hallmarks, including uncontrolled and limitless proliferation, invasion of tumor cells, promotion of tumor angiogenesis and evasion of apoptosis and resistance to growth suppressors, by highlighting the latest findings. Further on, given the paramount role of integrins in cancer, we will present novel strategies for integrin inhibition that are starting to emerge, promising a hopeful future regarding cancer treatment.
Collapse
Affiliation(s)
- Daniela Bianconi
- Department of Internal Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Matthias Unseld
- Department of Internal Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, A-1090 Vienna, Austria.
| | - Gerald W Prager
- Department of Internal Medicine I, Comprehensive Cancer Center Vienna, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
106
|
Bianchini F, Peppicelli S, Fabbrizzi P, Biagioni A, Mazzanti B, Menchi G, Calorini L, Pupi A, Trabocchi A. Triazole RGD antagonist reverts TGFβ1-induced endothelial-to-mesenchymal transition in endothelial precursor cells. Mol Cell Biochem 2016; 424:99-110. [PMID: 27761847 PMCID: PMC5219041 DOI: 10.1007/s11010-016-2847-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/08/2016] [Indexed: 12/26/2022]
Abstract
Fibrosis is the dramatic consequence of a dysregulated reparative process in which activated fibroblasts (myofibroblasts) and Transforming Growth Factor β1 (TGFβ1) play a central role. When exposed to TGFβ1, fibroblast and epithelial cells differentiate in myofibroblasts; in addition, endothelial cells may undergo endothelial-to-mesenchymal transition (EndoMT) and actively participate to the progression of fibrosis. Recently, the role of αv integrins, which recognize the Arg-Gly-Asp (RGD) tripeptide, in the release and signal transduction activation of TGFβ1 became evident. In this study, we present a class of triazole-derived RGD antagonists that interact with αvβ3 integrin. Above different compounds, the RGD-2 specifically interferes with integrin-dependent TGFβ1 EndoMT in Endothelial Colony-Forming Cells (ECPCs) derived from circulating Endothelial Precursor Cells (ECPCs). The RGD-2 decreases the amount of membrane-associated TGFβ1, and reduces both ALK5/TGFβ1 type I receptor expression and Smad2 phosphorylation in ECPCs. We found that RGD-2 antagonist reverts EndoMT, reducing α-smooth muscle actin (α-SMA) and vimentin expression in differentiated ECPCs. Our results outline the critical role of integrin in fibrosis progression and account for the opportunity of using integrins as target for anti-fibrotic therapeutic treatment.
Collapse
Affiliation(s)
- Francesca Bianchini
- Department of Clinical and Experimental Biomedical Science "Mario Serio", University of Florence, Florence, Italy.
| | - Silvia Peppicelli
- Department of Clinical and Experimental Biomedical Science "Mario Serio", University of Florence, Florence, Italy
| | | | - Alessio Biagioni
- Department of Clinical and Experimental Biomedical Science "Mario Serio", University of Florence, Florence, Italy
| | | | - Gloria Menchi
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy.,Interdepartmental Center for Preclinical Development of Molecular Imaging (CISPIM), University of Florence, Florence, Italy
| | - Lido Calorini
- Department of Clinical and Experimental Biomedical Science "Mario Serio", University of Florence, Florence, Italy
| | - Alberto Pupi
- Department of Clinical and Experimental Biomedical Science "Mario Serio", University of Florence, Florence, Italy.,Interdepartmental Center for Preclinical Development of Molecular Imaging (CISPIM), University of Florence, Florence, Italy
| | - Andrea Trabocchi
- Department of Chemistry "Ugo Schiff", University of Florence, Florence, Italy.,Interdepartmental Center for Preclinical Development of Molecular Imaging (CISPIM), University of Florence, Florence, Italy
| |
Collapse
|
107
|
Mandic L, Traxler D, Gugerell A, Zlabinger K, Lukovic D, Pavo N, Goliasch G, Spannbauer A, Winkler J, Gyöngyösi M. Molecular Imaging of Angiogenesis in Cardiac Regeneration. CURRENT CARDIOVASCULAR IMAGING REPORTS 2016; 9:27. [PMID: 27683600 PMCID: PMC5018257 DOI: 10.1007/s12410-016-9389-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Myocardial infarction (MI) leading to heart failure displays an important cause of death worldwide. Adequate restoration of blood flow to prevent this transition is a crucial factor to improve long-term morbidity and mortality. Novel regenerative therapies have been thoroughly investigated within the past decades. RECENT FINDINGS Increased angiogenesis in infarcted myocardium has shown beneficial effects on the prognosis of MI; therefore, the proangiogenic capacity of currently tested treatments is of specific interest. Molecular imaging to visualize formation of new blood vessels in vivo displays a promising option to monitor proangiogenic effects of regenerative substances. SUMMARY Based on encouraging results in preclinical models, molecular angiogenesis imaging has recently been applied in a small set of patients. This article reviews recent literature on noninvasive in vivo molecular imaging of angiogenesis after MI as an integral part of cardiac regeneration.
Collapse
Affiliation(s)
- Ljubica Mandic
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Denise Traxler
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alfred Gugerell
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Katrin Zlabinger
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Dominika Lukovic
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Noemi Pavo
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Georg Goliasch
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Andreas Spannbauer
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Johannes Winkler
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
108
|
Interaction between integrin α5 and PDE4D regulates endothelial inflammatory signalling. Nat Cell Biol 2016; 18:1043-53. [PMID: 27595237 PMCID: PMC5301150 DOI: 10.1038/ncb3405] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 08/03/2016] [Indexed: 12/16/2022]
Abstract
Atherosclerosis is primarily a disease of lipid metabolism and inflammation; however, it is also closely associated with endothelial extracellular matrix (ECM) remodelling, with fibronectin accumulating in the laminin-collagen basement membrane. To investigate how fibronectin modulates inflammation in arteries, we replaced the cytoplasmic tail of the fibronectin receptor integrin α5 with that of the collagen/laminin receptor integrin α2. This chimaera suppressed inflammatory signalling in endothelial cells on fibronectin and in knock-in mice. Fibronectin promoted inflammation by suppressing anti-inflammatory cAMP. cAMP was activated through endothelial prostacyclin secretion; however, this was ECM-independent. Instead, cells on fibronectin suppressed cAMP via enhanced phosphodiesterase (PDE) activity, through direct binding of integrin α5 to phosphodiesterase-4D5 (PDE4D5), which induced PP2A-dependent dephosphorylation of PDE4D5 on the inhibitory site Ser651. In vivo knockdown of PDE4D5 inhibited inflammation at athero-prone sites. These data elucidate a molecular mechanism linking ECM remodelling and inflammation, thereby identifying a new class of therapeutic targets.
Collapse
|
109
|
Hoja-Łukowicz D, Przybyło M, Duda M, Pocheć E, Bubka M. On the trail of the glycan codes stored in cancer-related cell adhesion proteins. Biochim Biophys Acta Gen Subj 2016; 1861:3237-3257. [PMID: 27565356 DOI: 10.1016/j.bbagen.2016.08.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 07/22/2016] [Accepted: 08/14/2016] [Indexed: 12/14/2022]
Abstract
Changes in the profile of protein glycosylation are a hallmark of ongoing neoplastic transformation. A unique set of tumor-associated carbohydrate antigens expressed on the surface of malignant cells may serve as powerful diagnostic and therapeutic targets. Cell-surface proteins with altered glycosylation affect the growth, proliferation and survival of those cells, and contribute to their acquisition of the ability to migrate and invade. They may also facilitate tumor-induced immunosuppression and the formation of distant metastases. Deciphering the information encoded in these particular glycan portions of glycoconjugates may shed light on the mechanisms of cancer progression and metastasis. A majority of the related review papers have focused on overall changes in the patterns of cell-surface glycans in various cancers, without pinpointing the molecular carriers of these glycan structures. The present review highlights the ways in which particular tumor-associated glycan(s) coupled with a given membrane-bound protein influence neoplastic cell behavior during the development and progression of cancer. We focus on altered glycosylated cell-adhesion molecules belonging to the cadherin, integrin and immunoglobulin-like superfamilies, examined in the context of molecular interactions.
