251
|
Zanotelli MR, Reinhart-King CA. Mechanical Forces in Tumor Angiogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1092:91-112. [PMID: 30368750 PMCID: PMC6986816 DOI: 10.1007/978-3-319-95294-9_6] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A defining hallmark of cancer and cancer development is upregulated angiogenesis. The vasculature formed in tumors is structurally abnormal, not organized in the conventional hierarchical arrangement, and more permeable than normal vasculature. These features contribute to leaky, tortuous, and dilated blood vessels, which act to create heterogeneous blood flow, compression of vessels, and elevated interstitial fluid pressure. As such, abnormalities in the tumor vasculature not only affect the delivery of nutrients and oxygen to the tumor, but also contribute to creating an abnormal tumor microenvironment that further promotes tumorigenesis. The role of chemical signaling events in mediating tumor angiogenesis has been well researched; however, the relative contribution of physical cues and mechanical regulation of tumor angiogenesis is less understood. Growing research indicates that the physical microenvironment plays a significant role in tumor progression and promoting abnormal tumor vasculature. Here, we review how mechanical cues found in the tumor microenvironment promote aberrant tumor angiogenesis. Specifically, we discuss the influence of matrix stiffness and mechanical stresses in tumor tissue on tumor vasculature, as well as the mechanosensory pathways utilized by endothelial cells to respond to the physical cues found in the tumor microenvironment. We also discuss the impact of the resulting aberrant tumor vasculature on tumor progression and therapeutic treatment.
Collapse
Affiliation(s)
- Matthew R Zanotelli
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Cynthia A Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
252
|
Yu C, Xing M, Sun S, Guan G, Wang L. In vitro evaluation of vascular endothelial cell behaviors on biomimetic vascular basement membranes. Colloids Surf B Biointerfaces 2019; 182:110381. [PMID: 31351274 DOI: 10.1016/j.colsurfb.2019.110381] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022]
Abstract
Vascular basement membrane (VBM) is a thin layer of fibrous extracellular matrix linking endothelium, and collagen type IV (COL IV) is its main composition. VBM plays a crucial role in anchoring down the endothelium to its loose connective tissue underneath. For vascular grafts, constructing biomimetic VBMs on the luminal surface is thus an effective approach to improve endothelialization in situ. In the present work, three types of polycaprolactone (PCL) membranes were produced and characterized through cell counting kit-8 (CCK-8) assay, adhesion force and elastic modulus test to examine the influence of fiber diameter and membrane composition on vascular endothelial cell (EC) behaviors. The PCL membranes with finer fibers of 54.77 nm (PCL-54) could biomimic the nanotopography of VBMs more efficiently than 544.64 nm (PCL-544), and they were more suitable for Pig iliac endothelium cells (PIECs) adhesion and proliferation, meanwhile, inducing higher elastic modulus and adhesion force of PIECs. On this foundation, we further immobilized COL IV onto PCL-54 (PCL-COL IV) to biomimic VBMs compositionally. Results showed that PIECs on PCL-COL IV exhibited the highest viability and proliferation. Besides, quantitative data indicated that the elastic modulus of the PIECs on PCL-COL IV (4441.00 Pa) was as two times higher than that on PCL-54 (2312.26 Pa), and the adhesion force grew to 1120.99 pN from 673.58 pN of PIECs on PCL-54. In summary, the PCL-COL IV membranes show high similarity with the native VBMs in terms of structure and composition, suggesting a promising potential for surface modification to vascular grafts for improved endothelialization.
