51
|
Xu X, Chen X, Li J. Natural protein bioinspired materials for regeneration of hard tissues. J Mater Chem B 2020; 8:2199-2215. [DOI: 10.1039/d0tb00139b] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
This review describes the protein bioinspired materials for the repair of hard tissues such as enamel, dentin and bone.
Collapse
Affiliation(s)
- Xinyuan Xu
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Xingyu Chen
- College of Medicine
- Southwest Jiaotong University
- Chengdu 610003
- China
| | - Jianshu Li
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu 610065
- P. R. China
| |
Collapse
|
52
|
Mammoto A, Mammoto T. Vascular Niche in Lung Alveolar Development, Homeostasis, and Regeneration. Front Bioeng Biotechnol 2019; 7:318. [PMID: 31781555 PMCID: PMC6861452 DOI: 10.3389/fbioe.2019.00318] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/25/2019] [Indexed: 12/28/2022] Open
Abstract
Endothelial cells (ECs) constitute small capillary blood vessels and contribute to delivery of nutrients, oxygen and cellular components to the local tissues, as well as to removal of carbon dioxide and waste products from the tissues. Besides these fundamental functions, accumulating evidence indicates that capillary ECs form the vascular niche. In the vascular niche, ECs reciprocally crosstalk with resident cells such as epithelial cells, mesenchymal cells, and immune cells to regulate development, homeostasis, and regeneration in various organs. Capillary ECs supply paracrine factors, called angiocrine factors, to the adjacent cells in the niche and orchestrate these processes. Although the vascular niche is anatomically and functionally well-characterized in several organs such as bone marrow and neurons, the effects of endothelial signals on other resident cells and anatomy of the vascular niche in the lung have not been well-explored. This review discusses the role of alveolar capillary ECs in the vascular niche during development, homeostasis and regeneration.
Collapse
Affiliation(s)
- Akiko Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Tadanori Mammoto
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
53
|
Clauder F, Czerniak AS, Friebe S, Mayr SG, Scheinert D, Beck-Sickinger AG. Endothelialization of Titanium Surfaces by Bioinspired Cell Adhesion Peptide Coatings. Bioconjug Chem 2019; 30:2664-2674. [DOI: 10.1021/acs.bioconjchem.9b00573] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Affiliation(s)
- Franziska Clauder
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Anne Sophie Czerniak
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| | - Sabrina Friebe
- Leibniz-Institute of Surface Engineering (IOM), Permoserstrasse 15, 04318 Leipzig, Germany
| | - Stefan G. Mayr
- Leibniz-Institute of Surface Engineering (IOM), Permoserstrasse 15, 04318 Leipzig, Germany
| | - Dierk Scheinert
- Department of Angiology, University Hospital Leipzig, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Annette G. Beck-Sickinger
- Institute of Biochemistry, Faculty of Life Sciences, Leipzig University, Brüderstrasse 34, 04103 Leipzig, Germany
| |
Collapse
|
54
|
The role of elastin-derived peptides in human physiology and diseases. Matrix Biol 2019; 84:81-96. [PMID: 31295577 DOI: 10.1016/j.matbio.2019.07.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/03/2019] [Accepted: 07/07/2019] [Indexed: 12/12/2022]
Abstract
Once considered as inert, the extracellular matrix recently revealed to be biologically active. Elastin is one of the most important components of the extracellular matrix. Many vital organs including arteries, lungs and skin contain high amounts of elastin to assure their correct function. Physiologically, the organism contains a determined quantity of elastin from the early development which may remain physiologically constant due to its very long half-life and very low turnover. Taking into consideration the continuously ongoing challenges during life, there is a physiological degradation of elastin into elastin-derived peptides which is accentuated in several disease states such as obstructive pulmonary diseases, atherosclerosis and aortic aneurysm. These elastin-derived peptides have been shown to have various biological effects mediated through their interaction with their cognate receptor called elastin receptor complex eliciting several signal transduction pathways. In this review, we will describe the production and the biological effects of elastin-derived peptides in physiology and pathology.
Collapse
|
55
|
Arora S, Yim EKF, Toh YC. Environmental Specification of Pluripotent Stem Cell Derived Endothelial Cells Toward Arterial and Venous Subtypes. Front Bioeng Biotechnol 2019; 7:143. [PMID: 31259171 PMCID: PMC6587665 DOI: 10.3389/fbioe.2019.00143] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022] Open
Abstract
Endothelial cells (ECs) are required for a multitude of cardiovascular clinical applications, such as revascularization of ischemic tissues or endothelialization of tissue engineered grafts. Patient derived primary ECs are limited in number, have donor variabilities and their in vitro phenotypes and functions can deteriorate over time. This necessitates the exploration of alternative EC sources. Although there has been a recent surge in the use of pluripotent stem cell derived endothelial cells (PSC-ECs) for various cardiovascular clinical applications, current differentiation protocols yield a heterogeneous EC population, where their specification into arterial or venous subtypes is undefined. Since arterial and venous ECs are phenotypically and functionally different, inappropriate matching of exogenous ECs to host sites can potentially affect clinical efficacy, as exemplified by venous graft mismatch when placed into an arterial environment. Therefore, there is a need to design and employ environmental cues that can effectively modulate PSC-ECs into a more homogeneous arterial or venous phenotype for better adaptation to the host environment, which will in turn contribute to better application efficacy. In this review, we will first give an overview of the developmental and functional differences between arterial and venous ECs. This provides the foundation for our subsequent discussion on the different bioengineering strategies that have been investigated to varying extent in providing biochemical and biophysical environmental cues to mature PSC-ECs into arterial or venous subtypes. The ability to efficiently leverage on a combination of biochemical and biophysical environmental cues to modulate intrinsic arterio-venous specification programs in ECs will greatly facilitate future translational applications of PSC-ECs. Since the development and maintenance of arterial and venous ECs in vivo occur in disparate physio-chemical microenvironments, it is conceivable that the application of these environmental factors in customized combinations or magnitudes can be used to selectively mature PSC-ECs into an arterial or venous subtype.
Collapse
Affiliation(s)
- Seep Arora
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore.,Biomedical Institute for Global Health Research and Technology (BIGHEART), National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Program, National University of Singapore, Singapore, Singapore
| |
Collapse
|
56
|
Sarker MD, Naghieh S, Sharma NK, Ning L, Chen X. Bioprinting of Vascularized Tissue Scaffolds: Influence of Biopolymer, Cells, Growth Factors, and Gene Delivery. JOURNAL OF HEALTHCARE ENGINEERING 2019; 2019:9156921. [PMID: 31065331 PMCID: PMC6466897 DOI: 10.1155/2019/9156921] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 02/03/2019] [Indexed: 01/16/2023]
Abstract
Over the past decades, tissue regeneration with scaffolds has achieved significant progress that would eventually be able to solve the worldwide crisis of tissue and organ regeneration. While the recent advancement in additive manufacturing technique has facilitated the biofabrication of scaffolds mimicking the host tissue, thick tissue regeneration remains challenging to date due to the growing complexity of interconnected, stable, and functional vascular network within the scaffold. Since the biological performance of scaffolds affects the blood vessel regeneration process, perfect selection and manipulation of biological factors (i.e., biopolymers, cells, growth factors, and gene delivery) are required to grow capillary and macro blood vessels. Therefore, in this study, a brief review has been presented regarding the recent progress in vasculature formation using single, dual, or multiple biological factors. Besides, a number of ways have been presented to incorporate these factors into scaffolds. The merits and shortcomings associated with the application of each factor have been highlighted, and future research direction has been suggested.
Collapse
Affiliation(s)
- M. D. Sarker
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Saman Naghieh
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - N. K. Sharma
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Liqun Ning
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
57
|
Jin HS, Song K, Baek JH, Lee JE, Kim DJ, Nam GW, Kang NJ, Lee DW. Identification of Matrix Metalloproteinase-1-Suppressive Peptides in Feather Keratin Hydrolysate. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:12719-12729. [PMID: 30395462 DOI: 10.1021/acs.jafc.8b05213] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Inhibition of matrix metalloproteinases (MMPs), which degrade collagen and elastin in the dermis of normal skin, is a key strategy for anti-skin aging. In this study, we identified five low-molecular-weight (LMW, <1 kDa) MMP-1-suppressive peptides in feather keratin hydrolysate (FKH) obtained by anaerobic digestion with an extremophilic bacterium. FKH was first subjected to ultrafiltration, followed by size-exclusion chromatography and liquid chromatography/electrospray ionization tandem mass spectrometry analysis. Chemically synthesized peptides identical to the sequences identified suppressed MMP expression in human dermal fibroblasts (HDFs). To investigate the impact of the MMP-1-suppressive peptides on the signaling pathway, we performed antibody array phosphorylation profiling of HDFs. The results suggested that the peptide GGFDL regulates ultraviolet-B-induced MMP-1 expression by inhibiting mitogen-activated protein kinases and nuclear factor κB signaling pathways as well as histone modification. Thus, LMW feather keratin peptides could serve as novel bioactive compounds to protect the skin against intrinsic and extrinsic factors.
Collapse
Affiliation(s)
- Hyeon-Su Jin
- Department of Biotechnology , Yonsei University , Seoul 03722 , South Korea
| | - Kyeongseop Song
- School of Food Science and Biotechnology , Kyungpook National University , Daegu 41566 , South Korea
| | - Je-Hyun Baek
- Center of Biomedical Mass Spectrometry (CBMS) , DiatechKorea Company, Limited , Seoul 05808 , South Korea
| | - Jae-Eun Lee
- School of Food Science and Biotechnology , Kyungpook National University , Daegu 41566 , South Korea
| | - Da Jeong Kim
- School of Food Science and Biotechnology , Kyungpook National University , Daegu 41566 , South Korea
| | - Gae-Won Nam
- School of Cosmetics , Seowon University , Cheongju 28674 , South Korea
| | - Nam Joo Kang
- School of Food Science and Biotechnology , Kyungpook National University , Daegu 41566 , South Korea
| | - Dong-Woo Lee
- Department of Biotechnology , Yonsei University , Seoul 03722 , South Korea
| |
Collapse
|
58
|
Using a Classifier Fusion Strategy to Identify Anti-angiogenic Peptides. Sci Rep 2018; 8:14062. [PMID: 30218091 PMCID: PMC6138733 DOI: 10.1038/s41598-018-32443-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/07/2018] [Indexed: 12/27/2022] Open
Abstract
Anti-angiogenic peptides perform distinct physiological functions and potential therapies for angiogenesis-related diseases. Accurate identification of anti-angiogenic peptides may provide significant clues to understand the essential angiogenic homeostasis within tissues and develop antineoplastic therapies. In this study, an ensemble predictor is proposed for anti-angiogenic peptide prediction by fusing an individual classifier with the best sensitivity and another individual one with the best specificity. We investigate predictive capabilities of various feature spaces with respect to the corresponding optimal individual classifiers and ensemble classifiers. The accuracy and Matthew’s Correlation Coefficient (MCC) of the ensemble classifier trained by Bi-profile Bayes (BpB) features are 0.822 and 0.649, respectively, which represents the highest prediction results among the investigated prediction models. Discriminative features are obtained from BpB using the Relief algorithm followed by the Incremental Feature Selection (IFS) method. The sensitivity, specificity, accuracy, and MCC of the ensemble classifier trained by the discriminative features reach up to 0.776, 0.888, 0.832, and 0.668, respectively. Experimental results indicate that the proposed method is far superior to the previous study for anti-angiogenic peptide prediction.