Collapse
Affiliation(s)
- Dorota Hoja-Łukowicz
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Małgorzata Duda
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Ewa Pocheć
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Monika Bubka
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| |
Collapse
|
110
|
Liu H, Gong X, Jing X, Ding X, Yao Y, Huang Y, Fan Y. Shear stress with appropriate time-step and amplification enhances endothelial cell retention on vascular grafts. J Tissue Eng Regen Med 2016; 11:2965-2978. [DOI: 10.1002/term.2196] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/09/2016] [Accepted: 03/14/2016] [Indexed: 01/08/2023]
Affiliation(s)
- Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering; Beihang University; Beijing People's Republic of China
| | - Xianghui Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering; Beihang University; Beijing People's Republic of China
| | - Xiaohui Jing
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering; Beihang University; Beijing People's Republic of China
| | - Xili Ding
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering; Beihang University; Beijing People's Republic of China
| | - Yuan Yao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering; Beihang University; Beijing People's Republic of China
| | - Yan Huang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering; Beihang University; Beijing People's Republic of China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering; Beihang University; Beijing People's Republic of China
- National Research Centre for Rehabilitation Technical Aids; Beijing People's Republic of China
| |
Collapse
|
111
|
Samardzija C, Luwor RB, Quinn MA, Kannourakis G, Findlay JK, Ahmed N. Coalition of Oct4A and β1 integrins in facilitating metastasis in ovarian cancer. BMC Cancer 2016; 16:432. [PMID: 27390927 PMCID: PMC4939035 DOI: 10.1186/s12885-016-2458-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 06/24/2016] [Indexed: 12/16/2022] Open
Abstract
Background Ovarian cancer is a metastatic disease and one of the leading causes of gynaecology malignancy-related deaths in women. Cancer stem cells (CSCs) are key contributors of cancer metastasis and relapse. Integrins are a family of cell surface receptors which allow interactions between cells and their surrounding microenvironment and play a fundamental role in promoting metastasis. This study investigates the molecular mechanism which associates CSCs and integrins in ovarian cancer metastasis. Methods The expression of Oct4A in high-grade serous ovarian tumors and normal ovaries was determined by immunofluorescence analysis. The functional role of Oct4A was evaluated by generating stable knockdown (KD) of Oct4A clones in an established ovarian cancer cell line HEY using shRNA-mediated silencing. The expression of integrins in cell lines was evaluated by flow cytometry. Spheroid forming ability, adhesion and the activities of matrix metalloproteinases 9/2 (MMP-9/2) was measured by in vitro functional assays and gelatin zymography. These observations were further validated in in vivo mouse models using Balb/c nu/nu mice. Results We report significantly elevated expression of Oct4A in high-grade serous ovarian tumors compared to normal ovarian tissues. The expression of Oct4A in ovarian cancer cell lines correlated with their CSC-related sphere forming abilities. The suppression of Oct4A in HEY cells resulted in a significant diminution of integrin β1 expression and associated α5 and α2 subunits compared to vector control cells. This was associated with a reduced adhesive ability on collagen and fibronectin and decreased secretion of pro-MMP2 in Oct4A KD cells compared to vector control cells. In vivo, Oct4A knock down (KD) cells produced tumors which were significantly smaller in size and weight compared to tumors derived from vector control cells. Immunohistochemical analyses of Oct4A KD tumor xenografts demonstrated a significant loss of cytokeratin 7 (CK7), Glut-1 as well as CD34 and CD31 compared to vector control cell-derived xenografts. Conclusion The expression of Oct4A may be crucial to promote and sustain integrin-mediated extracellular matrix (ECM) remodeling requisite for tumor metastasis in ovarian cancer patients.
Collapse
Affiliation(s)
- Chantel Samardzija
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Melbourne, VIC, 3052, Australia
| | - Rodney B Luwor
- Department of Surgery, University of Melbourne, Parkville, Melbourne, VIC, 3052, Australia
| | - Michael A Quinn
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Melbourne, VIC, 3052, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Suites 23-26, 106-110 Lydiard Street South, Ballarat Technology Central Park, Ballarat, 3353, Australia.,Federation University Australia, Ballarat, VIC, 3010, Australia
| | - Jock K Findlay
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Melbourne, VIC, 3052, Australia.,The Hudson Institute of Medical Research, Clayton, Melbourne, VIC, 3168, Australia
| | - Nuzhat Ahmed
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Melbourne, VIC, 3052, Australia. .,Fiona Elsey Cancer Research Institute, Suites 23-26, 106-110 Lydiard Street South, Ballarat Technology Central Park, Ballarat, 3353, Australia. .,Federation University Australia, Ballarat, VIC, 3010, Australia. .,The Hudson Institute of Medical Research, Clayton, Melbourne, VIC, 3168, Australia.
| |
Collapse
|
112
|
Lee KJ, Lim D, Yoo YH, Park EJ, Lee SH, Yadav BK, Lee YK, Park JH, Kim D, Park KH, Hahn JH. Paired Ig-Like Type 2 Receptor-Derived Agonist Ligands Ameliorate Inflammatory Reactions by Downregulating β1 Integrin Activity. Mol Cells 2016; 39:557-65. [PMID: 27306643 PMCID: PMC4959021 DOI: 10.14348/molcells.2016.0079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/16/2016] [Accepted: 05/19/2016] [Indexed: 11/27/2022] Open
Abstract
The paired immunoglobulin-like type 2 receptor (PILR) family consists of two functionally opposite members, inhibitory PILRα and activating PILRβ receptors. PILRs are widely expressed in various immune cells and interact with their ligands, especially CD99 expressed on activated T cells, to participate in immune responses. Here we investigated whether PILR-derived agonists inhibit β1 integrin activity as ligands for CD99. PILR-derived peptides as well as PILR-Fc fusion proteins prevented cell adhesion to fibronectin through the regulation of β1 integrin activity. Especially, PILRpep3, a representative 3-mer peptide covering the conserved motifs of the PILR extracellular domain, prevented the clustering and activation of β1 integrin by dephosphorylating FAK and vinculin, which are major components of focal adhesion. In addition, PILRpep3 inhibited transendothelial migration of monocytes as well as endothelial cell tube formation. Furthermore, upon intraperitoneal injection of PILRpep3 into mice with collagen-induced arthritis, the inflammatory response of rheumatoid arthritis was strongly suppressed. Taken together, these results suggest that PILR-derived agonist ligands may prevent the inflammatory reactions of rheumatoid arthritis by activating CD99.
Collapse
Affiliation(s)
- Kyoung-Jin Lee
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| | - Dongyoung Lim
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| | - Yeon Ho Yoo
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| | - Eun-Ji Park
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| | - Sun-Hee Lee
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| | - Birendra Kumar Yadav
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| | - Yong-Ki Lee
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| | - Jeong Hyun Park
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| | - Daejoong Kim
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| | - Kyeong Han Park
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| | - Jang-Hee Hahn
- Department of Anatomy and Cell Biology, School of Medicine, Kangwon National University, Chuncheon 200-701,
Korea
| |
Collapse
|
113
|
Hall AP, Westwood FR, Wadsworth PF. Review of the Effects of Anti-Angiogenic Compounds on the Epiphyseal Growth Plate. Toxicol Pathol 2016; 34:131-47. [PMID: 16537292 DOI: 10.1080/01926230600611836] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The formation of new blood vessels from a pre-existing vascular bed, termed “angiogenesis,” is of critical importance for the growth and development of the animal since it is required for the growth of the skeleton during endochondral ossification, development and cycling of the corpus luteum and uterus, and for the repair of tissues during wound healing. “Vasculogenesis,” the de novo formation of blood vessels is also important for the proper function and development of the vascular system in the embryo. New blood vessel formation is a prominent feature and permissive factor in the relentless progression of many human diseases, one of the most important examples of which is neoplasia. It is for this reason that angiogenesis is considered to be one of the hallmarks of cancer. The development of new classes of drugs that inhibit the growth and proper functioning of new blood vessels in vivo is likely to provide significant therapeutic benefit in the treatment of cancer, as well as other conditions where angiogenesis is a strong driver to the disease process. During the preclinical safety testing of these drugs, it is becoming increasingly clear that their in vivo efficacy is reflected in the profile of “expected toxicity” (resulting from pharmacology) observed in laboratory animals, so much so, that this profile of “desired” toxicity may act as a signature for their anti-angiogenic effect. In this article we review the major mechanisms controlling angiogenesis and its role during endochondral ossification. We also review the effects of perturbation of endochondral ossification through four mechanisms—inhibition of vascular endothelial growth factor (VEGF), pp60 c-Src kinase and matrix metalloproteinases as well as disruption of the blood supply with vascular targeting agents. Inhibition through each of these mechanisms appears to have broadly similar effects on the epiphyseal growth plate characterised by thickening due to the retention of hypertrophic chondrocytes resulting from the inhibition of angiogenesis. In contrast, in the metaphysis there are differing effects reflecting the specific role of these targets at this site.
Collapse
Affiliation(s)
- Anthony P Hall
- AstraZeneca, Mereside, Alderley Park, Macclesfield, Cheshire SK10 4TG, England.
| | | | | |
Collapse
|
114
|
Mas-Moruno C, Fraioli R, Rechenmacher F, Neubauer S, Kapp TG, Kessler H. αvβ3- or α5β1-Integrin-Selective Peptidomimetics for Surface Coating. Angew Chem Int Ed Engl 2016; 55:7048-67. [PMID: 27258759 DOI: 10.1002/anie.201509782] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Indexed: 12/21/2022]
Abstract
Engineering biomaterials with integrin-binding activity is a very powerful approach to promote cell adhesion, modulate cell behavior, and induce specific biological responses at the surface level. The aim of this Review is to illustrate the evolution of surface-coating molecules in this field: from peptides and proteins with relatively low integrin-binding activity and receptor selectivity to highly active and selective peptidomimetic ligands. In particular, we will bring into focus the difficult challenge of achieving selectivity between the two closely related integrin subtypes αvβ3 and α5β1. The functionalization of surfaces with such peptidomimetics opens the way for a new generation of highly specific cell-instructive surfaces to dissect the biological role of integrin subtypes and for application in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering, Universitat Politècnica de Catalunya (UPC), Diagonal 647, 08028, Barcelona, Spain.
| | - Roberta Fraioli
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering, Universitat Politècnica de Catalunya (UPC), Diagonal 647, 08028, Barcelona, Spain
| | - Florian Rechenmacher
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Stefanie Neubauer
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Tobias G Kapp
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Horst Kessler
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany.