Collapse
Affiliation(s)
- Chenglong Yu
- Engineering Research Center of Technical Textile, Ministry of Education, Key Laboratory of Textile Science and Technology of Ministry of Education, Key Laboratory of Textile Industry for Biomedical Textile materials and Technology, College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| | - Meiyi Xing
- Engineering Research Center of Technical Textile, Ministry of Education, Key Laboratory of Textile Science and Technology of Ministry of Education, Key Laboratory of Textile Industry for Biomedical Textile materials and Technology, College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| | - Shibo Sun
- Engineering Research Center of Technical Textile, Ministry of Education, Key Laboratory of Textile Science and Technology of Ministry of Education, Key Laboratory of Textile Industry for Biomedical Textile materials and Technology, College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| | - Guoping Guan
- Engineering Research Center of Technical Textile, Ministry of Education, Key Laboratory of Textile Science and Technology of Ministry of Education, Key Laboratory of Textile Industry for Biomedical Textile materials and Technology, College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China.
| | - Lu Wang
- Engineering Research Center of Technical Textile, Ministry of Education, Key Laboratory of Textile Science and Technology of Ministry of Education, Key Laboratory of Textile Industry for Biomedical Textile materials and Technology, College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China.
| |
Collapse
|
253
|
Dikici S, Claeyssens F, MacNeil S. Decellularised baby spinach leaves and their potential use in tissue engineering applications: Studying and promoting neovascularisation. J Biomater Appl 2019; 34:546-559. [DOI: 10.1177/0885328219863115] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Serkan Dikici
- Kroto Research Institute, University of Sheffield, Sheffield, UK
| | | | - Sheila MacNeil
- Kroto Research Institute, University of Sheffield, Sheffield, UK
| |
Collapse
|
254
|
Seasonal adaptations of the hypothalamo-neurohypophyseal system of the dromedary camel. PLoS One 2019; 14:e0216679. [PMID: 31211771 PMCID: PMC6581255 DOI: 10.1371/journal.pone.0216679] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 04/24/2019] [Indexed: 12/12/2022] Open
Abstract
The “ship” of the Arabian and North African deserts, the one-humped dromedary camel (Camelus dromedarius) has a remarkable capacity to survive in conditions of extreme heat without needing to drink water. One of the ways that this is achieved is through the actions of the antidiuretic hormone arginine vasopressin (AVP), which is made in a specialised part of the brain called the hypothalamo-neurohypophyseal system (HNS), but exerts its effects at the level of the kidney to provoke water conservation. Interestingly, our electron microscopy studies have shown that the ultrastructure of the dromedary HNS changes according to season, suggesting that in the arid conditions of summer the HNS is in an activated state, in preparation for the likely prospect of water deprivation. Based on our dromedary genome sequence, we have carried out an RNAseq analysis of the dromedary HNS in summer and winter. Amongst the 171 transcripts found to be significantly differentially regulated (>2 fold change, p value <0.05) there is a significant over-representation of neuropeptide encoding genes, including that encoding AVP, the expression of which appeared to increase in summer. Identification of neuropeptides in the HNS and analysis of neuropeptide profiles in extracts from individual camels using mass spectrometry indicates that overall AVP peptide levels decreased in the HNS during summer compared to winter, perhaps due to increased release during periods of dehydration in the dry season.
Collapse
|
255
|
da Silva LP, Reis RL, Correlo VM, Marques AP. Hydrogel-Based Strategies to Advance Therapies for Chronic Skin Wounds. Annu Rev Biomed Eng 2019; 21:145-169. [DOI: 10.1146/annurev-bioeng-060418-052422] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chronic skin wounds are the leading cause of nontraumatic foot amputations worldwide and present a significant risk of morbidity and mortality due to the lack of efficient therapies. The intrinsic characteristics of hydrogels allow them to benefit cutaneous healing essentially by supporting a moist environment. This property has long been explored in wound management to aid in autolytic debridement. However, chronic wounds require additional therapeutic features that can be provided by a combination of hydrogels with biochemical mediators or cells, promoting faster and better healing. We survey hydrogel-based approaches with potential to improve the healing of chronic wounds by reviewing their effects as observed in preclinical models. Topics covered include strategies to ablate infection and resolve inflammation, the delivery of bioactive agents to accelerate healing, and tissue engineering approaches for skin regeneration. The article concludes by considering the relevance of treating chronic skin wounds using hydrogel-based strategies.