Collapse
|
59
|
Salesse S, Odoul L, Chazée L, Garbar C, Duca L, Martiny L, Mahmoudi R, Debelle L. Elastin molecular aging promotes MDA-MB-231 breast cancer cell invasiveness. FEBS Open Bio 2018; 8:1395-1404. [PMID: 30186741 PMCID: PMC6120250 DOI: 10.1002/2211-5463.12455] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/30/2018] [Accepted: 05/15/2018] [Indexed: 01/22/2023] Open
Abstract
Elastin is a long-lived extracellular matrix protein responsible for the structural integrity and function of tissues. Breast cancer elastosis is a complex phenomenon resulting in both the deposition of elastotic masses and the local production of elastin fragments. In invasive human breast cancers, an increase in elastosis is correlated with severity of the disease and age of the patient. Elastin-derived peptides (EDPs) are a hallmark of aging and are matrikines - matrix fragments having the ability to regulate cell physiology. They are known to promote processes linked to tumor progression, but their effects on breast cancer cells remain unexplored. Our data show that EDPs enhance the invasiveness of MDA-MB-231 breast cancer cells through the engagement of matrix metalloproteases 14 and 2. We therefore suggest that elastosis and/or an aged stroma could promote breast cancer cell invasiveness.
Collapse
Affiliation(s)
- Stéphanie Salesse
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| | - Ludivine Odoul
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| | - Lise Chazée
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| | - Christian Garbar
- Biopathology Department Institut Jean Godinot-Unicancer Reims France.,DERM-I-C EA7319 Université de Reims Champagne Ardenne France
| | - Laurent Duca
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| | - Laurent Martiny
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| | - Rachid Mahmoudi
- Faculty of Medicine, EA3797 University of Reims Champagne-Ardenne France.,Department of Geriatrics and Internal Medicine Maison Blanche Hospital Reims University Hospitals France
| | - Laurent Debelle
- UMR CNRS/URCA 7369 SFR CAP Santé Faculty of Sciences University of Reims Champagne-Ardenne France
| |
Collapse
|
60
|
Impact of Elastin-Derived Peptide VGVAPG on Matrix Metalloprotease-2 and -9 and the Tissue Inhibitor of Metalloproteinase-1, -2, -3 and -4 mRNA Expression in Mouse Cortical Glial Cells In Vitro. Neurotox Res 2018; 35:100-110. [PMID: 30062663 PMCID: PMC6313372 DOI: 10.1007/s12640-018-9935-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 12/22/2022]
Abstract
Degradation products of elastin, i.e. elastin-derived peptides (EDPs), are involved in various physiological and pathological processes. EDPs are detectable in cerebrospinal fluid in healthy people and in patients after ischemic stroke. However, to date, no studies concerning the role of EDP in the nervous system were conducted. Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) play important roles during the repair phases of cerebral ischemia, particularly during angiogenesis and reestablishment of cerebral blood flow. Therefore, the aim of this study was to investigate the impact of the specific elastin-derived peptide VGVAPG on Mmp-2, -9 and Timp-1, -2, -3 and -4 mRNA expression in mouse cortical glial cells in vitro. Primary glial cells were maintained in DMEM/F12 without phenol red supplemented with 10% fetal bovine serum and the cells were exposed to 50 nM, 1 and 50 μM of the VGVAPG peptide. After 3 and 6 h of exposition to the peptide, expression of Mmp-2, -9 and Timp-1, -2, -3 and -4 mRNA was measured. Moreover, siRNA gene knockdown, cytotoxicity and apoptosis measurement were included in our experiments, which showed that VGVAPG in a wide range of concentrations exhibited neither proapoptotic nor cytotoxic properties in mouse glial cells in vitro. The peptides enhanced mRNA expression of Timp-2 and Timp-3 genes in an elastin-binding protein (EBP)-dependent manner. However, changes in mRNA expression of Mmp-2, Mmp-9 and Timp-4 were partially EBP-dependent. The decrease in mRNA expression of Timp-1 was EBP-independent. However, further studies underlying the VGVAPG peptide’s mechanism of action in the nervous system are necessary.
Collapse
|
61
|
Collagen-Elastin and Collagen-Glycosaminoglycan Scaffolds Promote Distinct Patterns of Matrix Maturation and Axial Vascularization in Arteriovenous Loop-Based Soft Tissue Flaps. Ann Plast Surg 2018; 79:92-100. [PMID: 28542070 DOI: 10.1097/sap.0000000000001096] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Autologous free flaps are the criterion standard for reconstructions of complex soft tissue defects; however, they are limited by donor-site morbidities. The arteriovenous (AV) loop model enables the generation of soft tissue constructs based on acellular dermal matrices with a functional microvasculature and minimal donor site morbidity. The ideal scaffold for AV loop-based tissue engineering has not been determined. METHODS AV loops were placed into subcutaneous isolation chambers filled with either a collagen-elastin scaffold or a collagen-glycosaminoglycan scaffold in the thighs of rats. Matrix elasticity, neoangiogenesis, cell migration, and proliferation were compared after 14 and 28 days. RESULTS Mean vessel count and area had increased in both matrices at 28 compared with 14 days. Collagen-elastin matrices showed a higher mean vessel count and area compared with collagen-glycosaminoglycan matrices at 14 days. At 28 days, a more homogeneous vascular network and higher cell counts were observed in collagen-elastin matrices. Collagen-glycosaminoglycan matrices, however, exhibited less volume loss at day 28. CONCLUSIONS Collagen-based scaffolds are suitable for soft tissue engineering in conjunction with the AV loop technique. These scaffolds exhibit distinct patterns of angiogenesis, cell migration, and proliferation and may in the future serve as the basis of tissue-engineered free flaps as an individualized treatment concept for critical wounds.
Collapse
|
62
|
Da Silva J, Lameiras P, Beljebbar A, Berquand A, Villemin M, Ramont L, Dukic S, Nuzillard JM, Molinari M, Gautier M, Brassart-Pasco S, Brassart B. Structural characterization and in vivo pro-tumor properties of a highly conserved matrikine. Oncotarget 2018; 9:17839-17857. [PMID: 29707150 PMCID: PMC5915158 DOI: 10.18632/oncotarget.24894] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/25/2018] [Indexed: 01/13/2023] Open
Abstract
Elastin-derived peptides (EDPs) exert protumor activities by increasing tumor growth, migration and invasion. A number of studies have highlighted the potential of VGVAPG consensus sequence-derived elastin-like polypeptides whose physicochemical properties and biocompatibility are particularly suitable for in vivo applications, such as drug delivery and tissue engineering. However, among the EDPs, the influence of elastin-derived nonapeptides (xGxPGxGxG consensus sequence) remains unknown. Here, we show that the AGVPGLGVG elastin peptide (AG-9) present in domain-26 of tropoelastin is more conserved than the VGVAPG elastin peptide (VG-6) from domain-24 in mammals. The results demonstrate that the structural features of AG-9 and VG-6 peptides are similar. CD, NMR and FTIR spectroscopies show that AG-9 and VG-6 present the same conformation, which includes a mixture of random coils and β-turn structures. On the other hand, the supraorganization differs between peptides, as demonstrated by AFM. The VG-6 peptide gathers in spots, whereas the AG-9 peptide aggregates into short amyloid-like fibrils. An in vivo study showed that AG-9 peptides promote tumor progression to a greater extent than do VG-6 peptides. These results were confirmed by in vitro studies such as 2D and 3D proliferation assays, migration assays, adhesion assays, proteinase secretion studies and pseudotube formation assays to investigate angiogenesis. Our findings suggest the possibility that the AG-9 peptide present in patient sera may dramatically influence cancer progression and could be used in the design of new, innovative antitumor therapies.
Collapse
Affiliation(s)
- Jordan Da Silva
- UMR CNRS/URCA 7369 MEDyC, Université de Reims Champagne Ardenne, UFR Médecine, 51095 Reims, France
| | - Pedro Lameiras
- ICMR, CNRS UMR 7312, UFR de Pharmacie, Université de Reims Champagne-Ardenne, 51096 Reims, France
| | - Abdelilah Beljebbar
- UMR CNRS/URCA 7369 MEDyC, Université de Reims Champagne Ardenne, UFR Médecine, 51095 Reims, France
| | - Alexandre Berquand
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51100 Reims, France
| | - Matthieu Villemin
- UMR CNRS/URCA 7369 MEDyC, Université de Reims Champagne Ardenne, UFR Médecine, 51095 Reims, France
| | - Laurent Ramont
- UMR CNRS/URCA 7369 MEDyC, Université de Reims Champagne Ardenne, UFR Médecine, 51095 Reims, France
- CHU de Reims, Laboratoire Central de Biochimie, 51092 Reims, France
| | - Sylvain Dukic
- UMR CNRS/URCA 7369 MEDyC, Université de Reims Champagne Ardenne, UFR Médecine, 51095 Reims, France
| | - Jean-Marc Nuzillard
- ICMR, CNRS UMR 7312, UFR de Pharmacie, Université de Reims Champagne-Ardenne, 51096 Reims, France
| | - Michael Molinari
- Laboratoire de Recherche en Nanosciences, LRN-EA4682, Université de Reims Champagne-Ardenne, 51100 Reims, France
| | - Mathieu Gautier
- Laboratoire de Physiologie Cellulaire et Moléculaire, LPCM - EA4667, Université de Picardie Jules Verne, UFR Sciences, F-80039 Amiens, France
| | - Sylvie Brassart-Pasco
- UMR CNRS/URCA 7369 MEDyC, Université de Reims Champagne Ardenne, UFR Médecine, 51095 Reims, France
| | - Bertrand Brassart
- UMR CNRS/URCA 7369 MEDyC, Université de Reims Champagne Ardenne, UFR Médecine, 51095 Reims, France
| |
Collapse
|
63
|
Boraldi F, Moscarelli P, Bochicchio B, Pepe A, Salvi AM, Quaglino D. Heparan sulfates facilitate harmless amyloidogenic fibril formation interacting with elastin-like peptides. Sci Rep 2018; 8:3115. [PMID: 29449596 PMCID: PMC5814424 DOI: 10.1038/s41598-018-21472-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/31/2018] [Indexed: 12/12/2022] Open
Abstract
Heparan sulfates (HSs) modulate tissue elasticity in physiopathological conditions by interacting with various matrix constituents as tropoelastin and elastin-derived peptides. HSs bind also to protein moieties accelerating amyloid formation and influencing cytotoxic properties of insoluble fibrils. Interestingly, amyloidogenic polypeptides, despite their supposed pathogenic role, have been recently explored as promising bio-nanomaterials due to their unique and interesting properties. Therefore, we investigated the interactions of HSs, obtained from different sources and exhibiting various degree of sulfation, with synthetic amyloidogenic elastin-like peptides (ELPs), also looking at the effects of these interactions on cell viability and cell behavior using in vitro cultured fibroblasts, as a prototype of mesenchymal cells known to modulate the soft connective tissue environment. Results demonstrate, for the first time, that HSs, with differences depending on their sulfation pattern and chain length, interact with ELPs accelerating aggregation kinetics and amyloid-like fibril formation as well as self-association. Furthermore, these fibrils do not negatively affect fibroblasts’ cell growth and parameters of redox balance, and influence cellular adhesion properties. Data provide information for a better understanding of the interactions altering the elastic component in aging and in pathologic conditions and may pave the way for the development of composite matrix-based biomaterials.