| |
Collapse
|
115
|
Mas-Moruno C, Fraioli R, Rechenmacher F, Neubauer S, Kapp TG, Kessler H. αvβ3- oder α5β1-Integrin-selektive Peptidmimetika für die Oberflächenbeschichtung. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201509782] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering; Universitat Politècnica de Catalunya (UPC); Diagonal 647 08028 Barcelona Spanien
| | - Roberta Fraioli
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering; Universitat Politècnica de Catalunya (UPC); Diagonal 647 08028 Barcelona Spanien
| | - Florian Rechenmacher
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Stefanie Neubauer
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Tobias G. Kapp
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Horst Kessler
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| |
Collapse
|
116
|
Wang WQ, Wang FH, Qin WX, Liu HY, Lu B, Chung C, Zhu J, Gu Q, Shi W, Wen C, Wu F, Zhang K, Sun XD. Joint Antiangiogenic Effect of ATN-161 and Anti-VEGF Antibody in a Rat Model of Early Wet Age-Related Macular Degeneration. Mol Pharm 2016; 13:2881-90. [PMID: 27089240 DOI: 10.1021/acs.molpharmaceut.6b00056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The wet form of age-related macular degeneration (AMD) is a leading cause of blindness among elderly Americans and is characterized by abnormal vessel growth, termed choroidal neovascularization (CNV). Integrin α5β1 is a transmembrane receptor that binds matrix macromolecules and proteinases to stimulate angiogenesis. We recently demonstrated that integrin α5β1 plays a critical role in the development of choroidal neovascularization. In this study, we determined the role and underlying mechanisms of integrin α5β1 in angiogenesis in human choroidal endothelial cells and evaluated the antiangiogenic effects of delivering a combination therapy of ATN-161, an integrin α5β1 inhibitor, and an anti-VEGF monoclonal antibody to rats with laser-induced CNV. Vascular endothelial growth factor (VEGF) is a signaling protein that stimulates vasculogenesis and angiogenesis through a pathway that is distinct from the integrin α5β1 signaling pathway. Our results indicate that fibronectin binds to integrin α5β1 and synergizes VEGF-induced angiogenesis via two independent signaling pathways, FN/integrin α5β1/FAK/ERK1/2 and FN/integrin α5β1/FAK/AKT. Integrin α5 knockdown by shRNA inhibits endothelial cell migration, tube formation, and proliferation, while ATN-161 only partially decreases integrin α5 function. Treatment with ATN-161 combined with anti-VEGF antibody showed joint effects in attenuating angiogenesis. In summary, our results provide the first evidence for the mechanisms by which integrin α5β1 is involved in ocular pathological neovascularization in vivo, suggesting that dual inhibition of integrin α5β1 and VEGF may be a promising novel therapeutic strategy for CNV in wet AMD.
Collapse
Affiliation(s)
- Wen-Qiu Wang
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China.,Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Feng-Hua Wang
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China
| | - Wen-Xin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, 200032, China
| | - Hai-Yun Liu
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China
| | - Bing Lu
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China
| | - Christopher Chung
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Jie Zhu
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Qing Gu
- Shanghai Key Laboratory of Fundus Disease and Eye Research Institute, Shanghai JiaoTong University , Shanghai 200080, China
| | - William Shi
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States.,Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Sichuan 610041, China
| | - Cindy Wen
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States.,Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Sichuan 610041, China
| | - Frances Wu
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Kang Zhang
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States.,Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Sichuan 610041, China.,Veterans Administration Healthcare System , San Diego, California 92161, United States
| | - Xiao-Dong Sun
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China.,Shanghai Key Laboratory of Fundus Disease and Eye Research Institute, Shanghai JiaoTong University , Shanghai 200080, China
| |
Collapse
|
117
|
Fortunato TM, Beltrami C, Emanueli C, De Bank PA, Pula G. Platelet lysate gel and endothelial progenitors stimulate microvascular network formation in vitro: tissue engineering implications. Sci Rep 2016; 6:25326. [PMID: 27141997 PMCID: PMC4855184 DOI: 10.1038/srep25326] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 04/14/2016] [Indexed: 12/22/2022] Open
Abstract
Revascularisation is a key step for tissue regeneration and complete organ engineering. We describe the generation of human platelet lysate gel (hPLG), an extracellular matrix preparation from human platelets able to support the proliferation of endothelial colony forming cells (ECFCs) in 2D cultures and the formation of a complete microvascular network in vitro in 3D cultures. Existing extracellular matrix preparations require addition of high concentrations of recombinant growth factors and allow only limited formation of capillary-like structures. Additional advantages of our approach over existing extracellular matrices are the absence of any animal product in the composition hPLG and the possibility of obtaining hPLG from patients to generate homologous scaffolds for re-implantation. This discovery has the potential to accelerate the development of regenerative medicine applications based on implantation of microvascular networks expanded ex vivo or the generation of fully vascularised organs.
Collapse
Affiliation(s)
- Tiago M Fortunato
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Cristina Beltrami
- Bristol Heart Institute, School of Clinical Sciences University of Bristol, Bristol, UK
| | - Costanza Emanueli
- Bristol Heart Institute, School of Clinical Sciences University of Bristol, Bristol, UK
| | - Paul A De Bank
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| | - Giordano Pula
- Department of Pharmacy and Pharmacology, University of Bath, Bath, UK
| |
Collapse
|
118
|
Almqvist S, Kleinman HK, Werthén M, Thomsen P, Agren MS. Effects of amelogenins on angiogenesis-associated processes of endothelial cells. J Wound Care 2016; 20:68, 70-5. [PMID: 21378680 DOI: 10.12968/jowc.2011.20.2.68] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
OBJECTIVE To study the effects of an amelogenin mixture on integrin-dependent adhesion, DNA synthesis and apoptosis of cultured human dermal microvascular endothelial cells and angiogenesis in an organotypic assay. METHOD Immobilised antibodies against specific integrins (alpha-1, alpha-2, alpha-3, alpha-4, alpha-5, alpha-v, ß1, ß2, ß3, ß4, ß6, alpha-vß3, alpha-vß5 and alpha-5ß1) were used to capture treated human dermal microvascular endothelial cells, which were detected colourimetrically. DNA synthesis of the cells was monitored by 5-bromo-2'- deoxyuridine incorporation and apoptosis by a TdT-mediated dUTP nick-end labelling technique. Tubule formation from aortic arches of 13-d-old chick embryos were followed over 48h. RESULTS The amelogenin mixture increased microvessel outgrowth by 76% (p < 0.01, n=12) from the aortic explants. Also, amelogenins increased the adhesion (p < 0.01, n = 5) by multiple angiogenesis associated integrin subunits and alpha-vß3, alpha-vß5 and alpha-5ß1 heterodimers on human dermal microvascular endothelial cells at a non-mitogenic concentration (100 µg/ml). Conversely, amelogenins at 1,000 µg/ml decreased microvessel formation possibly due to attenuation of corresponding integrins despite increasing (p < 0.001, n = 8) DNA synthesis. No significant apoptosis was detected in human dermal microvascular endothelial cells cultured on Matrigel with and without amelogenins. CONCLUSION Increased surface expression of integrins on endothelial cells may contribute to the proangiogenic property of amelogenins.
Collapse
Affiliation(s)
- S Almqvist
- Department of Biomaterials, Sahlgrenska Academy at the University of Gotheburg, Göteborg, Sweden
| | | | | | | | | |
Collapse
|
119
|
Anti-angiogenic mechanism of IPS-05002, a novel antagonist against integrin a5β1, determined by ProteoChip-based antibody array. BIOCHIP JOURNAL 2016. [DOI: 10.1007/s13206-016-0303-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
120
|
Zeng X, Ma YH, Chen YF, Qiu XC, Wu JL, Ling EA, Zeng YS. Autocrine fibronectin from differentiating mesenchymal stem cells induces the neurite elongation in vitro and promotes nerve fiber regeneration in transected spinal cord injury. J Biomed Mater Res A 2016; 104:1902-11. [PMID: 26991461 PMCID: PMC5101622 DOI: 10.1002/jbm.a.35720] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Revised: 03/02/2016] [Accepted: 03/11/2016] [Indexed: 01/08/2023]
Abstract
Extracellular matrix (ECM) expression is temporally and spatially regulated during the development of stem cells. We reported previously that fibronectin (FN) secreted by bone marrow mesenchymal stem cells (MSCs) was deposited on the surface of gelatin sponge (GS) soon after culture. In this study, we aimed to assess the function of accumulated FN on neuronal differentiating MSCs as induced by Schwann cells (SCs) in three dimensional transwell co‐culture system. The expression pattern and amount of FN of differentiating MSCs was examined by immunofluorescence, Western blot and immunoelectron microscopy. The results showed that FN accumulated inside GS scaffold, although its mRNA expression in MSCs was progressively decreased during neural induction. MSC‐derived neuron‐like cells showed spindle‐shaped cell body and long extending processes on FN‐decorated scaffold surface. However, after blocking of FN function by application of monoclonal antibodies, neuron‐like cells showed flattened cell body with short and thick neurites, together with decreased expression of integrin β1. In vivo transplantation study revealed that autocrine FN significantly facilitated endogenous nerve fiber regeneration in spinal cord transection model. Taken together, the present results showed that FN secreted by MSCs in the early stage accumulated on the GS scaffold and promoted the neurite elongation of neuronal differentiating MSCs as well as nerve fiber regeneration after spinal cord injury. This suggests that autocrine FN has a dynamic influence on MSCs in a three dimensional culture system and its potential application for treatment of traumatic spinal cord injury. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 104A: 1902–1911, 2016.