Collapse
Affiliation(s)
- Lucília P. da Silva
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
- Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, 4805-017 Barco, Guimarães, Portugal
| | - Vitor M. Correlo
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
- Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, 4805-017 Barco, Guimarães, Portugal
| | - Alexandra P. Marques
- 3B's Research Group, I3B's: Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, and Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Barco, Guimarães, Portugal;, , ,
- ICVS/3B's: PT Government Associate Laboratory, 4710-057 Braga, Guimarães, Portugal
- Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, 4805-017 Barco, Guimarães, Portugal
| |
Collapse
|
256
|
Korntner S, Lehner C, Gehwolf R, Wagner A, Grütz M, Kunkel N, Tempfer H, Traweger A. Limiting angiogenesis to modulate scar formation. Adv Drug Deliv Rev 2019; 146:170-189. [PMID: 29501628 DOI: 10.1016/j.addr.2018.02.010] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 01/22/2018] [Accepted: 02/26/2018] [Indexed: 02/06/2023]
Abstract
Angiogenesis, the process of new blood vessel formation from existing blood vessels, is a key aspect of virtually every repair process. During wound healing an extensive, but immature and leaky vascular plexus forms which is subsequently reduced by regression of non-functional vessels. More recent studies indicate that uncontrolled vessel growth or impaired vessel regression as a consequence of an excessive inflammatory response can impair wound healing, resulting in scarring and dysfunction. However, in order to elucidate targetable factors to promote functional tissue regeneration we need to understand the molecular and cellular underpinnings of physiological angiogenesis, ranging from induction to resolution of blood vessels. Especially for avascular tissues (e.g. cornea, tendon, ligament, cartilage, etc.), limiting rather than boosting vessel growth during wound repair potentially is beneficial to restore full tissue function and may result in favourable long-term healing outcomes.
Collapse
|
257
|
Lopes Junior E, Leite HP, Konstantyner T. Selenium and selenoproteins: from endothelial cytoprotection to clinical outcomes. Transl Res 2019; 208:85-104. [PMID: 30738860 DOI: 10.1016/j.trsl.2019.01.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 02/07/2023]
Abstract
The role of the vascular endothelium in inflammation was demonstrated experimentally through biomarkers of endothelial dysfunction and cytoprotection. Selenium is a trace element essential for cell protection against oxidative lesions triggered by reactive oxygen species or inflammatory responses. Preclinical studies have demonstrated a relationship between adhesion molecules as biomarkers of endothelial dysfunction and selenoproteins as biomarkers of selenium status under conditions that mimic different diseases. Most studies in humans indicate an association between selenium deficiency and increased risk of morbidity and mortality, yet the pathophysiology of selenium in endothelial activation remains unknown. Here, we summarize selenium-dependent endothelial function evaluation techniques and focus on the role of selenium in endothelial cytoprotection according to current scientific knowledge. Most studies on the role of selenium in endothelial processes show selenium-dependent endothelial functions and explain how cells and tissues adapt to inflammatory insults. Taken together, these studies show an increase in adhesion molecules and a decrease in the expression of selenoproteins following a decreased exposure to selenium. Few clinical trials have enough methodological quality to be included in meta-analysis on the benefits of selenium supplementation. Furthermore, the methodology adopted in many studies does not consider the relevant findings on the pathophysiology of endothelial dysfunction. Preclinical studies should be more frequently integrated into clinical studies to provide clearer views on the role of selenium status in endothelial cytoprotection.
Collapse
Affiliation(s)
- Emilio Lopes Junior
- Discipline of Nutrition and Metabolism, Department of Pediatrics, Federal University of São Paulo, São Paulo, Brazil
| | - Heitor Pons Leite
- Discipline of Nutrition and Metabolism, Department of Pediatrics, Federal University of São Paulo, São Paulo, Brazil.