Collapse
Affiliation(s)
- Federica Boraldi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Pasquale Moscarelli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Antonietta Pepe
- Department of Sciences, University of Basilicata, Potenza, Italy
| | - Anna M Salvi
- Department of Sciences, University of Basilicata, Potenza, Italy
| | - Daniela Quaglino
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| |
Collapse
|
64
|
Ricard-Blum S, Vallet SD. Fragments generated upon extracellular matrix remodeling: Biological regulators and potential drugs. Matrix Biol 2017; 75-76:170-189. [PMID: 29133183 DOI: 10.1016/j.matbio.2017.11.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/05/2017] [Accepted: 11/07/2017] [Indexed: 12/13/2022]
Abstract
The remodeling of the extracellular matrix (ECM) by several protease families releases a number of bioactive fragments, which regulate numerous biological processes such as autophagy, angiogenesis, adipogenesis, fibrosis, tumor growth, metastasis and wound healing. We review here the proteases which generate bioactive ECM fragments, their ECM substrates, the major bioactive ECM fragments, together with their biological properties and their receptors. The translation of ECM fragments into drugs is challenging and would take advantage of an integrative approach to optimize the design of pre-clinical and clinical studies. This could be done by building the contextualized interaction network of the ECM fragment repertoire including their parent proteins, remodeling proteinases, and their receptors, and by using mathematical disease models.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, F-69622 Villeurbanne cedex, France.
| | - Sylvain D Vallet
- Univ Lyon, University Claude Bernard Lyon 1, CNRS, INSA Lyon, CPE, Institute of Molecular and Supramolecular Chemistry and Biochemistry, UMR 5246, F-69622 Villeurbanne cedex, France.
| |
Collapse
|
65
|
McNally R, Alqudah A, Obradovic D, McClements L. Elucidating the Pathogenesis of Pre-eclampsia Using In Vitro Models of Spiral Uterine Artery Remodelling. Curr Hypertens Rep 2017; 19:93. [PMID: 29063290 PMCID: PMC5653699 DOI: 10.1007/s11906-017-0786-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
PURPOSE OF REVIEW The aim of the study is to perform a critical assessment of in vitro models of pre-eclampsia using complementary human and cell line-based studies. Molecular mechanisms involved in spiral uterine artery (SUA) remodelling and trophoblast functionality will also be discussed. RECENT FINDINGS A number of proteins and microRNAs have been implicated as key in SUA remodelling, which could be explored as early biomarkers or therapeutic targets for prevention of pre-eclampsia. Various 2D and 3D in vitro models involving trophoblast cells, endothelial cells, immune cells and placental tissue were discussed to elucidate the pathogenesis of pre-eclampsia. Nevertheless, pre-eclampsia is a multifactorial disease, and the mechanisms involved in its pathogenesis are complex and still largely unknown. Further studies are required to provide better understanding of the key processes leading to inappropriate placental development which is the root cause of pre-eclampsia. This new knowledge could identify novel biomarkers and treatment strategies.
Collapse
Affiliation(s)
- Ross McNally
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Abdelrahim Alqudah
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Danilo Obradovic
- Institute of Pathology, University of Belgrade, Belgrade, 11,000, Serbia
| | - Lana McClements
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
66
|
Kobayashi K, Jokaji R, Miyazawa-Hira M, Takatsuka S, Tanaka A, Ooi K, Nakamura H, Kawashiri S. Elastin‑derived peptides are involved in the processes of human temporomandibular disorder by inducing inflammatory responses in synovial cells. Mol Med Rep 2017; 16:3147-3154. [PMID: 28714016 PMCID: PMC5548023 DOI: 10.3892/mmr.2017.7012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 05/30/2017] [Indexed: 11/06/2022] Open
Abstract
Temporomandibular joint dysfunction (TMD) is a collection of clinical symptoms that involve masticatory muscles and the temporomandibular joint (TMJ). Common symptoms include limited jaw motion and joint sound/pain, along with TMJ disc displacement. TMD is frequently associated with synovitis, a chronic inflammation of the synovium. Fibroblast-like synovial cells have been identified to produce several inflammatory mediators and may have an important role in the progression of TMJ inflammation. Degradation of the extracellular matrix molecule elastin may lead to the release of bioactive peptides. The present study aimed to explore the role of elastin-derived peptides (EDPs) in human temporomandibular disorders. Therefore, interleukin-6 (IL-6) expression in the synovial fluid obtained from patients with TMD correlated significantly with two clinical parameters, specifically TMJ locking and pain/jaw function on a visual analog scale (VAS). To the best of our knowledge, this is the first study to determine that the concentration of EDPs in synovial fluid from patients with TMD may also be significantly correlated with the duration of TMJ locking, the VAS score and IL-6 expression. In vitro, EDPs act on human TMJ synovial cells to promote upregulation of IL-6 and the elastin-degrading enzyme matrix metalloproteinase-12 (MMP-12). The upregulation of IL-6 and MMP-12 expression by EDPs may be mediated through elastin-binding proteins (EBP) and a protein kinase A signalling cascade. These findings suggest a model for inflammation in the TMJ where EDPs are generated by harmful mechanical stimuli, induce both a pro-inflammatory cascade and increase expression of MMP-12 through activation of the EBP signalling cascade. This may lead to further increases in EDP levels, establishing a positive feedback loop leading to chronic inflammation in the TMJ. Therefore, significantly elevated levels of EDPs and IL-6 in the synovial fluid of the TMJ may be indicators of the pathological conditions of the joint.
Collapse
Affiliation(s)
- Kazuhiko Kobayashi
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa 920‑8640, Japan
| | - Rei Jokaji
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa 920‑8640, Japan
| | - Mayuko Miyazawa-Hira
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa 920‑8640, Japan
| | - Shigeyuki Takatsuka
- Department of Oral and Maxillofacial Surgery, Public Central Hospital of Matto Ishikawa, Hakusan, Ishikawa 924-8588, Japan
| | - Akira Tanaka
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa 920‑8640, Japan
| | - Kazuhiro Ooi
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa 920‑8640, Japan
| | - Hiroyuki Nakamura
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa 920‑8640, Japan
| | - Shuichi Kawashiri
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa 920‑8640, Japan
| |
Collapse
|
67
|
Abstract
A characteristic feature of liver cirrhosis is the accumulation of large amounts of connective tissue with the prevailing content of type I collagen. Elastin is a minor connective tissue component in normal liver but it is actively synthesized by hepatic stellate cells and portal fibroblasts in diseased liver. The accumulation of elastic fibers in later stages of liver fibrosis may contribute to the decreasing reversibility of the disease with advancing time. Elastin is formed by polymerization of tropoelastin monomers. It is an amorphous protein highly resistant to the action of proteases that forms the core of elastic fibers. Microfibrils surrounding the core are composed of fibrillins that bind a number of proteins involved in fiber formation. They include microfibril-associated glycoproteins (MAGPs), microfibrillar-associated proteins (MFAPs) and fibulins. Lysyl oxidase (LOX) and lysyl oxidase-like proteins (LOXLs) are responsible for tropoelastin cross-linking and polymerization. TGF-β complexes attached to microfibrils release this cytokine and influence the behavior of the cells in the neighborhood. The role of TGF-β as the main profibrotic cytokine in the liver is well-known and the release of the cytokines of TGF-β superfamily from their storage in elastic fibers may affect the course of fibrosis. Elastic fibers are often studied in the tissues where they provide elasticity and resilience but their role is no longer viewed as purely mechanical. Tropoelastin, elastin polymer and elastin peptides resulting from partial elastin degradation influence fibroblastic and inflammatory cells as well as angiogenesis. A similar role may be performed by elastin in the liver. This article reviews the results of the research of liver elastic fibers on the background of the present knowledge of elastin biochemistry and physiology. The regulation of liver elastin synthesis and degradation may be important for the outcome of liver fibrosis.
Collapse
Affiliation(s)
- Jiří Kanta
- Department of Medical Biochemistry, Faculty of Medicine in Hradec Kralove, Charles University in Prague Hradec Kralove, Czechia
| |
Collapse
|
68
|
Sheets AR, Massey CJ, Cronk SM, Iafrati MD, Herman IM. Matrix- and plasma-derived peptides promote tissue-specific injury responses and wound healing in diabetic swine. J Transl Med 2016; 14:197. [PMID: 27369317 PMCID: PMC4930589 DOI: 10.1186/s12967-016-0946-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 06/17/2016] [Indexed: 12/21/2022] Open
Abstract
Background Non-healing wounds are a major global health concern and account for the majority of non-traumatic limb amputations worldwide. However, compared to standard care practices, few advanced therapeutics effectively resolve these injuries stemming from cardiovascular disease, aging, and diabetes-related vasculopathies. While matrix turnover is disrupted in these injuries, debriding enzymes may promote healing by releasing matrix fragments that induce cell migration, proliferation, and morphogenesis, and plasma products may also stimulate these processes. Thus, we created matrix- and plasma-derived peptides, Comb1 and UN3, which induce cellular injury responses in vitro, and accelerate healing in rodent models of non-healing wounds. However, the effects of these peptides in non-healing wounds in diabetes are not known. Here, we interrogated whether these peptides stimulate healing in a diabetic porcine model highly reminiscent of human healing impairments in type 1 and type 2-diabetes. Methods After 3–6 weeks of streptozotocin-induced diabetes, full-thickness wounds were surgically created on the backs of adult female Yorkshire swine under general anesthesia. Comb1 and UN3 peptides or sterile saline (negative control) were administered to wounds daily for 3–7 days. Following sacrifice, wound tissues were harvested, and quantitative histological and immunohistochemical analyses were performed for wound closure, angiogenesis and granulation tissue deposition, along with quantitative molecular analyses of factors critical for angiogenesis, epithelialization, and dermal matrix remodeling. Results Comb1 and UN3 significantly increase re-epithelialization and angiogenesis in diabetic porcine wounds, compared to saline-treated controls. Additionally, fluorescein-conjugated Comb1 labels keratinocytes, fibroblasts, and vascular endothelial cells in porcine wounds, and Far western blotting reveals these cell populations express multiple fluorescein-Comb1-interacting proteins in vitro. Further, peptide treatment increases mRNA expression of several pro-angiogenic, epithelializing, and matrix-remodeling factors, importantly including balanced inductions in matrix metalloproteinase-2, -9, and tissue inhibitor of metalloproteinases-1, lending further insight into their mechanisms. Conclusions Comb1 and UN3 stimulate wound resolution in diabetic Yorkshire swine through upregulation of multiple reparative growth factors and cytokines, especially matrix metalloproteinases and inhibitors that may aid in reversing the proteolytic imbalance characteristic of chronically inflamed non-healing wounds. Together, these peptides should have great therapeutic potential for all patients in need of healing, regardless of injury etiology.