Collapse
Affiliation(s)
- Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
| | - Yuan-Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
- Department of Histology and Embryology, Guangdong Medical University, Zhanjiang, 524023, China
| | - Yuan-Feng Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xue-Cheng Qiu
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
| | - Jin-Lang Wu
- Department of Electron Microscope, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510120, China
- Co-innovation Center of Neuroregeneration, Jiangsu, 226019, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|
121
|
Currier NV, Ackerman SE, Kintzing JR, Chen R, Filsinger Interrante M, Steiner A, Sato AK, Cochran JR. Targeted Drug Delivery with an Integrin-Binding Knottin-Fc-MMAF Conjugate Produced by Cell-Free Protein Synthesis. Mol Cancer Ther 2016; 15:1291-300. [PMID: 27197305 DOI: 10.1158/1535-7163.mct-15-0881] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 03/17/2016] [Indexed: 11/16/2022]
Abstract
Antibody-drug conjugates (ADC) have generated significant interest as targeted therapeutics for cancer treatment, demonstrating improved clinical efficacy and safety compared with systemic chemotherapy. To extend this concept to other tumor-targeting proteins, we conjugated the tubulin inhibitor monomethyl-auristatin-F (MMAF) to 2.5F-Fc, a fusion protein composed of a human Fc domain and a cystine knot (knottin) miniprotein engineered to bind with high affinity to tumor-associated integrin receptors. The broad expression of integrins (including αvβ3, αvβ5, and α5β1) on tumor cells and their vasculature makes 2.5F-Fc an attractive tumor-targeting protein for drug delivery. We show that 2.5F-Fc can be expressed by cell-free protein synthesis, during which a non-natural amino acid was introduced into the Fc domain and subsequently used for site-specific conjugation of MMAF through a noncleavable linker. The resulting knottin-Fc-drug conjugate (KFDC), termed 2.5F-Fc-MMAF, had approximately 2 drugs attached per KFDC. 2.5F-Fc-MMAF inhibited proliferation in human glioblastoma (U87MG), ovarian (A2780), and breast (MB-468) cancer cells to a greater extent than 2.5F-Fc or MMAF alone or added in combination. As a single agent, 2.5F-Fc-MMAF was effective at inducing regression and prolonged survival in U87MG tumor xenograft models when administered at 10 mg/kg two times per week. In comparison, tumors treated with 2.5F-Fc or MMAF were nonresponsive, and treatment with a nontargeted control, CTRL-Fc-MMAF, showed a modest but not significant therapeutic effect. These studies provide proof-of-concept for further development of KFDCs as alternatives to ADCs for tumor targeting and drug delivery applications. Mol Cancer Ther; 15(6); 1291-300. ©2016 AACR.
Collapse
Affiliation(s)
- Nicolas V Currier
- Division of Pediatric Hematology/Oncology, Stanford Medical School, Stanford, California
| | | | - James R Kintzing
- Department of Bioengineering, Stanford University, Stanford, California
| | - Rishard Chen
- Sutro Biopharma, Inc., South San Francisco, California
| | | | | | - Aaron K Sato
- Sutro Biopharma, Inc., South San Francisco, California
| | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, California. Department of Chemical Engineering, Stanford University, Stanford, California.
| |
Collapse
|
122
|
Adhesion molecules and the extracellular matrix as drug targets for glioma. Brain Tumor Pathol 2016; 33:97-106. [PMID: 26992378 DOI: 10.1007/s10014-016-0261-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 03/07/2016] [Indexed: 12/14/2022]
Abstract
The formation of tumor vasculature and cell invasion along white matter tracts have pivotal roles in the development and progression of glioma. A better understanding of the mechanisms of angiogenesis and invasion in glioma will aid the development of novel therapeutic strategies. The processes of angiogenesis and invasion cause the production of an array of adhesion molecules and extracellular matrix (ECM) components. This review focuses on the role of adhesion molecules and the ECM in malignant glioma. The results of clinical trials using drugs targeted against adhesion molecules and the ECM for glioma are also discussed.
Collapse
|
123
|
Baeyens N, Bandyopadhyay C, Coon BG, Yun S, Schwartz MA. Endothelial fluid shear stress sensing in vascular health and disease. J Clin Invest 2016; 126:821-8. [PMID: 26928035 DOI: 10.1172/jci83083] [Citation(s) in RCA: 414] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Endothelial cells transduce the frictional force from blood flow (fluid shear stress) into biochemical signals that regulate gene expression and cell behavior via specialized mechanisms and pathways. These pathways shape the vascular system during development and during postnatal and adult life to optimize flow to tissues. The same pathways also contribute to atherosclerosis and vascular malformations. This Review covers recent advances in basic mechanisms of flow signaling and the involvement of these mechanisms in vascular physiology, remodeling, and these diseases. We propose that flow sensing pathways that govern normal morphogenesis can contribute to disease under pathological conditions or can be altered to induce disease. Viewing atherosclerosis and vascular malformations as instances of pathological morphogenesis provides a unifying perspective that may aid in developing new therapies.
Collapse
|
124
|
Topalovski M, Brekken RA. Matrix control of pancreatic cancer: New insights into fibronectin signaling. Cancer Lett 2015; 381:252-8. [PMID: 26742464 DOI: 10.1016/j.canlet.2015.12.027] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a highly metastatic disease that resists most current therapies. A defining characteristic of PDA is an intense fibrotic response that promotes tumor cell invasion and chemoresistance. Efforts to understand the complex relationship between the tumor and its extracellular network and to therapeutically perturb tumor-stroma interactions are ongoing. Fibronectin (FN), a provisional matrix protein abundant in PDA stroma but not normal tissues, supports metastatic spread and chemoresistance of this deadly disease. FN also supports angiogenesis, which is required for even hypovascular tumors such as PDA to develop and progress. Targeting components of the tumor stroma, such as FN, can effectively reduce tumor growth and spread while also enhancing delivery of chemotherapy. Here, we review the molecular mechanisms by which FN drives angiogenesis, metastasis and chemoresistance in PDA. In light of these new findings, we also discuss therapeutic strategies to inhibit FN signaling.
Collapse
Affiliation(s)
- Mary Topalovski
- Hamon Center for Therapeutic Oncology Research and the Division of Surgical Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research and the Division of Surgical Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Departments of Surgery and Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
125
|
Notni J, Steiger K, Hoffmann F, Reich D, Kapp TG, Rechenmacher F, Neubauer S, Kessler H, Wester HJ. Complementary, Selective PET Imaging of Integrin Subtypes α5β1 and αvβ3 Using 68Ga-Aquibeprin and 68Ga-Avebetrin. J Nucl Med 2015; 57:460-6. [DOI: 10.2967/jnumed.115.165720] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
126
|
D’Alessandria C, Pohle K, Rechenmacher F, Neubauer S, Notni J, Wester HJ, Schwaiger M, Kessler H, Beer AJ. In vivo biokinetic and metabolic characterization of the 68Ga-labelled α5β1-selective peptidomimetic FR366. Eur J Nucl Med Mol Imaging 2015; 43:953-963. [DOI: 10.1007/s00259-015-3218-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/06/2015] [Indexed: 01/04/2023]
|
127
|
A fibronectin mimetic motif improves integrin mediated cell biding to recombinant spider silk matrices. Biomaterials 2015; 74:256-66. [PMID: 26461118 DOI: 10.1016/j.biomaterials.2015.10.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 10/01/2015] [Accepted: 10/05/2015] [Indexed: 01/08/2023]
Abstract
The cell binding motif RGD is the most widely used peptide to improve cell binding properties of various biomaterials, including recombinant spider silk. In this paper we use genetic engineering to further enhance the cell supportive capacity of spider silk by presenting the RGD motif as a turn loop, similar to the one found in fibronectin (FN), but in the silk stabilized by cysteines, and therefore denoted FNCC. Human primary cells cultured on FNCC-silk showed increased attachment, spreading, stress fiber formation and focal adhesions, not only compared to RGD-silk, but also to silk fused with linear controls of the RGD containing motif from fibronectin. Cell binding to FNCC-silk was shown to involve the α5β1 integrin, and to support proliferation and migration of keratinocytes. The FNCC-silk protein allowed efficient assembly, and could even be transformed into free standing films, on which keratinocytes could readily form a monolayer culture. The results hold promise for future applications within tissue engineering.