| | - Tulio Konstantyner
- Discipline of Nutrition and Metabolism, Department of Pediatrics, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
258
|
Ragelle H, Goncalves A, Kustermann S, Antonetti DA, Jayagopal A. Organ-On-A-Chip Technologies for Advanced Blood-Retinal Barrier Models. J Ocul Pharmacol Ther 2019; 36:30-41. [PMID: 31140899 PMCID: PMC6985766 DOI: 10.1089/jop.2019.0017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 04/02/2019] [Indexed: 12/16/2022] Open
Abstract
The blood-retinal barrier (BRB) protects the retina by maintaining an adequate microenvironment for neuronal function. Alterations of the junctional complex of the BRB and consequent BRB breakdown in disease contribute to a loss of neuronal signaling and vision loss. As new therapeutics are being developed to prevent or restore barrier function, it is critical to implement physiologically relevant in vitro models that recapitulate the important features of barrier biology to improve disease modeling, target validation, and toxicity assessment. New directions in organ-on-a-chip technology are enabling more sophisticated 3-dimensional models with flow, multicellularity, and control over microenvironmental properties. By capturing additional biological complexity, organs-on-chip can help approach actual tissue organization and function and offer additional tools to model and study disease compared with traditional 2-dimensional cell culture. This review describes the current state of barrier biology and barrier function in ocular diseases, describes recent advances in organ-on-a-chip design for modeling the BRB, and discusses the potential of such models for ophthalmic drug discovery and development.
Collapse
Affiliation(s)
- Héloïse Ragelle
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Andreia Goncalves
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Harbor, Michigan
| | - Stefan Kustermann
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - David A. Antonetti
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Ann Harbor, Michigan
| | - Ashwath Jayagopal
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
259
|
Lee D, Chambers M. A bilayer tissue culture model of the bovine alveolus. F1000Res 2019; 8:357. [PMID: 31448101 PMCID: PMC6685456 DOI: 10.12688/f1000research.18696.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/27/2019] [Indexed: 08/02/2024] Open
Abstract
The epithelial lining of the lung is often the first point of interaction between the host and inhaled pathogens, allergens and medications. Epithelial cells are therefore the main focus of studies which aim to shed light on host-pathogen interactions, to dissect the mechanisms of local host immunity and study toxicology. If these studies are not to be conducted exclusively in vivo, it is imperative that in vitro models are developed with a high in vitro- in vivo correlation. We describe here a co-culture bilayer model of the bovine alveolus, designed to overcome some of the limitations encountered with mono-culture and live animal models. Our system includes bovine pulmonary arterial endothelial cells (BPAECs) seeded onto a permeable membrane in 24 well Transwell format. The BPAECs are overlaid with immortalised bovine alveolar type II epithelial cells and the bilayer cultured at air-liquid interface for 14 days before use; in our case to study host-mycobacterial interactions. Characterisation of novel cell lines and the bilayer model have provided compelling evidence that immortalised bovine alveolar type II cells are an authentic substitute for primary alveolar type II cells and their culture as a bilayer in conjunction with BPAECs provides a physiologically relevant in vitro model of the bovine alveolus. The bilayer model may be used to study dynamic intracellular and extracellular host-pathogen interactions, using proteomics, genomics, live cell imaging, in-cell ELISA and confocal microscopy. The model presented in this article enables other researchers to establish an in vitro model of the bovine alveolus that is easy to set up, malleable and serves as a comparable alternative to in vivo models, whilst allowing study of early host-pathogen interactions, currently not feasible in vivo. The model therefore achieves one of the 3Rs objectives in that it replaces the use of animals in research of bovine respiratory diseases.