Collapse
Affiliation(s)
- Anthony R Sheets
- Graduate Program in Cellular & Molecular Physiology, The Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, 02111, USA.,Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA.,The Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Conner J Massey
- The Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Stephen M Cronk
- The Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA
| | - Mark D Iafrati
- The Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA.,Department of Surgery, Division of Vascular Surgery, Tufts Medical Center, 800 Washington St., Boston, MA, 02111, USA
| | - Ira M Herman
- Graduate Program in Cellular & Molecular Physiology, The Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, 02111, USA. .,Graduate Program in Cell, Molecular and Developmental Biology, The Sackler School of Graduate Biomedical Sciences, Tufts University, Boston, MA, 02111, USA. .,Department of Developmental, Molecular and Chemical Biology, School of Medicine, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA. .,The Center for Innovations in Wound Healing Research, School of Medicine, Tufts University, 136 Harrison Ave, Boston, MA, 02111, USA.
| |
Collapse
|
69
|
Heinz A, Huertas ACM, Schräder CU, Pankau R, Gosch A, Schmelzer CEH. Elastins from patients with Williams-Beuren syndrome and healthy individuals differ on the molecular level. Am J Med Genet A 2016; 170:1832-42. [DOI: 10.1002/ajmg.a.37638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 03/10/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Andrea Heinz
- Faculty of Natural Sciences I, Institute of Pharmacy; Martin Luther University Halle-Wittenberg; Halle (Saale) Germany
| | - Angela C. Mora Huertas
- Faculty of Natural Sciences I, Institute of Pharmacy; Martin Luther University Halle-Wittenberg; Halle (Saale) Germany
| | - Christoph U. Schräder
- Faculty of Natural Sciences I, Institute of Pharmacy; Martin Luther University Halle-Wittenberg; Halle (Saale) Germany
| | - Rainer Pankau
- Finkelstein-Klinik für Kinder-und Jugendmedizin; Heidekreis-Klinikum; Walsrode Germany
| | - Angela Gosch
- Fakultät für angewandte Sozialwissenschaften FK 11; Hochschule München; München Germany
| | - Christian E. H. Schmelzer
- Faculty of Natural Sciences I, Institute of Pharmacy; Martin Luther University Halle-Wittenberg; Halle (Saale) Germany
| |
Collapse
|
70
|
Gümbel D, Ackerl M, Napp M, Daeschlein G, Spranger N, Stope MB, Ekkernkamp A, Matthes G. Retrospektive Analyse von56 Weichteildefekten nach einzeitiger Rekonstruktion unter Verwendung von Dermisersatzpräparaten. J Dtsch Dermatol Ges 2016; 14:595-602. [PMID: 27240065 DOI: 10.1111/ddg.12874_g] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
HINTERGRUND Der Verschluss von Wunden mit ausgeprägtem Weichteilschaden stellt eine chirurgische Herausforderung dar und erfordert häufig umfangreiche plastische Operationen sowie freie Lappenplastiken. Die Kombination von Dermisersatzpräparaten und Spalthauttransplantationen ist eine innovative Methode die zur Versorgung von komplexen Verletzungen der Extremitäten angewandt werden kann. Wir haben diese Technik in das Standard-Handwerkszeug bei komplexen Verletzungen der Extremitäten aufgenommen. Die klinischen Ergebnisse von 56 behandelten Patienten werden vorgestellt. PATIENTEN UND METHODEN In 44 Fällen (78,6 %) wurde die beschriebene Methode an Defekten der unteren Extremitäten verwendet, einschließlich sieben Personen (12,5 %), die sich einer Stumpfdeckung nach Amputation unterzogen. Zwölf Defekte (21,4 %) befanden sich an den oberen Extremitäten. In zwei Fällen (3,6 %) wurde die Matriderm(®) -Matrix verwendet, um Nerven von unmittelbar angrenzenden chirurgischen Implantaten zu schützen. ERGEBNISSE Bei 41 Patienten (73,2 %) kam es zur Einheilung des Transplantats ohne Komplikationen. Fünfzehn Patienten (26,8 %) zeigten eine gestörte Wundheilung nach Defektverschluss, die unter konservativer Therapie zur Ausheilung gebracht werden konnte. Ein Patient (1,8 %) zeigte ein Transplantatversagen, was eine Revisionsoperation erforderlich machte. Umfangreiche plastische Rekonstruktionen mussten bei keinem Patienten angewandt werden. SCHLUSSFOLGERUNGEN Bei Fällen, in denen ausgedehnte plastische Operationen nicht möglich oder nicht erwünscht sind, ist die Verwendung von Dermisersatzpräparaten in Kombination mit Spalthauttransplantationen eine vielversprechende Alternative zum Wundverschluss bei ausgedehnten Weichteilschäden.
Collapse
Affiliation(s)
- Denis Gümbel
- Zentrum für Orthopädie, Unfallchirurgie und Rehabilitative Medizin, Klinik und Poliklinik für Unfall-, Wiederherstellungschirurgie und Rehabilitative Medizin, Universitätsmedizin, Greifswald.,Klinik für Unfallchirurgie und Orthopädie, BG Klinikum Unfallkrankenhaus Berlin gGmbH, Berlin
| | - Martin Ackerl
- Klinik für Unfallchirurgie und Orthopädie, BG Klinikum Unfallkrankenhaus Berlin gGmbH, Berlin
| | - Matthias Napp
- Zentrum für Orthopädie, Unfallchirurgie und Rehabilitative Medizin, Klinik und Poliklinik für Unfall-, Wiederherstellungschirurgie und Rehabilitative Medizin, Universitätsmedizin, Greifswald
| | - Georg Daeschlein
- Klinik und Poliklinik für Hautkrankheiten, Universitätsmedizin, Greifswald
| | - Nikolai Spranger
- Klinik für Unfallchirurgie und Orthopädie, BG Klinikum Unfallkrankenhaus Berlin gGmbH, Berlin
| | - Matthias B Stope
- Klinik und Poliklinik für Urologie, Urologisches Forschungslabor, Universitätsmedizin, Greifswald
| | - Axel Ekkernkamp
- Zentrum für Orthopädie, Unfallchirurgie und Rehabilitative Medizin, Klinik und Poliklinik für Unfall-, Wiederherstellungschirurgie und Rehabilitative Medizin, Universitätsmedizin, Greifswald.,Klinik für Unfallchirurgie und Orthopädie, BG Klinikum Unfallkrankenhaus Berlin gGmbH, Berlin
| | - Gerrit Matthes
- Zentrum für Orthopädie, Unfallchirurgie und Rehabilitative Medizin, Klinik und Poliklinik für Unfall-, Wiederherstellungschirurgie und Rehabilitative Medizin, Universitätsmedizin, Greifswald.,Klinik für Unfallchirurgie und Orthopädie, BG Klinikum Unfallkrankenhaus Berlin gGmbH, Berlin
| |
Collapse
|
71
|
Gümbel D, Ackerl M, Napp M, Daeschlein G, Spranger N, Stope MB, Ekkernkamp A, Matthes G. Retrospective analysis of 56 soft tissue defects treated with one-stage reconstruction using dermal skin substitutes. J Dtsch Dermatol Ges 2016; 14:595-601. [DOI: 10.1111/ddg.12874] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Denis Gümbel
- Department of Trauma and Reconstructive Surgery; University Medicine Greifswald; Greifswald Germany
- Department of Trauma and Orthopedic Surgery; Unfallkrankenhaus Berlin; Berlin Germany
| | - Martin Ackerl
- Department of Trauma and Orthopedic Surgery; Unfallkrankenhaus Berlin; Berlin Germany
| | - Matthias Napp
- Department of Trauma and Reconstructive Surgery; University Medicine Greifswald; Greifswald Germany
| | - Georg Daeschlein
- Department of Dermatology; University Medicine Greifswald; Greifswald Germany
| | - Nikolai Spranger
- Department of Trauma and Orthopedic Surgery; Unfallkrankenhaus Berlin; Berlin Germany
| | - Matthias B. Stope
- Department of Urology; Research Laboratory; University Medicine Greifswald; Greifswald Germany
| | - Axel Ekkernkamp
- Department of Trauma and Reconstructive Surgery; University Medicine Greifswald; Greifswald Germany
- Department of Trauma and Orthopedic Surgery; Unfallkrankenhaus Berlin; Berlin Germany
| | - Gerrit Matthes
- Department of Trauma and Reconstructive Surgery; University Medicine Greifswald; Greifswald Germany
- Department of Trauma and Orthopedic Surgery; Unfallkrankenhaus Berlin; Berlin Germany
| |
Collapse
|
72
|
Ferraro V, Anton M, Santé-Lhoutellier V. The “sisters” α-helices of collagen, elastin and keratin recovered from animal by-products: Functionality, bioactivity and trends of application. Trends Food Sci Technol 2016. [DOI: 10.1016/j.tifs.2016.03.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
73
|
Scandolera A, Odoul L, Salesse S, Guillot A, Blaise S, Kawecki C, Maurice P, El Btaouri H, Romier-Crouzet B, Martiny L, Debelle L, Duca L. The Elastin Receptor Complex: A Unique Matricellular Receptor with High Anti-tumoral Potential. Front Pharmacol 2016; 7:32. [PMID: 26973522 PMCID: PMC4777733 DOI: 10.3389/fphar.2016.00032] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/03/2016] [Indexed: 12/29/2022] Open
Abstract
Elastin, one of the longest-lived proteins, confers elasticity to tissues with high mechanical constraints. During aging or pathophysiological conditions such as cancer progression, this insoluble polymer of tropoelastin undergoes an important degradation leading to the release of bioactive elastin-derived peptides (EDPs), named elastokines. EDP exhibit several biological functions able to drive tumor development by regulating cell proliferation, invasion, survival, angiogenesis, and matrix metalloproteinase expression in various tumor and stromal cells. Although, several receptors have been suggested to bind elastokines (αvβ3 and αvβ5 integrins, galectin-3), their main receptor remains the elastin receptor complex (ERC). This heterotrimer comprises a peripheral subunit, named elastin binding protein (EBP), associated to the protective protein/cathepsin A (PPCA). The latter is bound to a membrane-associated protein called Neuraminidase-1 (Neu-1). The pro-tumoral effects of elastokines have been linked to their binding onto EBP. Additionally, Neu-1 sialidase activity is essential for their signal transduction. Consistently, EDP-EBP interaction and Neu-1 activity emerge as original anti-tumoral targets. Interestingly, besides its direct involvement in cancer progression, the ERC also regulates diabetes outcome and thrombosis, an important risk factor for cancer development and a vascular process highly increased in patients suffering from cancer. In this review, we will describe ERC and elastokines involvement in cancer development suggesting that this unique receptor would be a promising therapeutic target. We will also discuss the pharmacological concepts aiming at blocking its pro-tumoral activities. Finally, its emerging role in cancer-associated complications and pathologies such as diabetes and thrombotic events will be also considered.
Collapse
Affiliation(s)
- Amandine Scandolera
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Ludivine Odoul
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Stéphanie Salesse
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Alexandre Guillot
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Sébastien Blaise
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Charlotte Kawecki
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Pascal Maurice
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Hassan El Btaouri
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Béatrice Romier-Crouzet
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Laurent Martiny
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Laurent Debelle
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| | - Laurent Duca
- UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences Reims, France
| |
Collapse
|
74
|
Ricard-Blum S, Vallet SD. Matricryptins Network with Matricellular Receptors at the Surface of Endothelial and Tumor Cells. Front Pharmacol 2016; 7:11. [PMID: 26869928 PMCID: PMC4740388 DOI: 10.3389/fphar.2016.00011] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/12/2016] [Indexed: 12/11/2022] Open
Abstract
The extracellular matrix (ECM) is a source of bioactive fragments called matricryptins or matrikines resulting from the proteolytic cleavage of extracellular proteins (e.g., collagens, elastin, and laminins) and proteoglycans (e.g., perlecan). Matrix metalloproteinases (MMPs), cathepsins, and bone-morphogenetic protein-1 release fragments, which regulate physiopathological processes including tumor growth, metastasis, and angiogenesis, a pre-requisite for tumor growth. A number of matricryptins, and/or synthetic peptides derived from them, are currently investigated as potential anti-cancer drugs both in vitro and in animal models. Modifications aiming at improving their efficiency and their delivery to their target cells are studied. However, their use as drugs is not straightforward. The biological activities of these fragments are mediated by several receptor families. Several matricryptins may bind to the same matricellular receptor, and a single matricryptin may bind to two different receptors belonging or not to the same family such as integrins and growth factor receptors. Furthermore, some matricryptins interact with each other, integrins and growth factor receptors crosstalk and a signaling pathway may be regulated by several matricryptins. This forms an intricate 3D interaction network at the surface of tumor and endothelial cells, which is tightly associated with other cell-surface associated molecules such as heparan sulfate, caveolin, and nucleolin. Deciphering the molecular mechanisms underlying the behavior of this network is required in order to optimize the development of matricryptins as anti-cancer agents.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- University Claude Bernard Lyon 1, UMR 5246 Centre National de la Recherche Scientifique - University Lyon 1 - Institut National des Sciences Appliquées de Lyon - École Supérieure de Chimie Physique Électronique de Lyon Villeurbanne, France
| | - Sylvain D Vallet
- University Claude Bernard Lyon 1, UMR 5246 Centre National de la Recherche Scientifique - University Lyon 1 - Institut National des Sciences Appliquées de Lyon - École Supérieure de Chimie Physique Électronique de Lyon Villeurbanne, France
| |
Collapse
|
75
|
|
76
|
Ricard-Blum S, Vallet SD. Proteases decode the extracellular matrix cryptome. Biochimie 2015; 122:300-13. [PMID: 26382969 DOI: 10.1016/j.biochi.2015.09.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 09/11/2015] [Indexed: 12/24/2022]
Abstract
The extracellular matrix is comprised of 1100 core-matrisome and matrisome-associated proteins and of glycosaminoglycans. This structural scaffold contributes to the organization and mechanical properties of tissues and modulates cell behavior. The extracellular matrix is dynamic and undergoes constant remodeling, which leads to diseases if uncontrolled. Bioactive fragments, called matricryptins, are released from the extracellular proteins by limited proteolysis and have biological activities on their own. They regulate numerous physiological and pathological processes such as angiogenesis, cancer, diabetes, wound healing, fibrosis and infectious diseases and either improve or worsen the course of diseases depending on the matricryptins and on the molecular and biological contexts. Several protease families release matricryptins from core-matrisome and matrisome-associated proteins both in vitro and in vivo. The major proteases, which decrypt the extracellular matrix, are zinc metalloproteinases of the metzincin superfamily (matrixins, adamalysins and astacins), cysteine proteinases and serine proteases. Some matricryptins act as enzyme inhibitors, further connecting protease and matricryptin fates and providing intricate regulation of major physiopathological processes such as angiogenesis and tumorigenesis. They strengthen the role of the extracellular matrix as a key player in tissue failure and core-matrisome and matrisome-associated proteins as important therapeutic targets.