Collapse
|
128
|
Ye P, Li Z, Jiang H, Liu T. SNPs in microRNA-binding sites in the ITGB1 and ITGB3 3'-UTR increase colorectal cancer risk. Cell Biochem Biophys 2015; 70:601-7. [PMID: 24777809 DOI: 10.1007/s12013-014-9962-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The purpose of the study was to investigate the potential associations between single-nucleotide polymorphisms (SNPs) in microRNA (miRNA)-binding sites in the integrin beta-1 (ITGB1) gene and integrin beta-3 (ITGB3) gene 3'-untranslated regions, and colorectal cancer (CRC) susceptibility in a Chinese population. A hospital-based case-control study was performed in 200 patients with CRC and 200 matched healthy donors. Two SNPs in miRNA binding of ITGB1 and ITGB3 genes (rs17468 and rs2317676) were genotyped by polymerase chain reaction-restrict fragment length polymorphism assay. The association between genotypes and CRC risk was evaluated by computing the odds ratio (OR) and 95 % confidence interval (CI) from multivariate unconditional logistic regression analyses. The frequency of the T genotype in ITGB1 rs17468 and G genotype in ITGB3 rs2317676 occurred more frequently in CRC patients than in controls (P < 0.05). We found that CT and TT genotypes of rs17468 were associated with a significantly increased risk of CRC (OR = 1.67, 95 % CI = 1.090-2.559 for CT + TT vs. CC), also the AG and GG genotype in ITGB3 rs2317676 (OR = 1.65, 95 % CI = 1.114-2.458 for AG + GG vs. AA). In conclusion, our results showed that both the ITGB1 rs17468 SNP and ITGB3 rs2317676 SNP were associated with an increased risk of CRC, which suggests that these 2 SNPs might contribute to CRC risk in a Chinese population.
Collapse
Affiliation(s)
- Pingjiang Ye
- Department of Colorectal Surgery, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing, 312000, Zhejiang, China
| | | | | | | |
Collapse
|
129
|
Wang K, Seo BR, Fischbach C, Gourdon D. Fibronectin Mechanobiology Regulates Tumorigenesis. Cell Mol Bioeng 2015; 9:1-11. [PMID: 26900407 PMCID: PMC4746220 DOI: 10.1007/s12195-015-0417-4] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 08/08/2015] [Indexed: 12/25/2022] Open
Abstract
Fibronectin (Fn) is an essential extracellular matrix (ECM) glycoprotein involved in both physiological and pathological processes. The structure–function relationship of Fn has been and is still being studied, as changes in its molecular structure are integral in regulating (or dysregulating) its biological activities via its cell, matrix component, and growth factor binding sites. Fn comprises three types of repeating modules; among them, FnIII modules are mechanically unstable domains that may be extended/unfolded upon cell traction and either uncover cryptic binding sites or disrupt otherwise exposed binding sites. Cells assemble Fn into a fibrillar network; its conformational flexibility implicates Fn as a critical mechanoregulator of the ECM. Fn has been shown to contribute to altered stroma remodeling during tumorigenesis. This review will discuss (i) the significance of the structure–function relationship of Fn at both the molecular and the matrix scales, (ii) the role of Fn mechanobiology in the regulation of tumorigenesis, and (iii) Fn-related advances in cancer therapy development.
Collapse
Affiliation(s)
- Karin Wang
- Department of Materials Science and Engineering, Cornell University, 327 Bard Hall, Ithaca, NY 14853 USA ; Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Bo Ri Seo
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853 USA
| | - Claudia Fischbach
- Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853 USA ; Kavli Institute at Cornell for Nanoscale Science, Cornell University, Ithaca, NY 14853 USA
| | - Delphine Gourdon
- Department of Materials Science and Engineering, Cornell University, 327 Bard Hall, Ithaca, NY 14853 USA ; Department of Biomedical Engineering, Cornell University, Ithaca, NY 14853 USA
| |
Collapse
|
130
|
Setyawati MI, Tay CY, Docter D, Stauber RH, Leong DT. Understanding and exploiting nanoparticles' intimacy with the blood vessel and blood. Chem Soc Rev 2015; 44:8174-99. [PMID: 26239875 DOI: 10.1039/c5cs00499c] [Citation(s) in RCA: 230] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While the blood vessel is seldom the target tissue, almost all nanomedicine will interact with blood vessels and blood at some point of time along its life cycle in the human body regardless of their intended destination. Despite its importance, many bionanotechnologists do not feature endothelial cells (ECs), the blood vessel cells, or consider blood effects in their studies. Including blood vessel cells in the study can greatly increase our understanding of the behavior of any given nanomedicine at the tissue of interest or to understand side effects that may occur in vivo. In this review, we will first describe the diversity of EC types found in the human body and their unique behaviors and possibly how these important differences can implicate nanomedicine behavior. Subsequently, we will discuss about the protein corona derived from blood with foci on the physiochemical aspects of nanoparticles (NPs) that dictate the protein corona characteristics. We would also discuss about how NPs characteristics can affect uptake by the endothelium. Subsequently, mechanisms of how NPs could cross the endothelium to access the tissue of interest. Throughout the paper, we will share some novel nanomedicine related ideas and insights that were derived from the understanding of the NPs' interaction with the ECs. This review will inspire more exciting nanotechnologies that had accounted for the complexities of the real human body.
Collapse
Affiliation(s)
- Magdiel Inggrid Setyawati
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | | | | | | | | |
Collapse
|
131
|
Guasch J, Conings B, Neubauer S, Rechenmacher F, Ende K, Rolli CG, Kappel C, Schaufler V, Micoulet A, Kessler H, Boyen HG, Cavalcanti-Adam EA, Spatz JP. Segregation versus colocalization: orthogonally functionalized binary micropatterned substrates regulate the molecular distribution in focal adhesions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:3737-3747. [PMID: 25981929 DOI: 10.1002/adma.201500900] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 04/05/2015] [Indexed: 06/04/2023]
Abstract
Orthogonally functionalized binary micropatterned substrates are produced using a novel protocol. The use of adequate peptido-mimetics enables an unprecedented segregation of purified αvβ3 and α5β1 integrins in adjacent microislands and evidences the preference of U2OS cells to colocalize such receptors. Moreover, this tendency can be altered by varying the geometry and composition of the micropatterns.
Collapse
Affiliation(s)
- Judith Guasch
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| | - Bert Conings
- Institute for Materials Research, Wetenschapspark 1, Diepenbeek, B-3590, Belgium
| | - Stefanie Neubauer
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Institute for Advanced Study (IAS) and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstr. 4, Garching, D-85747, Germany
| | - Florian Rechenmacher
- Institute for Advanced Study (IAS) and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstr. 4, Garching, D-85747, Germany
| | - Karen Ende
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
| | - Claudio G Rolli
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| | - Christian Kappel
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
| | - Viktoria Schaufler
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| | - Alexandre Micoulet
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| | - Horst Kessler
- Institute for Advanced Study (IAS) and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstr. 4, Garching, D-85747, Germany
| | - Hans-Gerd Boyen
- Institute for Materials Research, Wetenschapspark 1, Diepenbeek, B-3590, Belgium
| | - Elisabetta Ada Cavalcanti-Adam
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| | - Joachim P Spatz
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| |
Collapse
|
132
|
Zhang R, Wang N, Zhang M, Zhang LN, Guo ZX, Luo XG, Zhou H, He HP, Zhang TC. Rho/MRTF-A-Induced Integrin Expression Regulates Angiogenesis in Differentiated Multipotent Mesenchymal Stem Cells. Stem Cells Int 2015; 2015:534758. [PMID: 25949242 PMCID: PMC4408638 DOI: 10.1155/2015/534758] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/13/2015] [Accepted: 03/18/2015] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are known to undergo endothelial differentiation in response to treatment with vascular endothelial growth factor (VEGF), but their angiogenic ability is poorly characterized. In the present study, we aimed to further investigate the role of Rho/MRTF-A in angiogenesis by MSCs and the effect of the Rho/MRTF-A pathway on the expression of integrins α1β1 and α5β1, which are known to mediate physiological and pathological angiogenesis. Our results showed that increased expression of α1, α5, and β1 was observed during angiogenesis of differentiated MSCs, and the Rho/MRTF-A signaling pathway was demonstrated to be involved in regulating the expression of integrins α1, α5, and β1. Luciferase reporter assay and ChIP assay determined that MRTF-A could bind to and transactivate the integrin α1 and α5 promoters. Treatment with the Rho inhibitor C3 transferase, the Rho-associated protein kinase (ROCK) inhibitor Y27632 or with shMRTF-A inhibited both the upregulation of α1, α5, and β1 as well as angiogenesis. Furthermore, in human umbilical vein endothelial cells (HUVECs), MRTF-A deletion led to marked reductions in cell migration and vessel network formation compared with the control. These data demonstrate that Rho/MRTF-A signaling is an important mediator that controls integrin gene expression during MSC-mediated angiogenic processes.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Nan Wang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Man Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Li-Nan Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhi-Xia Guo
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Xue-Gang Luo
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Hao Zhou
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Hong-Peng He
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Tong-Cun Zhang
- Key Laboratory of Industrial Microbiology, Ministry of Education and Tianjin City, College of Biotechnology, Tianjin University of Science and Technology, Tianjin 300457, China
- Department of Biochemistry, Medical College, Wuhan University of Science and Technology, Wuhan 430081, China
| |
Collapse
|
133
|
Murphy PA, Begum S, Hynes RO. Tumor angiogenesis in the absence of fibronectin or its cognate integrin receptors. PLoS One 2015; 10:e0120872. [PMID: 25807551 PMCID: PMC4373772 DOI: 10.1371/journal.pone.0120872] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/10/2015] [Indexed: 11/18/2022] Open
Abstract
Binding of α5β1 and αvβ3/β5 integrin receptors on the endothelium to their fibronectin substrate in the extracellular matrix has been targeted as a possible means of blocking tumor angiogenesis and tumor growth. However, clinical trials of blocking antibodies and peptides have been disappointing despite promising preclinical results, leading to questions about the mechanism of the inhibitors and the reasons for their failure. Here, using tissue-specific and inducible genetics to delete the α5 and αv receptors in the endothelium or their fibronectin substrate, either in the endothelium or globally, we show that both are dispensable for tumor growth, in transplanted tumors as well as spontaneous and angiogenesis-dependent RIP-Tag-driven pancreatic adenocarcinomas. In the nearly complete absence of fibronectin, no differences in vascular density or the deposition of basement membrane laminins, ColIV, Nid1, Nid2, or the TGFβ binding matrix proteins, fibrillin-1 and -2, could be observed. Our results reveal that fibronectin and the endothelial fibronectin receptor subunits, α5 and αv, are dispensable for tumor angiogenesis, suggesting that the inhibition of angiogenesis induced by antibodies or small molecules may occur through a dominant negative effect, rather than a simple functional block.