Collapse
Affiliation(s)
- Diane Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| | - Mark Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| |
Collapse
|
260
|
Abstract
The epithelial lining of the lung is often the first point of interaction between the host and inhaled pathogens, allergens and medications. Epithelial cells are therefore the main focus of studies which aim to shed light on host-pathogen interactions, to dissect the mechanisms of local host immunity and study toxicology. If these studies are not to be conducted exclusively in vivo, it is imperative that in vitro models are developed with a high in vitro- in vivo correlation. We describe here a co-culture model of the bovine alveolus, designed to overcome some of the limitations encountered with mono-culture and live animal models. Our system includes bovine pulmonary arterial endothelial cells (BPAECs) seeded onto a permeable membrane in 24 well Transwell format. The BPAECs are overlaid with immortalised bovine alveolar type II epithelial cells and cultured at air-liquid interface for 14 days before use; in our case to study host-mycobacterial interactions. Characterisation of novel cell lines and the co-culture model have provided compelling evidence that immortalised bovine alveolar type II cells are an authentic substitute for primary alveolar type II cells and their co-culture with BPAECs provides a physiologically relevant in vitro model of the bovine alveolus. The co-culture model may be used to study dynamic intracellular and extracellular host-pathogen interactions, using proteomics, genomics, live cell imaging, in-cell ELISA and confocal microscopy. The model presented in this article enables other researchers to establish an in vitro model of the bovine alveolus that is easy to set up, malleable and serves as a comparable alternative to in vivo models, whilst allowing study of early host-pathogen interactions, currently not feasible in vivo. The model therefore achieves one of the 3Rs objectives in that it replaces the use of animals in research of bovine respiratory diseases.
Collapse
Affiliation(s)
- Diane Lee
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| | - Mark Chambers
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, GU2 7AL, UK
| |
Collapse
|
261
|
Zhao H, Chappell JC. Microvascular bioengineering: a focus on pericytes. J Biol Eng 2019; 13:26. [PMID: 30984287 PMCID: PMC6444752 DOI: 10.1186/s13036-019-0158-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/15/2019] [Indexed: 12/26/2022] Open
Abstract
Capillaries within the microcirculation are essential for oxygen delivery and nutrient/waste exchange, among other critical functions. Microvascular bioengineering approaches have sought to recapitulate many key features of these capillary networks, with an increasing appreciation for the necessity of incorporating vascular pericytes. Here, we briefly review established and more recent insights into important aspects of pericyte identification and function within the microvasculature. We then consider the importance of including vascular pericytes in various bioengineered microvessel platforms including 3D culturing and microfluidic systems. We also discuss how vascular pericytes are a vital component in the construction of computational models that simulate microcirculation phenomena including angiogenesis, microvascular biomechanics, and kinetics of exchange across the vessel wall. In reviewing these topics, we highlight the notion that incorporating pericytes into microvascular bioengineering applications will increase their utility and accelerate the translation of basic discoveries to clinical solutions for vascular-related pathologies.
Collapse
Affiliation(s)
- Huaning Zhao
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, 2 Riverside Circle, Roanoke, VA 24016 USA.,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061 USA
| | - John C Chappell
- Center for Heart and Reparative Medicine, Fralin Biomedical Research Institute, 2 Riverside Circle, Roanoke, VA 24016 USA.,Department of Biomedical Engineering and Mechanics, Virginia Polytechnic State Institute and State University, Blacksburg, VA 24061 USA.,3Department of Basic Science Education, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016 USA
| |
Collapse
|
262
|
Biswas S, Chakrabarti S. Increased Extracellular Matrix Protein Production in Chronic Diabetic Complications: Implications of Non-Coding RNAs. Noncoding RNA 2019; 5:E30. [PMID: 30909482 PMCID: PMC6468528 DOI: 10.3390/ncrna5010030] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 12/16/2022] Open
Abstract
Management of chronic diabetic complications remains a major medical challenge worldwide. One of the characteristic features of all chronic diabetic complications is augmented production of extracellular matrix (ECM) proteins. Such ECM proteins are deposited in all tissues affected by chronic complications, ultimately causing organ damage and dysfunction. A contributing factor to this pathogenetic process is glucose-induced endothelial damage, which involves phenotypic transformation of endothelial cells (ECs). This phenotypic transition of ECs, from a quiescent state to an activated dysfunctional state, can be mediated through alterations in the synthesis of cellular proteins. In this review, we discussed the roles of non-coding RNAs, specifically microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), in such processes. We further outlined other epigenetic mechanisms regulating the biogenesis and/or function of non-coding RNAs. Overall, we believe that better understanding of such molecular processes may lead to the development of novel biomarkers and therapeutic strategies in the future.
Collapse
Affiliation(s)
- Saumik Biswas
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A5A5, Canada.