Collapse
Affiliation(s)
- Sylvie Ricard-Blum
- UMR 5086 CNRS - Université Lyon 1, 7 Passage du Vercors, 69367 Lyon Cedex 07, France.
| | - Sylvain D Vallet
- UMR 5086 CNRS - Université Lyon 1, 7 Passage du Vercors, 69367 Lyon Cedex 07, France.
| |
Collapse
|
77
|
Yu Y, Wise SG, Michael PL, Bax DV, Yuen GSC, Hiob MA, Yeo GC, Filipe EC, Dunn LL, Chan KH, Hajian H, Celermajer DS, Weiss AS, Ng MKC. Characterization of Endothelial Progenitor Cell Interactions with Human Tropoelastin. PLoS One 2015; 10:e0131101. [PMID: 26115013 PMCID: PMC4482626 DOI: 10.1371/journal.pone.0131101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 05/28/2015] [Indexed: 01/23/2023] Open
Abstract
The deployment of endovascular implants such as stents in the treatment of cardiovascular disease damages the vascular endothelium, increasing the risk of thrombosis and promoting neointimal hyperplasia. The rapid restoration of a functional endothelium is known to reduce these complications. Circulating endothelial progenitor cells (EPCs) are increasingly recognized as important contributors to device re-endothelialization. Extracellular matrix proteins prominent in the vessel wall may enhance EPC-directed re-endothelialization. We examined attachment, spreading and proliferation on recombinant human tropoelastin (rhTE) and investigated the mechanism and site of interaction. EPCs attached and spread on rhTE in a dose dependent manner, reaching a maximal level of 56±3% and 54±3%, respectively. EPC proliferation on rhTE was comparable to vitronectin, fibronectin and collagen. EDTA, but not heparan sulfate or lactose, reduced EPC attachment by 81±3%, while full attachment was recovered after add-back of manganese, inferring a classical integrin-mediated interaction. Integrin αVβ3 blocking antibodies decreased EPC adhesion and spreading on rhTE by 39±3% and 56±10% respectively, demonstrating a large contribution from this specific integrin. Attachment of EPCs on N-terminal rhTE constructs N25 and N18 accounted for most of this interaction, accompanied by comparable spreading. In contrast, attachment and spreading on N10 was negligible. αVβ3 blocking antibodies reduced EPC spreading on both N25 and N18 by 45±4% and 42±14%, respectively. In conclusion, rhTE supports EPC binding via an integrin mechanism involving αVβ3. N25 and N18, but not N10 constructs of rhTE contribute to EPC binding. The regulation of EPC activity by rhTE may have implications for modulation of the vascular biocompatibility of endovascular implants.
Collapse
Affiliation(s)
- Young Yu
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, 2050, Australia
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Steven G. Wise
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia
- * E-mail:
| | - Praveesuda L. Michael
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Daniel V. Bax
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia
| | - Gloria S. C. Yuen
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Matti A. Hiob
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia
| | - Giselle C. Yeo
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia
| | - Elysse C. Filipe
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Louise L. Dunn
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Kim H. Chan
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, 2050, Australia
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Hamid Hajian
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - David S. Celermajer
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, 2050, Australia
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| | - Anthony S. Weiss
- School of Molecular Bioscience, University of Sydney, Sydney, NSW, 2006, Australia
- Bosch Institute, University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, University of Sydney, Sydney, NSW, 2006, Australia
| | - Martin K. C. Ng
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, NSW, 2050, Australia
- The Heart Research Institute, Sydney, NSW, 2042, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
78
|
Scandolera A, Rabenoelina F, Chaintreuil C, Rusciani A, Maurice P, Blaise S, Romier-Crouzet B, El Btaouri H, Martiny L, Debelle L, Duca L. Uncoupling of Elastin Complex Receptor during In Vitro Aging Is Related to Modifications in Its Intrinsic Sialidase Activity and the Subsequent Lactosylceramide Production. PLoS One 2015; 10:e0129994. [PMID: 26086247 PMCID: PMC4473072 DOI: 10.1371/journal.pone.0129994] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/15/2015] [Indexed: 12/16/2022] Open
Abstract
Degradation of elastin leads to the production of elastin-derived peptides (EDP), which exhibit several biological effects, such as cell proliferation or protease secretion. Binding of EDP on the elastin receptor complex (ERC) triggers lactosylceramide (LacCer) production and ERK1/2 activation following ERC Neu-1 subunit activation. The ability for ERC to transduce signals is lost during aging, but the mechanism involved is still unknown. In this study, we characterized an in vitro model of aging by subculturing human dermal fibroblasts. This model was used to understand the loss of EDP biological activities during aging. Our results show that ERC uncoupling does not rely on Neu-1 or PPCA mRNA or protein level changes. Furthermore, we observe that the membrane targeting of these subunits is not affected with aging. However, we evidence that Neu-1 activity and LacCer production are altered. Basal Neu-1 catalytic activity is strongly increased in aged cells. Consequently, EDP fail to promote Neu-1 catalytic activity and LacCer production in these cells. In conclusion, we propose, for the first time, an explanation for ERC uncoupling based on the age-related alterations of Neu-1 activity and LacCer production that may explain the loss of EDP-mediated effects occurring during aging.
Collapse
Affiliation(s)
- Amandine Scandolera
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
| | - Fanja Rabenoelina
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
| | - Carine Chaintreuil
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
| | - Anthony Rusciani
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
| | - Pascal Maurice
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
| | - Sébastien Blaise
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
| | - Béatrice Romier-Crouzet
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
| | - Hassan El Btaouri
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
| | - Laurent Martiny
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
| | - Laurent Debelle
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
| | - Laurent Duca
- Laboratoire Signalisation et Récepteurs Matriciels (SiRMa), UMR CNRS/URCA 7369, SFR CAP Santé, Université de Reims Champagne Ardenne, Faculté des Sciences, Reims, France
- * E-mail:
| |
Collapse
|
79
|
Qin Z. Soluble elastin peptides in cardiovascular homeostasis: Foe or ally. Peptides 2015; 67:64-73. [PMID: 25794852 DOI: 10.1016/j.peptides.2015.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 03/04/2015] [Accepted: 03/09/2015] [Indexed: 12/23/2022]
Abstract
Elastin peptides, also known as elastin-derived peptides or elastokines, are soluble polypeptides in blood and tissue. The blood levels of elastin peptides are usually low but can increase during cardiovascular diseases, such as atherosclerosis, aortic aneurysm and diabetes with vascular complications. Generally, elastin peptides are derived from the degradation of insoluble elastic polymers. The biological activities of elastin peptides are bidirectional, e.g., a pro-inflammatory effect on monocyte migration induction vs. a protective effect on vasodilation promotion. However, recent in vivo studies have demonstrated that elastin peptides promote the formation of atherosclerotic plaques in hypercholesterolemic mice and induce hyperglycemia and elevations in plasma lipid levels in fasted mice. More important, the detrimental effects induced by elastin peptides can be largely inhibited by genetic or pharmacological blockade of the elastin receptor complex or by neutralization of an antibody against elastin peptides. These studies indicate new therapeutic strategies for the treatment of cardiovascular diseases by targeting elastin peptide metabolism. Therefore, the goal of this review is to summarize current knowledge about elastin peptides relevant to cardiovascular pathologies to further delineate their potential application in cardiovascular disease.
Collapse
Affiliation(s)
- Zhenyu Qin
- Division of Vascular Surgery, Department of Surgery, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States.
| |
Collapse
|
80
|
Effect of elastin-derived peptides on the production of tissue inhibitor of metalloproteinase-1, -2, and -3 and the ratios in various endothelial cell lines. Exp Ther Med 2015; 9:2245-2250. [PMID: 26136968 DOI: 10.3892/etm.2015.2429] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 02/05/2015] [Indexed: 12/25/2022] Open
Abstract
Tissue inhibitors of metalloproteinases (TIMPs) control the activity of metalloproteinases. Elastin-derived peptides (EDPs) are generated as a result of the degradation of elastin fibers. The EDPs bind to the elastin receptor and exert numerous biological effects. The aim of the present study was to compare the production of TIMP-1, TIMP-2 and TIMP-3 and their ratios in human endothelial cells (ECs) derived from three clinically important vascular localizations (coronary arteries, aorta and iliac artery), and evaluate the influence of a well-known EDP, κ-elastin. The highest concentration of TIMP-1 was identified in the aortic ECs, while the lowest concentration was observed in the ECs derived from the coronary artery. The opposite pattern was observed for TIMP-2 production. When the TIMP-3 concentration was analyzed in the three EC lines, no statistically significant differences were observed. Application of κ-elastin was found to decrease the TIMP-1 concentration in the aortic ECs, while an increase in the TIMP-1 concentration was observed in the ECs derived from the iliac artery. The most significant decrease in TIMP-2 concentration following κ-elastin administration was observed in the ECs obtained from the coronary arteries. Furthermore, the highest concentration of κ-elastin resulted in an increase in TIMP-3 production in the ECs derived from the coronary arteries. The following ratios of the TIMP concentrations were calculated: TIMP-1/TIMP-2, TIMP-1/TIMP-3 and TIMP-2/TIMP-3. Each ratio presented different values for the ECs obtained from the various localizations. In the majority of cases, the addition of κ-elastin did not significantly change these proportions. Therefore, these indicators may be characteristic features that can be used to describe ECs in various clinically important vascular localizations.