Collapse
Affiliation(s)
- Patrick A. Murphy
- Howard Hughes Medical Institute, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Shahinoor Begum
- Howard Hughes Medical Institute, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Richard O. Hynes
- Howard Hughes Medical Institute, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
134
|
Wade RJ, Bassin EJ, Gramlich WM, Burdick JA. Nanofibrous hydrogels with spatially patterned biochemical signals to control cell behavior. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:1356-62. [PMID: 25640972 PMCID: PMC4412590 DOI: 10.1002/adma.201404993] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 12/01/2014] [Indexed: 05/18/2023]
Abstract
The ability to spatially pattern biochemical signals into nanofibrous materials using thiol-ene reactions of thiolated molecules to presented norbornene groups is demonstrated. This approach is used to pattern three molecules independently within one scaffold, to pattern molecules through the depth of a scaffold, and to spatially control cell adhesion and morphology.
Collapse
Affiliation(s)
- Ryan J Wade
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | | | | | | |
Collapse
|
135
|
Moore MC, Pandolfi V, McFetridge PS. Novel human-derived extracellular matrix induces in vitro and in vivo vascularization and inhibits fibrosis. Biomaterials 2015; 49:37-46. [PMID: 25725553 DOI: 10.1016/j.biomaterials.2015.01.022] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/23/2014] [Accepted: 01/20/2015] [Indexed: 10/24/2022]
Abstract
The inability to vascularize engineered organs and revascularize areas of infarction has been a major roadblock to delivering successful regenerative medicine therapies to the clinic. These investigations detail an isolated human extracellular matrix derived from the placenta (hPM) that induces vasculogenesis in vitro and angiogenesis in vivo within bioengineered tissues, with significant immune reductive properties. Compositional analysis showed ECM components (fibrinogen, laminin), angiogenic cytokines (angiogenin, FGF), and immune-related cytokines (annexins, DEFA1) in near physiological ratios. Gene expression profiles of endothelial cells seeded onto the matrix displayed upregulation of angiogenic genes (TGFB1, VEGFA), remodeling genes (MMP9, LAMA5) and vascular development genes (HAND2, LECT1). Angiogenic networks displayed a time dependent stability in comparison to current in vitro approaches that degrade rapidly. In vivo, matrix-dosed bioscaffolds showed enhanced angiogenesis and significantly reduced fibrosis in comparison to current angiogenic biomaterials. Implementation of this human placenta derived extracellular matrix provides an alternative to Matrigel and, due to its human derivation, its development may have significant clinical applications leading to advances in therapeutic angiogenesis techniques and tissue engineering.
Collapse
Affiliation(s)
- Marc C Moore
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, JG-56 Biomedical Sciences Building, P.O. Box 116131, Gainesville, FL 32611-6131, USA
| | - Vittoria Pandolfi
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, JG-56 Biomedical Sciences Building, P.O. Box 116131, Gainesville, FL 32611-6131, USA
| | - Peter S McFetridge
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, JG-56 Biomedical Sciences Building, P.O. Box 116131, Gainesville, FL 32611-6131, USA.
| |
Collapse
|
136
|
Ng SR, O'Hare D. An iridium oxide microelectrode for monitoring acute local pH changes of endothelial cells. Analyst 2015; 140:4224-31. [DOI: 10.1039/c5an00377f] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A microelectrode on a chip was modified to detect the local pH changes of the attached endothelial cells under the stimulation of thrombin.
Collapse
Affiliation(s)
- Shu Rui Ng
- Department of Bioengineering
- Imperial College London
- UK SW7 2AZ
- School of Chemical and Biomedical Engineering
- Division of Bioengineering
| | - Danny O'Hare
- Department of Bioengineering
- Imperial College London
- UK SW7 2AZ
| |
Collapse
|
137
|
Kim JW, Cochran FV, Cochran JR. A chemically cross-linked knottin dimer binds integrins with picomolar affinity and inhibits tumor cell migration and proliferation. J Am Chem Soc 2014; 137:6-9. [PMID: 25486381 PMCID: PMC4304478 DOI: 10.1021/ja508416e] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
![]()
Molecules that target and inhibit
αvβ3, αvβ5, and α5β1 integrins have
generated great interest
because of the role of these receptors in mediating angiogenesis and
metastasis. Attempts to increase the binding affinity and hence the
efficacy of integrin inhibitors by dimerization have been marginally
effective. In the present work, we achieved this goal by using oxime-based
chemical conjugation to synthesize dimers of integrin-binding cystine
knot (knottin) miniproteins with low-picomolar binding affinity to
tumor cells. A non-natural amino acid containing an aminooxy side
chain was introduced at different locations within a knottin monomer
and reacted with dialdehyde-containing cross-linkers of different
lengths to create knottin dimers with varying molecular topologies.
Dimers cross-linked through an aminooxy functional group located near
the middle of the protein exhibited higher apparent binding affinity
to integrin-expressing tumor cells compared with dimers cross-linked
through an aminooxy group near the C-terminus. In contrast, the cross-linker
length had no effect on the integrin binding affinity. A chemical-based
dimerization strategy was critical, as knottin dimers created through
genetic fusion to a bivalent antibody domain exhibited only modest
improvement (less than 5-fold) in tumor cell binding relative to the
knottin monomer. The best oxime-conjugated knottin dimer achieved
an unprecedented 150-fold increase in apparent binding affinity over
the knottin monomer. Also, this dimer bound 3650-fold stronger and
inhibited tumor cell migration and proliferation compared with cilengitide,
an integrin-targeting peptidomimetic that performed poorly in recent
clinical trials, suggesting promise for further therapeutic development.
Collapse
Affiliation(s)
- Jun W Kim
- Departments of †Bioengineering and ‡Chemical Engineering, Stanford University , Stanford, California 94305, United States
| | | | | |
Collapse
|
138
|
Santhakumar R, Vidyasekar P, Verma RS. Cardiogel: a nano-matrix scaffold with potential application in cardiac regeneration using mesenchymal stem cells. PLoS One 2014; 9:e114697. [PMID: 25521816 PMCID: PMC4270637 DOI: 10.1371/journal.pone.0114697] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/13/2014] [Indexed: 01/05/2023] Open
Abstract
3-Dimensional conditions for the culture of Bone Marrow-derived Stromal/Stem Cells (BMSCs) can be generated with scaffolds of biological origin. Cardiogel, a cardiac fibroblast-derived Extracellular Matrix (ECM) has been previously shown to promote cardiomyogenic differentiation of BMSCs and provide protection against oxidative stress. To determine the matrix composition and identify significant proteins in cardiogel, we investigated the differences in the composition of this nanomatrix and a BMSC-derived ECM scaffold, termed as ‘mesogel’. An optimized protocol was developed that resulted in efficient decellularization while providing the maximum yield of ECM. The proteins were sequentially solubilized using acetic acid, Sodium Dodecyl Sulfate (SDS) and Dithiothreitol (DTT). These proteins were then analyzed using surfactant-assisted in-solution digestion followed by nano-liquid chromatography and tandem mass spectrometry (nLC-MS/MS). The results of these analyses revealed significant differences in their respective compositions and 17 significant ECM/matricellular proteins were differentially identified between cardiogel and mesogel. We observed that cardiogel also promoted cell proliferation, adhesion and migration while enhancing cardiomyogenic differentiation and angiogenesis. In conclusion, we developed a reproducible method for efficient extraction and solubilization of in vitro cultured cell-derived extracellular matrix. We report several important proteins differentially identified between cardiogel and mesogel, which can explain the biological properties of cardiogel. We also demonstrated the cardiomyogenic differentiation and angiogenic potential of cardiogel even in the absence of any external growth factors. The transplantation of Bone Marrow derived Stromal/Stem Cells (BMSCs) cultured on such a nanomatrix has potential applications in regenerative therapy for Myocardial Infarction (MI).