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A5A5, Canada.
| |
Collapse
|
263
|
Gao J, Hu H, Wang X. Clinically relevant concentrations of lidocaine inhibit tumor angiogenesis through suppressing VEGF/VEGFR2 signaling. Cancer Chemother Pharmacol 2019; 83:1007-1015. [PMID: 30887179 DOI: 10.1007/s00280-019-03815-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/13/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Angiogenesis, the formation of blood vessel, is required for invasive tumor growth and metastasis. In this study, the effects of lidocaine, an amide-link local anesthetic, on angiogenesis and tumor growth were investigated. METHODS In vitro angiogenesis assays were conducted using human umbilical vascular endothelial cell (HUVEC). Essential molecules involved in vascular endothelial growth factor (VEGF) signaling were analyzed. Tumor angiogenesis was analyzed using in vivo mouse tumor model. RESULTS Lidocaine at clinically relevant concentrations inhibited angiogenesis. Lidocaine inhibited endothelial cell in vitro capillary network formation on Matrigel through interfering early stage of angiogenesis. In addition, lidocaine inhibited vascular endothelial growth factor (VEGF)-stimulated endothelial cell migration and proliferation without affecting cell adhesion. Lidocaine also induced endothelial cell apoptosis in the presence of VEGF. Lidocaine suppressed VEGF-activated phosphorylation of VEGF receptor 2 (VEGFR2), PLCγ-PKC-MAPK and FAK-paxillin in endothelial cells, demonstrating that VEGF, PLC, MAPK and FAK-paxillin suppression is associated with the antiangiogenic effect of lidocaine. Importantly, the in vitro observations were translatable to in vivo B16 melanoma mouse model. Lidocaine significantly inhibited tumor angiogenesis, leading to delay of tumor growth. CONCLUSIONS This study is the first to report that lidocaine acts as an angiogenesis inhibitor. The findings provide preclinical evidence into the potential mechanisms by which lidocaine may negatively affect cancer growth and metastasis.
Collapse
Affiliation(s)
- Jin Gao
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Jingzhou Road 136, Xiangcheng District, Xiangyang, Hubei Province, 441021, China
| | - Huimin Hu
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Jingzhou Road 136, Xiangcheng District, Xiangyang, Hubei Province, 441021, China
| | - Xuesong Wang
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Jingzhou Road 136, Xiangcheng District, Xiangyang, Hubei Province, 441021, China.
| |
Collapse
|
264
|
Epidermal Growth Factor Like-domain 7 and miR-126 are abnormally expressed in diffuse Systemic Sclerosis fibroblasts. Sci Rep 2019; 9:4589. [PMID: 30872612 PMCID: PMC6418261 DOI: 10.1038/s41598-019-39485-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/12/2018] [Indexed: 11/10/2022] Open
Abstract
Systemic sclerosis (SSc) is characterized by microangiopathy with impaired reparative angiogenesis and fibrosis. Epidermal Growth Factor Like-domain 7 (EGFL7), firstly described in endothelial cells plays a pivotal role in angiogenesis. Fibroblasts (FBs) are involved in vascular remodeling, under physiological and pathological conditions. In this study, we investigated: (i) the expression of EGFL7 and its miR-126 in patients affected by diffuse cutaneous SSc (dcSSc); (ii) the ability of Transforming Growth Factor-beta (TGF-β) to modulate EGFL7 expression; (iii) the ability of EGFL7 to modulate COL1A1 expression and proliferation/migration, and (iv) the functional role of EGFL7 on angiogenesis. Patients were divided in 2 subsets: patients fulfilling the classification criteria in less than one year from Raynaud’s Phenomenon onset (Early Onset Subset–EOS), and all the others (Long Standing Subset–LSS). We show that EGFL7 expression is increased in EOS dcSSc skin and cultured FBs. EGFL7 is inducible by TGF-β on Healthy Controls (HC) FBs but not in SSc-FBs. EGFL7 decreases COL1A1 expression in EOS SSc-FBs while EGFL7 silencing up-regulates COL1A1 expression. EGFL7 promotes migration/invasion of EOS SSc-FBs but not proliferation. Finally, SSc-FBs, partially inhibit angiogenesis in organotypic coculture assays, and this is reversed by treatment with human recombinant (rh)EGFL7. We conclude that EGFL7 and its specific microRNA miR-126 may be involved in the pathogenesis of SSc vasculopathy and fibrosis.