Collapse
|
81
|
Munjal C, Opoka AM, Osinska H, James JF, Bressan GM, Hinton RB. TGF-β mediates early angiogenesis and latent fibrosis in an Emilin1-deficient mouse model of aortic valve disease. Dis Model Mech 2015; 7:987-96. [PMID: 25056700 PMCID: PMC4107327 DOI: 10.1242/dmm.015255] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Aortic valve disease (AVD) is characterized by elastic fiber fragmentation (EFF), fibrosis and aberrant angiogenesis. Emilin1 is an elastin-binding glycoprotein that regulates elastogenesis and inhibits TGF-β signaling, but the role of Emilin1 in valve tissue is unknown. We tested the hypothesis that Emilin1 deficiency results in AVD, mediated by non-canonical (MAPK/phosphorylated Erk1 and Erk2) TGF-β dysregulation. Using histology, immunohistochemistry, electron microscopy, quantitative gene expression analysis, immunoblotting and echocardiography, we examined the effects of Emilin1 deficiency (Emilin1−/−) in mouse aortic valve tissue. Emilin1 deficiency results in early postnatal cell-matrix defects in aortic valve tissue, including EFF, that progress to latent AVD and premature death. The Emilin1−/− aortic valve displays early aberrant provisional angiogenesis and late neovascularization. In addition, Emilin1−/− aortic valves are characterized by early valve interstitial cell activation and proliferation and late myofibroblast-like cell activation and fibrosis. Interestingly, canonical TGF-β signaling (phosphorylated Smad2 and Smad3) is upregulated constitutively from birth to senescence, whereas non-canonical TGF-β signaling (phosphorylated Erk1 and Erk2) progressively increases over time. Emilin1 deficiency recapitulates human fibrotic AVD, and advanced disease is mediated by non-canonical (MAPK/phosphorylated Erk1 and Erk2) TGF-β activation. The early manifestation of EFF and aberrant angiogenesis suggests that these processes are crucial intermediate factors involved in disease progression and therefore might provide new therapeutic targets for human AVD.
Collapse
Affiliation(s)
- Charu Munjal
- Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Amy M Opoka
- Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hanna Osinska
- Division of Molecular Cardiovascular Biology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jeanne F James
- Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Giorgio M Bressan
- Department of Molecular Medicine, University of Padua, 35121 Padua, Italy
| | - Robert B Hinton
- Division of Cardiology, The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| |
Collapse
|
82
|
Wang Y, Mithieux SM, Kong Y, Wang XQ, Chong C, Fathi A, Dehghani F, Panas E, Kemnitzer J, Daniels R, Kimble RM, Maitz PK, Li Z, Weiss AS. Tropoelastin incorporation into a dermal regeneration template promotes wound angiogenesis. Adv Healthc Mater 2015; 4:577-84. [PMID: 25469903 DOI: 10.1002/adhm.201400571] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2014] [Revised: 11/05/2014] [Indexed: 11/06/2022]
Abstract
Severe burn injury results in substantial skin loss and cannot be treated by autografts. The Integra Dermal Regeneration Template (IDRT) is the leading synthetic skin substitute because it allows for wound bed regeneration and wound healing. However, all substitutes suffer from slow blood vessel ingrowth and would benefit considerably from enhanced vascularization to nurture tissue repair. It is shown here that by incorporating the human elastic protein tropoelastin into a dermal regeneration template (TDRT) we can promote angiogenesis in wound healing. In small and large animal models comprising mice and pigs, the hybrid TDRT biomaterial and IDRT show similar contraction to autografts and decrease wound contraction compared to open wounds. In mice, TDRT accelerates early stage angiogenesis by 2 weeks, as evidenced by increased angiogenesis fluorescent radiant efficiency in live animal imaging and the expression of endothelial cell adhesion marker CD146. In the pig, a full thickness wound repair model confirms increased numbers of blood vessels in the regenerating areas of the dermis closest to the hypodermis and immediately below the epidermis at 2 weeks post-surgery. It is concluded that including tropoelastin in a dermal regeneration template has the potential to promote wound repair through enhanced vascularization.
Collapse
Affiliation(s)
- Yiwei Wang
- Burns Research Group, ANZAC Research Institute; University of Sydney; Concord NSW 2139 Australia
| | - Suzanne M. Mithieux
- School of Molecular Bioscience; University of Sydney; NSW 2006 Australia
- Charles Perkins Centre; University of Sydney; NSW 2006 Australia
| | - Yvonne Kong
- School of Molecular Bioscience; University of Sydney; NSW 2006 Australia
| | - Xue-Qing Wang
- Centre for Children's Burns and Trauma Research, Queensland Children's Medical Research Institute; University of Queensland; Australia
- Stuart Pegg Paediatric Burns Centre; Royal Children's Hospital; Brisbane Australia
| | - Cassandra Chong
- Burns Research Group, ANZAC Research Institute; University of Sydney; Concord NSW 2139 Australia
| | - Ali Fathi
- School of Chemical and Biomolecular Engineering; University of Sydney; NSW 2006 Australia
| | - Fariba Dehghani
- School of Chemical and Biomolecular Engineering; University of Sydney; NSW 2006 Australia
| | | | | | | | - Roy M. Kimble
- Centre for Children's Burns and Trauma Research, Queensland Children's Medical Research Institute; University of Queensland; Australia
- Stuart Pegg Paediatric Burns Centre; Royal Children's Hospital; Brisbane Australia
| | - Peter K. Maitz
- Burns Research Group, ANZAC Research Institute; University of Sydney; Concord NSW 2139 Australia
- Burns and Reconstructive Surgery Unit; Concord Repatriation General Hospital; NSW 2139 Australia
| | - Zhe Li
- Burns Research Group, ANZAC Research Institute; University of Sydney; Concord NSW 2139 Australia
- Burns and Reconstructive Surgery Unit; Concord Repatriation General Hospital; NSW 2139 Australia
| | - Anthony S. Weiss
- School of Molecular Bioscience; University of Sydney; NSW 2006 Australia
- Charles Perkins Centre; University of Sydney; NSW 2006 Australia
- Bosch Institute; University of Sydney; NSW 2006 Australia
| |
Collapse
|
83
|
Over-the-counter anti-ageing topical agents and their ability to protect and repair photoaged skin. Maturitas 2015; 80:265-72. [DOI: 10.1016/j.maturitas.2014.12.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 12/30/2014] [Indexed: 01/20/2023]
|
84
|
Golda A, Jurecka A, Gajda K, Tylki-Szymańska A, Lalik A. Human pulmonary artery endothelial cells in the model of mucopolysaccharidosis VI present a prohypertensive phenotype. Mol Genet Metab Rep 2015; 3:11-7. [PMID: 26937388 PMCID: PMC4750576 DOI: 10.1016/j.ymgmr.2015.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 02/20/2015] [Accepted: 02/21/2015] [Indexed: 12/18/2022] Open
Abstract
Background Mucopolysaccharidosis type VI (MPS VI) is an autosomal recessive lysosomal disorder caused by a deficient activity of N-acetylgalactosamine-4-sulfatase (ARSB). Pulmonary hypertension (PH) occurs in MPS VI patients and is a marker of bad prognosis. Malfunction of endothelium, which regulates vascular tonus and stimulates angiogenesis, can contribute to the occurrence of PH in MPS VI. Aim The aim of the study was to establish a human MPS VI cellular model of pulmonary artery endothelial cells (HPAECs) and evaluate how it affects factors that may trigger PH such as proliferation, apoptosis, expression of endothelial nitric oxide synthase (eNOS), natriuretic peptide type C (NPPC), and vascular endothelial growth factor A (VEGFA). Results Increasing concentrations of dermatan sulfate (DS) reduce the viability of the cells in both ARSB deficiency and controls, but hardly influence apoptosis. The expression of eNOS in HPAECs is reduced up to two thirds in the presence of DS. NPPC shows a biphasic expression reaction with an increase at 50 μg/mL DS and reduction at 0 and 100 μg/mL DS. The expression of VEGFA decreases with increasing DS concentrations and absence of elastin, and increases with increasing DS in the presence of elastin. Conclusion Our data suggest that MPS VI endothelium presents a prohypertensive phenotype due to the reduction of endothelium's proliferation ability and expression of vasorelaxing factors.
Collapse
Affiliation(s)
- Adam Golda
- Department of Cardiology, Gliwice Medical Center, Gliwice, Poland
| | - Agnieszka Jurecka
- Department of Pediatrics, Nutrition and Metabolic Diseases, The Children's Memorial Health Institute, Warsaw, Poland
| | - Karolina Gajda
- Systems Engineering Group, Faculty of Automatic Control, Electronics and Informatics, Silesian University of Technology, Gliwice, Poland
| | - Anna Tylki-Szymańska
- Department of Pediatrics, Nutrition and Metabolic Diseases, The Children's Memorial Health Institute, Warsaw, Poland
| | - Anna Lalik
- Systems Engineering Group, Faculty of Automatic Control, Electronics and Informatics, Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
85
|
Gaffney J, Solomonov I, Zehorai E, Sagi I. Multilevel regulation of matrix metalloproteinases in tissue homeostasis indicates their molecular specificity in vivo. Matrix Biol 2015; 44-46:191-9. [PMID: 25622911 DOI: 10.1016/j.matbio.2015.01.012] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/18/2015] [Accepted: 01/18/2015] [Indexed: 11/16/2022]
Abstract
The matrix metalloproteinases (MMPs) play a crucial role in irreversible remodeling of the extracellular matrix (ECM) in normal homeostasis and pathological states. Accumulating data from various studies strongly suggest that MMPs are tightly regulated, starting from the level of gene expression all the way to zymogen activation and endogenous inhibition, with each level controlled by multiple factors. Recent in vivo findings indicate that cell-ECM and cell-cell interactions, as well as ECM bio-active products, contribute an additional layer of regulation at all levels, indicating that individual MMP expression and activity in vivo are highly coordinated and tissue specific processes.
Collapse
Affiliation(s)
- Jean Gaffney
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel; Department of Natural Sciences, Baruch College, New York, NY, USA
| | - Inna Solomonov
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Eldar Zehorai
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Irit Sagi
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
86
|
Azevedo A, Prado AF, Antonio RC, Issa JP, Gerlach RF. Matrix metalloproteinases are involved in cardiovascular diseases. Basic Clin Pharmacol Toxicol 2014; 115:301-14. [PMID: 24974977 DOI: 10.1111/bcpt.12282] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 06/26/2014] [Indexed: 12/18/2022]
Abstract
This MiniReview describes the essential biochemical and molecular aspects of matrix metalloproteinases (MMPs) and briefly discusses how they engage in different diseases, with particular emphasis on cardiovascular diseases. There is compelling scientific evidence that many MMPs, especially MMP-2, play important roles in the development of cardiovascular diseases; inhibition of these enzymes is beneficial to many cardiovascular conditions, sometimes precluding or postponing end-organ damage and fatal outcomes. Conducting comprehensive discussions and further studies on how MMPs participate in cardiovascular diseases is important, because inhibition of these enzymes may be an alternative or an adjuvant for current cardiovascular disease therapy.