Collapse
Affiliation(s)
- Rajalakshmi Santhakumar
- Stem cell and Molecular Biology Lab, Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, Tamil Nadu, India
| | - Prasanna Vidyasekar
- Stem cell and Molecular Biology Lab, Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, Tamil Nadu, India
| | - Rama Shanker Verma
- Stem cell and Molecular Biology Lab, Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai, Tamil Nadu, India
- * E-mail:
| |
Collapse
|
139
|
H-CRRETAWAC-OH, a lead structure for the development of radiotracer targeting integrin α5β1? BIOMED RESEARCH INTERNATIONAL 2014; 2014:243185. [PMID: 25374888 PMCID: PMC4211326 DOI: 10.1155/2014/243185] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 05/24/2014] [Indexed: 11/18/2022]
Abstract
Imaging of angiogenic processes is of great interest in preclinical research as well as in clinical settings. The most commonly addressed target structure for imaging angiogenesis is the integrin α(v)β(3). Here we describe the synthesis and evaluation of [(18)F]FProp-Cys(*)-Arg-Arg-Glu-Thr-Ala-Trp-Ala-Cys(*)-OH, a radiolabelled peptide designed to selectively target the integrin α(5)β(1). Conjugation of 4-nitrophenyl-(RS)-2-[(18)F]fluoropropionate provided [(18)F]FProp-Cys(*)-Arg-Arg-Glu-Thr-Ala-Trp-Ala-Cys(*)-OH in high radiochemical purity (>95%) and a radiochemical yield of approx. 55%. In vitro evaluation showed α(5)β(1) binding affinity in the nanomolar range, whereas affinity to α(v)β(3) and α(IIb)β(3) was >50 μM. Cell uptake studies using human melanoma M21 (α(v)β(3)-positive and α(5)β(1)-negative), human melanoma M21-L (α(v)β(3)-negative and α(5)β(1)-negative), and human prostate carcinoma DU145 (α(v)β(3)-negative and α(5)β(1)-positive) confirmed receptor-specific binding. The radiotracer was stable in human serum and showed low protein binding. Biodistribution studies showed tumour uptake ranging from 2.5 to 3.5% ID/g between 30 and 120 min post-injection. However, blocking studies and studies using mice bearing α(5)β(1)-negative M21 tumours did not confirm receptor-specific uptake of [(18)F]FProp-Cys(*)-Arg-Arg-Glu-Thr-Ala-Trp-Ala-Cys(*)-OH, although this radiopeptide revealed high affinity and substantial selectivity to α(5)β(1) in vitro. Further experiments are needed to study the in vivo metabolism of this peptide and to develop improved radiopeptide candidates suitable for PET imaging of α(5)β(1) expression in vivo.
Collapse
|
140
|
Geary SM, Salem AK. Exploiting the tumor phenotype using biodegradable submicron carriers of chemotherapeutic drugs. Crit Rev Oncog 2014; 19:269-80. [PMID: 25271435 DOI: 10.1615/critrevoncog.2014011518] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor tissues possess characteristics that distinguish them from healthy tissues and make them attractive targets for submicron carriers of chemotherapeutic drugs (CTX). CTX are generally administered systemically in free form to cancer patients resulting in unwanted cytotoxic effects and placing limitations on the deliverable CTX dose. In an effort to raise the therapeutic index of CTX there are now liposome-based CTX formulations in clinical use that are more tumor specific than the free form of CTX. However, progression to liposome-based chemotherapy in the clinic has been slow and there have been no approved formulations introduced in the last decade. Alternative carrier systems such as those made from the biodegradable polymer poly(lactic-co-glycolic) acid (PLGA) have been investigated in preclinical settings with promising outcomes. Here we review the principle behind biodegradable submicron carriers as CTX delivery vehicles for solid tumors with a specific focUS on liposomes and PLGA-based carriers, highlighting the strengths and weaknesses of each system.
Collapse
Affiliation(s)
- Sean M Geary
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, Iowa
| |
Collapse
|
141
|
Jullienne A, Roberts JM, Pop V, Paul Murphy M, Head E, Bix GJ, Badaut J. Juvenile traumatic brain injury induces long-term perivascular matrix changes alongside amyloid-beta accumulation. J Cereb Blood Flow Metab 2014; 34:1637-45. [PMID: 25052558 PMCID: PMC4269722 DOI: 10.1038/jcbfm.2014.124] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 04/28/2014] [Accepted: 06/05/2014] [Indexed: 02/08/2023]
Abstract
In our juvenile traumatic brain injury (jTBI) model, emergence of cognitive dysfunctions was observed up to 6 months after trauma. Here we hypothesize that early brain injury induces changes in the neurovascular unit (NVU) that would be associated with amyloid-beta (Aβ) accumulation. We investigated NVU changes for up to 6 months in a rat jTBI model, with a focus on the efflux protein P-glycoprotein (P-gp) and on the basement membrane proteins perlecan and fibronectin, all known to be involved in Aβ clearance. Rodent-Aβ staining is present and increased after jTBI around cerebral blood microvessels, and the diameter of those is decreased by 25% and 34% at 2 and 6 months, respectively, without significant angiogenesis. P-glycoprotein staining in endothelium is decreased by 22% and parallels an increase of perlecan and fibronectin staining around cerebral blood vessels. Altogether, these results strongly suggest that the emergence of long-term behavioral dysfunctions observed in rodent jTBI may be related to endothelial remodeling at the blood-brain barrier alongside vascular dysfunction and altered Aβ trafficking. This study shows that it is important to consider jTBI as a vascular disorder with long-term consequences on cognitive functions.
Collapse
Affiliation(s)
- Amandine Jullienne
- Department of Pediatrics, Loma Linda University, Loma Linda, California, USA
| | - Jill M Roberts
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Viorela Pop
- Department of Pediatrics, Loma Linda University, Loma Linda, California, USA
| | - M Paul Murphy
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Gregory J Bix
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Jérôme Badaut
- 1] Department of Pediatrics, Loma Linda University, Loma Linda, California, USA [2] Department of Physiology, Loma Linda University, Loma Linda, California, USA [3] CNRS UMR 5287, Bordeaux University, Bordeaux, France
| |
Collapse
|
142
|
Ackerman SE, Wilson CM, Kahn SA, Kintzing JR, Jindal DA, Cheshier SH, Grant GA, Cochran JR. A Bioengineered Peptide that Localizes to and Illuminates Medulloblastoma: A New Tool with Potential for Fluorescence-Guided Surgical Resection. Cureus 2014; 6:e207. [PMID: 28729960 PMCID: PMC5515084 DOI: 10.7759/cureus.207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Tumors of the central nervous system are challenging to treat due to the limited effectiveness and associated toxicities of chemotherapy and radiation therapy. For tumors that can be removed surgically, extent of malignant tissue resection has been shown to correlate with disease progression, recurrence, and survival. Thus, improved technologies for real-time brain tumor imaging are critically needed as tools for guided surgical resection. We previously engineered a novel peptide that binds with high affinity and unique specificity to αVβ3, αVβ5, and α5β1 integrins, which are present on tumor cells, and the vasculature of many cancers, including brain tumors. In the current study, we conjugated this engineered peptide to a near infrared fluorescent dye (Alexa Fluor 680), and used the resulting molecular probe for non-invasive whole body imaging of patient-derived medulloblastoma xenograft tumors implanted in the cerebellum of mice. The engineered peptide exhibited robust targeting and illumination of intracranial medulloblastoma following both intravenous and intraperitoneal injection routes. In contrast, a variant of the engineered peptide containing a scrambled integrin-binding sequence did not localize to brain tumors, demonstrating that tumor-targeting is driven by specific integrin interactions. Ex vivo imaging was used to confirm the presence of tumor and molecular probe localization to the cerebellar region. These results warrant further clinical development of the engineered peptide as a tool for image-guided resection of central nervous system tumors.
Collapse
Affiliation(s)
| | | | - Suzana A. Kahn
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | | | | | - Samuel H. Cheshier
- Department of Neurosurgery and Neurology, Stanford University School of Medicine & Lucile Packard Children’s Hospital, Department of Neurosurgery and Neurology, Stanford University School of Medicine & Lucile Packard Children’s Hospital at Stanford
| | - Gerald A. Grant
- Department of Neurosurgery, Stanford University School of Medicine
| | - Jennifer R. Cochran
- Department of Bioengineering and (by courtesy) Chemical Engineering, Stanford University
| |
Collapse
|
143
|
Mateo J, Berlin J, de Bono JS, Cohen RB, Keedy V, Mugundu G, Zhang L, Abbattista A, Davis C, Gallo Stampino C, Borghaei H. A first-in-human study of the anti-α5β1 integrin monoclonal antibody PF-04605412 administered intravenously to patients with advanced solid tumors. Cancer Chemother Pharmacol 2014; 74:1039-46. [PMID: 25212537 PMCID: PMC4209234 DOI: 10.1007/s00280-014-2576-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 08/22/2014] [Indexed: 11/26/2022]
Abstract
Purpose A first-in-human clinical trial of a fully human, Fc-engineered IgG1 monoclonal antibody targeting integrin α5β1 was conducted to evaluate tolerability, maximum tolerated dose, pharmacokinetics, pharmacodynamics and preliminary anti-tumor activity. Methods Escalating doses of PF-04605412 were given IV on day 1, 28 and every 2 weeks thereafter to patients with advanced solid tumors until disease progression or unacceptable toxicity. Sequential dose cohorts were evaluated based on a modified 3 + 3 dose-escalation design. The starting dose was 7.5 mg based on preclinical data. Results Thirty-three patients were enrolled to six dose levels (7.5, 11.25, 16.9, 34, 68 and 136 mg). Twenty-three patients were evaluable for the primary endpoint (determination of the maximum tolerated dose). Five patients required permanent drug discontinuation due to acute infusion-related reactions, which occurred as grade 3 events in two patients. PK analysis indicated that the targeted drug exposure based on preclinical models was not achieved by the tolerated doses and PK modeling suggesting that doses at least fivefold higher would be necessary. No anti-tumor activity was observed. Conclusion Based on the safety data, the risks associated with the likelihood of significant cytokine-mediated infusion reactions at higher doses, the projected high dose necessary to affect on the biological target and the lack of anti-tumor activity at the doses explored, the trial was prematurely terminated without determining a formal maximum tolerated dose. Further clinical development of PF-04605412 has been discontinued. Electronic supplementary material The online version of this article (doi:10.1007/s00280-014-2576-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- J. Mateo
- The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, Sutton, UK
| | - J. Berlin
- Vanderbilt-Ingram Cancer Center, Nashville, TN USA
| | - J. S. de Bono
- The Royal Marsden NHS Foundation Trust and The Institute of Cancer Research, Sutton, UK
| | - R. B. Cohen
- Developmental Therapeutics Program and Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111 USA
| | - V. Keedy
- Vanderbilt-Ingram Cancer Center, Nashville, TN USA
| | | | | | | | | | | | - H. Borghaei
- Developmental Therapeutics Program and Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA 19111 USA
| |
Collapse
|
144
|
|
145
|
PET radiopharmaceuticals for imaging integrin expression: tracers in clinical studies and recent developments. BIOMED RESEARCH INTERNATIONAL 2014; 2014:871609. [PMID: 25013808 PMCID: PMC4072020 DOI: 10.1155/2014/871609] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 04/29/2014] [Indexed: 11/17/2022]
Abstract
Noninvasive determination of integrin expression has become an interesting approach in nuclear medicine. Since the discovery of the first 18F-labeled cyclic RGD peptide as radiotracer for imaging integrin αvβ3 expression in vivo, there have been carried out enormous efforts to develop RGD peptides for PET imaging. Moreover, in recent years, additional integrins, including α5β1 and αvβ6 came into the focus of pharmaceutical radiochemistry. This review will discuss the tracers already evaluated in clinical trials and summarize the preliminary outcome. It will also give an overview on recent developments to further optimize the first-generation compounds such as [18F]Galacto-RGD. This includes recently developed 18F-labeling strategies and also new approaches in 68Ga-complex chemistry. Furthermore, the approaches to develop radiopharmaceuticals targeting integrin α5β1 and αvβ6 will be summarized and discussed.
Collapse
|
146
|
Wei X, Chen X, Ying M, Lu W. Brain tumor-targeted drug delivery strategies. Acta Pharm Sin B 2014; 4:193-201. [PMID: 26579383 PMCID: PMC4629063 DOI: 10.1016/j.apsb.2014.03.001] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 02/07/2014] [Accepted: 02/24/2014] [Indexed: 12/18/2022] Open
Abstract
Despite the application of aggressive surgery, radiotherapy and chemotherapy in clinics, brain tumors are still a difficult health challenge due to their fast development and poor prognosis. Brain tumor-targeted drug delivery systems, which increase drug accumulation in the tumor region and reduce toxicity in normal brain and peripheral tissue, are a promising new approach to brain tumor treatments. Since brain tumors exhibit many distinctive characteristics relative to tumors growing in peripheral tissues, potential targets based on continuously changing vascular characteristics and the microenvironment can be utilized to facilitate effective brain tumor-targeted drug delivery. In this review, we briefly describe the physiological characteristics of brain tumors, including blood–brain/brain tumor barriers, the tumor microenvironment, and tumor stem cells. We also review targeted delivery strategies and introduce a systematic targeted drug delivery strategy to overcome the challenges.
Collapse
|
147
|
Turner CJ, Badu-Nkansah K, Crowley D, van der Flier A, Hynes RO. Integrin-α5β1 is not required for mural cell functions during development of blood vessels but is required for lymphatic-blood vessel separation and lymphovenous valve formation. Dev Biol 2014; 392:381-92. [PMID: 24858485 DOI: 10.1016/j.ydbio.2014.05.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/22/2014] [Accepted: 05/08/2014] [Indexed: 11/25/2022]
Abstract
Integrin α5β1 is essential for vascular development but it remains unclear precisely where and how it functions. Here, we report that deletion of the gene encoding the integrin-α5 subunit (Itga5) using the Pdgfrb-Cre transgenic mouse line, leads to oedema, haemorrhage and increased levels of embryonic lethality. Unexpectedly, these defects were not caused by loss of α5 from Pdgfrb-Cre expressing mural cells (pericytes and vascular smooth muscle cells), which wrap around the endothelium and stabilise blood vessels, nor by defects in the heart or great vessels, but were due to abnormal development of the lymphatic vasculature. Reminiscent of the pathologies seen in the human lymphatic malformation, fetal cystic hygroma, α5 mutants display defects both in the separation of their blood and lymphatic vasculature and in the formation of the lymphovenous valves. As a consequence, α5-deficient mice develop dilated, blood-filled lymphatic vessels and lymphatic capillaries that are ectopically covered with smooth muscle cells. Analysis of the expression of Pdgfrb during lymphatic development suggests that these defects probably arise from loss of α5β1 integrin in subsets of specialised Prox1(+)Pdgfrb(+) venous endothelial cells that are essential for the separation of the jugular lymph sac from the cardinal vein and formation of the lymphovenous valve leaflets.
Collapse
Affiliation(s)
- Christopher J Turner
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kwabena Badu-Nkansah
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Denise Crowley
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arjan van der Flier
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard O Hynes
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
148
|
Amado-Azevedo J, Valent ET, Van Nieuw Amerongen GP. Regulation of the endothelial barrier function: a filum granum of cellular forces, Rho-GTPase signaling and microenvironment. Cell Tissue Res 2014; 355:557-76. [PMID: 24633925 DOI: 10.1007/s00441-014-1828-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/24/2014] [Indexed: 12/20/2022]
Abstract
Although the endothelium is an extremely thin single-cell layer, it performs exceedingly well in preventing blood fluids from leaking into the surrounding tissues. However, specific pathological conditions can affect this cell layer, compromising the integrity of the barrier. Vascular leakage is a hallmark of many cardiovascular diseases and despite its medical importance, no specialized therapies are available to prevent it or reduce it. Small guanosine triphosphatases (GTPases) of the Rho family are known to be key regulators of various aspects of cell behavior and studies have shown that they can exert both positive and negative effects on endothelial barrier integrity. Moreover, extracellular matrix stiffness has now been implicated in the regulation of Rho-GTPase signaling, which has a direct impact on the integrity of endothelial junctions. However, knowledge about both the precise mechanism of this regulation and the individual contribution of the specific regulatory proteins remains fragmentary. In this review, we discuss recent findings concerning the balanced activities of Rho-GTPases and, in particular, aspects of the regulation of the endothelial barrier. We highlight the role of Rho-GTPases in the intimate relationships between biomechanical forces, microenvironmental influences and endothelial intercellular junctions, which are all interwoven in a beautiful filigree-like fashion.
Collapse
Affiliation(s)
- Joana Amado-Azevedo
- Laboratory for Physiology, Institute for Cardiovascular Research, VU University Medical Center, Van den Boechorststraat 7, 1081BT, Amsterdam, The Netherlands
| | | | | |
Collapse
|
149
|
Kostourou V, Papalazarou V. Non-collagenous ECM proteins in blood vessel morphogenesis and cancer. Biochim Biophys Acta Gen Subj 2014; 1840:2403-13. [PMID: 24576673 DOI: 10.1016/j.bbagen.2014.02.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/14/2014] [Accepted: 02/17/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND The extracellular matrix (ECM) is constituted by diverse composite structures, which determine the specific to each organ, histological architecture and provides cells with biological information, mechanical support and a scaffold for adhesion and migration. The pleiotropic effects of the ECM stem from the dynamic changes in its molecular composition and the ability to remodel in order to effectively regulate biological outcomes. Besides collagens, fibronectin and laminin are two major fiber-forming constituents of various ECM structures. SCOPE OF REVIEW This review will focus on the properties and the biological functions of non-collagenous extracellular matrix especially on laminin and fibronectin that are currently emerging as important regulators of blood vessel formation and function in health and disease. MAJOR CONCLUSIONS The ECM is a fundamental component of the microenvironment of blood vessels, with activities extending beyond providing a vascular scaffold; extremely versatile it directly or indirectly modulates all essential cellular functions crucial for angiogenesis, including cell adhesion, migration, proliferation, differentiation and lumen formation. Specifically, fibronectin and laminins play decisive roles in blood vessel morphogenesis both during embryonic development and in pathological conditions, such as cancer. GENERAL SIGNIFICANCE Emerging evidence demonstrates the importance of ECM function during embryonic development, organ formation and tissue homeostasis. A wealth of data also illustrates the crucial role of the ECM in several human pathophysiological processes, including fibrosis, skeletal diseases, vascular pathologies and cancer. Notably, several ECM components have been identified as potential therapeutic targets for various diseases, including cancer. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Vassiliki Kostourou
- Vascular Adhesion Lab, BSRC Alexander Fleming, 34 Fleming Str., Vari, 166 72 Athens, Greece
| | - Vassilis Papalazarou
- Vascular Adhesion Lab, BSRC Alexander Fleming, 34 Fleming Str., Vari, 166 72 Athens, Greece
| |
Collapse
|
150
|
Chung HJ, Mahalingam M. Angiogenesis, vasculogenic mimicry and vascular invasion in cutaneous malignant melanoma – implications for therapeutic strategies and targeted therapies. Expert Rev Anticancer Ther 2014; 14:621-39. [DOI: 10.1586/14737140.2014.883281] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|