Collapse
Affiliation(s)
- Aline Azevedo
- Department of Pharmacology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | | | | | | |
Collapse
|
87
|
Stahl PJ, Chan TR, Shen YI, Sun G, Gerecht S, Yu SM. Capillary Network-Like Organization of Endothelial Cells in PEGDA Scaffolds Encoded with Angiogenic Signals via Triple Helical Hybridization. ADVANCED FUNCTIONAL MATERIALS 2014; 24:3213-3225. [PMID: 25541582 PMCID: PMC4273917 DOI: 10.1002/adfm.201303217] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Survival of tissue engineered constructs after implantation depends on proper vascularization. The differentiation of endothelial cells into mature microvasculature requires dynamic interactions between cells, scaffold, and growth factors, which are difficult to recapitulate in artificial systems. Previously, photocrosslinked poly(ethylene glycol) diacrylate (PEGDA) hydrogels displaying collagen mimetic peptides (CMPs), dubbed PEGDA‐CMP, that can be further conjugated with bioactive molecules via CMP‐CMP triple helix hybridization were reported. Here, it is shown that a bifunctional peptide featuring pro‐angiogenic domain mimicking vascular endothelial growth factor (VEGF) and a collagen mimetic domain that can fold into a triple helix conformation can hybridize with CMP side chains of the PEGDA‐CMP hydrogel, which results in presentation of insoluble VEGF‐like signals to endothelial cells. Presentation of VEGF‐like signals on the surface of micropatterned scaffolds in this way transforms cells from a quiescent state to elongated and aligned phenotype suggesting that this system could be used to engineer organized microvasculature. It is also shown that the pro‐angiogenic peptide, when applied topically in combination with modified dextran/PEGDA hydrogels, can enhance neovascularization of burn wounds in mice demonstrating the potential clinical use of CMP‐mediated matrix‐bound bioactive molecules for dermal injuries.
Collapse
Affiliation(s)
- Patrick J Stahl
- Department of Materials Science and Engineering, Institute for NanoBioTechnology The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218 (USA)
| | - Tania R Chan
- Department of Materials Science and Engineering, Institute for NanoBioTechnology The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218 (USA)
| | - Yu-I Shen
- Department of Biomolecular and Chemical Engineering, Institute for NanoBioTechnology The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218 (USA)
| | - Guoming Sun
- Department of Biomolecular and Chemical Engineering, Institute for NanoBioTechnology The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218 (USA)
| | - Sharon Gerecht
- Department of Biomolecular and Chemical Engineering, Institute for NanoBioTechnology The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218 (USA)
| | - S Michael Yu
- Department of Bioengineering University of Utah 201 Presidents Circle, Salt Lake City, UT 84112 (USA) Institute for NanoBioTechnology The Johns Hopkins University, 3400 N. Charles St., Baltimore, MD 21218 (USA)
| |
Collapse
|
88
|
Elastin peptides regulate HT-1080 fibrosarcoma cell migration and invasion through an Hsp90-dependent mechanism. Br J Cancer 2014; 111:139-48. [PMID: 24874477 PMCID: PMC4090727 DOI: 10.1038/bjc.2014.239] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 04/04/2014] [Accepted: 04/10/2014] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The elastin-derived peptides (EDPs) exert protumoural activities by potentiating the secretion of matrix metalloproteinases (MMP) and the plasminogen-plasmin activating system. In the present paper, we studied heat-shock protein 90 (Hsp90) involvement in this mechanism. METHODS HT-1080 fibrosarcoma cell migration and invasion were studied in artificial wound assay and modified Boyden chamber assay, respectively. Heat-shock protein 90 was studied by western blot and immunofluorescence. Matrix metalloproteinase-2 and urokinase plasminogen activator (uPA) were studied by gelatin ± plasminogen zymography and immunofluorescence. Heat-shock protein 90 partners were studied by immunoprecipitation. Messenger RNA expression was studied using real-time PCR. Small interfering RNAs were used to confirm the essential role of Hsp90. RESULTS We showed that kappa-elastin and VGVAPG elastin hexapeptide stimulated Hsp90, pro-MMP-2 and uPA secretion within 6 h, whereas AGVPGLGVG and GRKRK peptides had no effect. No increase of mRNA level was observed. Heat-shock protein 90-specific inhibitors inhibit EDP-stimulated HT-1080 cell-invasive capacity and restrained EDP-stimulated pro-MMP-2 and uPA secretions. The inhibitory effect was reproduced by using Hsp90-blocking antibody or Hsp90 knockdown by siRNA. Heat-shock protein 90 interacted with and stabilised uPA and pro-MMP-2 in conditioned culture media of HT-1080 fibrosarcoma cells. CONCLUSIONS Taken together, our results demonstrate that EDPs exert protumoural activities through an Hsp90-dependent mechanism involving pro-MMP-2 and uPA.
Collapse
|
89
|
Liu S, Young SM, Varisco BM. Dynamic expression of chymotrypsin-like elastase 1 over the course of murine lung development. Am J Physiol Lung Cell Mol Physiol 2014; 306:L1104-16. [PMID: 24793170 DOI: 10.1152/ajplung.00126.2013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Postnatal lung development requires coordination of three processes (surface area expansion, microvascular growth, and matrix remodeling). Because normal elastin structure is important for lung morphogenesis, because physiological remodeling of lung elastin has never been defined, and because elastin remodeling is angiogenic, we sought to test the hypothesis that, during lung development, elastin is remodeled in a defined temporal-spatial pattern, that a novel protease is associated with this remodeling, and that angiogenesis is associated with elastin remodeling. By elastin in situ zymography, lung elastin remodeling increased 24-fold between embryonic day (E) 15.5 and postnatal day (PND) 14. Remodeling was restricted to major vessels and airways on PND1 with a sevenfold increase in alveolar wall elastin remodeling from PND1 to PND14. By inhibition assays and literature review, we identified chymotrypsin-like elastase 1 (CELA1) as a potential mediator of elastin remodeling. CELA1 mRNA levels increased 12-fold from E15.5 to PND9, and protein levels increased 3.4-fold from E18.5 to PND9. By costaining experiments, the temporal-spatial pattern of CELA1 expression matched that of elastin remodeling, and 58-85% of CELA1(+) cells were <10 μm from an elastase signal. An association between elastin remodeling and angiogenesis was tested by similar methods. At PND7 and PND14, 60-95% of angiogenin(+) cells were associated with elastin remodeling. Both elastase inhibition and CELA1 silencing impaired angiogenesis in vitro. Our data defines the temporal-spatial pattern of elastin remodeling during lung development, demonstrates an association of this remodeling with CELA1, and supports a role for elastin remodeling in regulating angiogenesis.
Collapse
Affiliation(s)
- Sheng Liu
- Division of Critical Care Medicine, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; and
| | - Sarah Marie Young
- Division of Critical Care Medicine, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; and
| | - Brian Michael Varisco
- Division of Critical Care Medicine, Cincinnati Children's Hospital Research Foundation, Cincinnati, Ohio; and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| |
Collapse
|
90
|
Du P, Subbiah R, Park JH, Park K. Vascular morphogenesis of human umbilical vein endothelial cells on cell-derived macromolecular matrix microenvironment. Tissue Eng Part A 2014; 20:2365-77. [PMID: 24517112 DOI: 10.1089/ten.tea.2013.0693] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Extracellular matrix (ECM) is a highly organized network of proteins and other macromolecules that plays a critical role in cell adhesion, migration, and differentiation. In this study, we hypothesize that ECM derived from in-vitro-cultured cells possesses unique surface texture, topography, and mechanical property, and consequently carries some distinct cues for vascular morphogenesis of human umbilical vein endothelial cells (ECs). Cell-derived matrix (CDM) was obtained by culturing fibroblasts, preosteoblasts, and chondrocytes, respectively, on coverslips and then by decellularizing them using detergents and enzymes. These matrices were named fibroblast-derived matrix (FDM), preosteoblast-derived matrix (PDM), and chondrocyte-derived matrix (CHDM). Immunofluorescence of each CDM shows that some of the matrix components are fibronectin (FN), type I collagen, and laminin. Atomic force microscopy analysis presented that average fiber diameter ranged from 2 to 7 μm and FDM holds much larger fibers. The matrix elasticity measurements revealed that average Young's modulus of CHDM (17.7 ± 4.2 kPa) was much greater than that of PDM (10.5 ± 1.1 kPa) or FDM (5.7 ± 0.5 kPa). During 5-day culture, EC morphologies were dramatically changed on PDM and FDM, but those on CHDM and gelatin were rather stable, regardless of time lapse. Cell migration assay discovered quicker repopulation of the scratched areas on PDM and FDM than on gelatin and CHDM. A capillary-like structure (CLS) assembly was also notable only in the PDM and FDM, as compared with CHDM, gelatin, or FN that were very poor in CLS formation. Quantitative analysis of mean CLS branch points and branch lengths demonstrated much better angiogenic activity of ECs on PDM and FDM. Interestingly, CLS formation was closely associated with matrix remodeling by ECs and the matrix clearance on PDM with time was sharply contrasted with that on CHDM that majority of the matrix FN was reserved. It was notable that membrane type 1-matrix metalloprotease was deeply involved in the process of matrix remodeling. This study indicates that specific matrix microenvironments are very critical for vascular morphogenesis of ECs, and thus, provide a nice platform for angiogenesis study as well as vascular tissue engineering.
Collapse
Affiliation(s)
- Ping Du
- 1 Center for Biomaterials, Korea Institute of Science and Technology , Seoul, Republic of Korea
| | | | | | | |
Collapse
|
91
|
Khavandgar Z, Roman H, Li J, Lee S, Vali H, Brinckmann J, Davis EC, Murshed M. Elastin haploinsufficiency impedes the progression of arterial calcification in MGP-deficient mice. J Bone Miner Res 2014; 29:327-37. [PMID: 23857752 DOI: 10.1002/jbmr.2039] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Revised: 05/18/2013] [Accepted: 06/10/2013] [Indexed: 01/12/2023]
Abstract
Matrix gla protein (MGP) is a potent inhibitor of extracellular matrix (ECM) mineralization. MGP-deficiency in humans leads to Keutel syndrome, a rare genetic disease hallmarked by abnormal soft tissue calcification. MGP-deficient (Mgp(-/-)) mice show progressive deposition of hydroxyapatite minerals in the arterial walls and die within 2 months of age. The mechanism of antimineralization function of MGP is not fully understood. We examined the progression of vascular calcification and expression of several chondrogenic/osteogenic markers in the thoracic aortas of Mgp(-/-) mice at various ages. Although cells with chondrocyte-like morphology have been reported in the calcified aorta, our gene expression data indicate that chondrogenic/osteogenic markers are not upregulated in the arteries prior to the initiation of calcification. Interestingly, arterial calcification in Mgp(-/-) mice appears first in the elastic laminae. Considering the known mineral scaffolding function of elastin (ELN), a major elastic lamina protein, we hypothesize that elastin content in the laminae is a critical determinant for arterial calcification in Mgp(-/-) mice. To investigate this, we performed micro-computed tomography (µCT) and histological analyses of the aortas of Mgp(-/-);Eln(+/-) mice and show that elastin haploinsufficiency significantly reduces arterial calcification in this strain. Our data suggest that MGP deficiency leads to alterations of vascular ECM that may in turn initiate arterial calcification.
Collapse
|
92
|
Annabi N, Tamayol A, Uquillas JA, Akbari M, Bertassoni LE, Cha C, Camci-Unal G, Dokmeci MR, Peppas NA, Khademhosseini A. 25th anniversary article: Rational design and applications of hydrogels in regenerative medicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2014; 26:85-123. [PMID: 24741694 PMCID: PMC3925010 DOI: 10.1002/adma.201303233] [Citation(s) in RCA: 911] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Hydrogels are hydrophilic polymer-based materials with high water content and physical characteristics that resemble the native extracellular matrix. Because of their remarkable properties, hydrogel systems are used for a wide range of biomedical applications, such as three-dimensional (3D) matrices for tissue engineering, drug-delivery vehicles, composite biomaterials, and as injectable fillers in minimally invasive surgeries. In addition, the rational design of hydrogels with controlled physical and biological properties can be used to modulate cellular functionality and tissue morphogenesis. Here, the development of advanced hydrogels with tunable physiochemical properties is highlighted, with particular emphasis on elastomeric, light-sensitive, composite, and shape-memory hydrogels. Emerging technologies developed over the past decade to control hydrogel architecture are also discussed and a number of potential applications and challenges in the utilization of hydrogels in regenerative medicine are reviewed. It is anticipated that the continued development of sophisticated hydrogels will result in clinical applications that will improve patient care and quality of life.
Collapse
Affiliation(s)
- Nasim Annabi
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Ali Tamayol
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jorge Alfredo Uquillas
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mohsen Akbari
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Luiz E. Bertassoni
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Chaenyung Cha
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Gulden Camci-Unal
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Mehmet R. Dokmeci
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nicholas A. Peppas
- Department of Biomedical Engineering, Biomedical Engineering Building 3.110B, The University of Texas at Austin, 1 University Station, C0800, Austin, Texas, 78712–1062, USA
| | - Ali Khademhosseini
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA. Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA. Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| |
Collapse
|
93
|
Regeneration of Human Dermis by a Multi-Headed Peptide. J Invest Dermatol 2014; 134:58-67. [DOI: 10.1038/jid.2013.290] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Revised: 05/30/2013] [Accepted: 06/05/2013] [Indexed: 11/08/2022]
|
94
|
Revi D, Vineetha VP, Muhamed J, Rajan A, Anilkumar TV. Porcine cholecyst-derived scaffold promotes full-thickness wound healing in rabbit. J Tissue Eng 2013; 4:2041731413518060. [PMID: 24555014 PMCID: PMC3927752 DOI: 10.1177/2041731413518060] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 11/27/2013] [Indexed: 12/22/2022] Open
Abstract
Graft-assisted healing is an important strategy for treating full-thickness skin wounds. This study evaluated the properties of porcine cholecyst-derived scaffold and its use for treating full-thickness skin wound in rabbit. The physical properties of cholecyst-derived scaffold were congenial for skin-graft application. Compared to a commercially available skin-graft substitute made of porcine small intestinal submucosa, the cholecyst-derived scaffold was rich in natural biomolecules like elastin and glycosaminoglycans. When used as a xenograft, it promoted healing with excess cell proliferation at early phases and acceptable collagen deposition in the later remodelling phases.
Collapse
Affiliation(s)
- Deepa Revi
- Division of Experimental Pathology Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| | - Vadavanath Prabhakaran Vineetha
- Division of Experimental Pathology Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| | - Jaseer Muhamed
- Division of Experimental Pathology Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| | - Akhila Rajan
- Division of Experimental Pathology Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| | - Thapasimuthu Vijayamma Anilkumar
- Division of Experimental Pathology Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum, India
| |
Collapse
|
95
|
Gunda V, Verma RK, Sudhakar YA. Inhibition of elastin peptide-mediated angiogenic signaling mechanism(s) in choroidal endothelial cells by the α6(IV)NC1 collagen fragment. Invest Ophthalmol Vis Sci 2013; 54:7828-35. [PMID: 24194191 DOI: 10.1167/iovs.12-10870] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE The inhibitory effects and mechanism(s) of type IV collagen α-6 chain-derived noncollagenous domain (α6[IV]NC1 or hexastatin) on elastin-derived peptide (EDP)-activated choroidal endothelial cell migration, kinase signaling, and membrane type 1 metalloproteinase (MT1-MMP) activation are explored. METHODS Mouse choroidal endothelial cells (MCECs) were incubated in media with soluble EDPs (kappa elastin, mouse elastin, and Val-Gly-Val-Ala-Pro-Gly [VGVAPG] hexapeptide) for different time intervals with or without α6(IV)NC1. The MCECs proliferation, migration, tube formation, MT1-MMP expression, and angiogenic signaling were analyzed in cells subjected to EDP and α6(IV)NC1 treatments. The MCECs also were subjected to EDPs, and specific inhibitors for evaluation of focal adhesion kinase (FAK) and protein kinase B (Akt) phosphorylation. RESULTS Kappa elastin, mouse elastin, and VGVAPG enhanced the migration, without affecting the proliferation of MCECs. The α6(IV)NC1 inhibited survival and EDP-activated migration of MCECs. The EDP-activated MCEC tube formation on matrigel also was inhibited by α6(IV)NC1. Further, EDP-activated MT1-MMP expression and FAK/phosphoinositide-3-kinase (PI-3K)/mammalian target of rapamycin (mToR)/Akt phosphorylation in MCECs, were reduced by α6(IV)NC1. The EDP-induced FAK and Akt phosphorylation was blocked by FAK- and Akt-specific inhibitors. CONCLUSIONS The EDPs and α6(IV)NC1 are identified to exhibit opposing effects on MCEC angiogenic behavior and signaling. The α6(IV)NC1 inhibited cell survival, EDP-mediated migration, MT1-MMP expression and, FAK/PI-3K/mToR/Akt phosphorylation in MCECs. This work demonstrates α6(IV)NC1 as a prospective endogenous molecule for the treatment of diseases involving choroidal neovascularization in the eye.
Collapse
Affiliation(s)
- Venugopal Gunda
- Cell Signaling, Retinal & Tumor Angiogenesis Laboratory, Boys Town National Research Hospital, Omaha, Nebraska
| | | | | |
Collapse
|
96
|
Lee P, Bax DV, Bilek MMM, Weiss AS. A novel cell adhesion region in tropoelastin mediates attachment to integrin αVβ5. J Biol Chem 2013; 289:1467-77. [PMID: 24293364 DOI: 10.1074/jbc.m113.518381] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Tropoelastin protein monomers assemble to form elastin. Cellular integrin αVβ3 binds RKRK at the C-terminal tail of tropoelastin. We probed cell interactions with tropoelastin by deleting the RKRK sequence to identify other cell-binding interactions within tropoelastin. We found a novel human dermal fibroblast attachment and spreading site on tropoelastin that is located centrally in the molecule. Inhibition studies demonstrated that this cell adhesion was not mediated by either elastin-binding protein or glycosaminoglycans. Cell interactions were divalent cation-dependent, indicating integrin dependence. Function-blocking monoclonal antibodies revealed that αV integrin(s) and integrin αVβ5 specifically were critical for cell adhesion to this part of tropoelastin. These data reveal a common αV integrin-binding theme for tropoelastin: αVβ3 at the C terminus and αVβ5 at the central region of tropoelastin. Each αV region contributes to fibroblast attachment and spreading, but they differ in their effects on cytoskeletal assembly.
Collapse
Affiliation(s)
- Pearl Lee
- From the School of Molecular Bioscience
| | | | | | | |
Collapse
|
97
|
Zhu B, Xu T, Yuan J, Guo X, Liu D. Transcriptome sequencing reveals differences between primary and secondary hair follicle-derived dermal papilla cells of the Cashmere goat (Capra hircus). PLoS One 2013; 8:e76282. [PMID: 24069460 PMCID: PMC3777969 DOI: 10.1371/journal.pone.0076282] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 08/22/2013] [Indexed: 12/30/2022] Open
Abstract
The dermal papilla is thought to establish the character and control the size of hair follicles. Inner Mongolia Cashmere goats (Capra hircus) have a double coat comprising the primary and secondary hair follicles, which have dramatically different sizes and textures. The Cashmere goat is rapidly becoming a potent model for hair follicle morphogenesis research. In this study, we established two dermal papilla cell lines during the anagen phase of the hair growth cycle from the primary and secondary hair follicles and clarified the similarities and differences in their morphology and growth characteristics. High-throughput transcriptome sequencing was used to identify gene expression differences between the two dermal papilla cell lines. Many of the differentially expressed genes are involved in vascularization, ECM-receptor interaction and Wnt/β-catenin/Lef1 signaling pathways, which intimately associated with hair follicle morphogenesis. These findings provide valuable information for research on postnatal morphogenesis of hair follicles.
Collapse
Affiliation(s)
- Bing Zhu
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Teng Xu
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Jianlong Yuan
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Xudong Guo
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
- * E-mail: (XG); (DL)
| | - Dongjun Liu
- The Key Laboratory of Mammalian Reproductive Biology and Biotechnology of the Ministry of Education, Inner Mongolia University, Hohhot, China
- * E-mail: (XG); (DL)
| |
Collapse
|
98
|
Sinha A, Vyavahare NR. High-glucose levels and elastin degradation products accelerate osteogenesis in vascular smooth muscle cells. Diab Vasc Dis Res 2013; 10:410-9. [PMID: 23754846 PMCID: PMC5403374 DOI: 10.1177/1479164113485101] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic disease in which the body either does not use or produce the glucose metabolising hormone insulin efficiently. Calcification of elastin in the arteries of diabetics is a major predictor of cardiovascular diseases. It has been previously shown that elastin degradation products work synergistically with transforming growth factor-beta 1 (TGF-β1) to induce osteogenesis in vascular smooth muscle cells. In this study, we tested the hypothesis that high concentration of glucose coupled with elastin degradation products and TGF-β1 (a cytokine commonly associated with diabetes) will cause a greater degree of osteogenesis compared to normal vascular cells. Thus, the goal of this study was to analyse the effects of high concentration of glucose, elastin peptides and TGF-β1 on bone-specific markers like alkaline phosphatase (ALP), osteocalcin (OCN) and runt-related transcription factor 2 (RUNX2). We demonstrated using relative gene expression and specific protein assays that elastin degradation products in the presence of high glucose cause the increase in expression of the specific elastin-laminin receptor-1 (ELR-1) and activin receptor-like kinase-5 (ALK-5) present on the surface of the vascular cells, in turn leading to overexpression of typical osteogenic markers like ALP, OCN and RUNX2. Conversely, blocking of ELR-1 and ALK-5 strongly suppressed the expression of the osteogenic proteins. In conclusion, our results indicate that glucose plays an important role in amplifying the osteogenesis induced by elastin peptides and TGF-β1, possibly by activating the ELR-1 and ALK-5 signalling pathways.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Elastin/pharmacology
- Glucose/metabolism
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Osteogenesis/drug effects
- Protein Serine-Threonine Kinases
- Rats
- Receptor, Transforming Growth Factor-beta Type I
- Receptors, Transforming Growth Factor beta
- Signal Transduction/drug effects
- Transforming Growth Factor beta1/metabolism
Collapse
Affiliation(s)
- Aditi Sinha
- Department of Bioengineering, Clemson University, Clemson, SC 29634, USA
| | | |
Collapse
|
99
|
Annabi N, Mithieux SM, Camci-Unal G, Dokmeci MR, Weiss AS, Khademhosseini A. Elastomeric Recombinant Protein-based Biomaterials. Biochem Eng J 2013; 77:110-118. [PMID: 23935392 DOI: 10.1016/j.bej.2013.05.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Elastomeric protein-based biomaterials, produced from elastin derivatives, are widely investigated as promising tissue engineering scaffolds due to their remarkable properties including substantial extensibility, long-term stability, self-assembly, high resilience upon stretching, low energy loss, and excellent biological activity. These elastomers are processed from different sources of soluble elastin such as animal-derived soluble elastin, recombinant human tropoelastin, and elastin-like polypeptides into various forms including three dimensional (3D) porous hydrogels, elastomeric films, and fibrous electrospun scaffolds. Elastin-based biomaterials have shown great potential for the engineering of elastic tissues such as skin, lung and vasculature. In this review, the synthesis and properties of various elastin-based elastomers with their applications in tissue engineering are described.
Collapse
Affiliation(s)
- Nasim Annabi
- Center for Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, 02139, USA ; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA ; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, Massachusetts, 02139, USA
| | | | | | | | | | | |
Collapse
|
100
|
Maurice P, Blaise S, Gayral S, Debelle L, Laffargue M, Hornebeck W, Duca L. Elastin fragmentation and atherosclerosis progression: The elastokine concept. Trends Cardiovasc Med 2013; 23:211-21. [DOI: 10.1016/j.tcm.2012.12.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 12/17/2012] [Accepted: 12/18/2012] [Indexed: 01/05/2023]
|