Collapse
|
265
|
O'Rourke BP, Kramer AH, Cao LL, Inayathullah M, Guzik H, Rajadas J, Nosanchuk JD, Sharp DJ. Fidgetin-Like 2 siRNA Enhances the Wound Healing Capability of a Surfactant Polymer Dressing. Adv Wound Care (New Rochelle) 2019; 8:91-100. [PMID: 30911440 PMCID: PMC6430983 DOI: 10.1089/wound.2018.0827] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/07/2018] [Indexed: 02/02/2023] Open
Abstract
Microtubules (MTs) are intracellular polymers that provide structure to the cell, serve as railways for intracellular transport, and regulate many cellular activities, including cell migration. The dynamicity and function of the MT cytoskeleton are determined in large part by its regulatory proteins, including the recently discovered MT severing enzyme Fidgetin-like 2 (FL2). Downregulation of FL2 expression with small interfering RNA (siRNA) results in a more than twofold increase in cell migration rate in vitro as well as translates into improved wound-healing outcomes in in vivo mouse models. Here we utilized a commercially available surfactant polymer dressing (SPD) as a vehicle to deliver FL2 siRNA. To this end we incorporated collagen microparticles containing FL2 siRNA into SPD (SPD-FL2-siRNA) for direct application to the injury site. Topical application of SPD-FL2 siRNA to murine models of full-thickness excision wounds and full-thickness burn wounds resulted in significant improvements in the rate and quality of wound healing, as measured clinically and histologically, compared with controls. Wound healing occurred more rapidly and with high fidelity, resulting in properly organized collagen substructure. Taken together, these findings indicate that the incorporation of FL2 siRNA into existing treatment options is a promising avenue to improve wound outcomes.
Collapse
Affiliation(s)
| | - Adam H. Kramer
- Physiology and Biophysics, and Albert Einstein College of Medicine, Bronx, New York
| | - Longyue L. Cao
- Department of Medicine, Children's Hospital Boston, Boston, Massachusetts
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Mohammed Inayathullah
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University, Palo Alto, California
| | - Hillary Guzik
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, New York
| | - Jayakumar Rajadas
- Biomaterials and Advanced Drug Delivery Laboratory, Stanford University, Palo Alto, California
| | - Joshua D. Nosanchuk
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - David J. Sharp
- MicroCures, Inc., Research and Development, Bronx, New York
- Physiology and Biophysics, and Albert Einstein College of Medicine, Bronx, New York
- Department of Ophthalmology and Visual Sciences, and Albert Einstein College of Medicine, Bronx, New York
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
266
|
Cold-inducible RNA-binding protein as a novel target to alleviate blood–brain barrier damage induced by cardiopulmonary bypass. J Thorac Cardiovasc Surg 2019; 157:986-996.e5. [DOI: 10.1016/j.jtcvs.2018.08.100] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/29/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022]
|
267
|
Ali MS, Wang X, Lacerda CMR. The effect of physiological stretch and the valvular endothelium on mitral valve proteomes. Exp Biol Med (Maywood) 2019; 244:241-251. [PMID: 30722697 PMCID: PMC6425102 DOI: 10.1177/1535370219829006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 01/09/2019] [Indexed: 11/15/2022] Open
Abstract
IMPACT STATEMENT This work is important to the field of heart valve pathophysiology as it provides new insights into molecular markers of mechanically induced valvular degeneration as well as the protective role of the valvular endothelium. These discoveries reported here advance our current knowledge of the valvular endothelium and how its removal essentially takes valve leaflets into an environmental shock. In addition, it shows that static conditions represent a mild pathological state for valve leaflets, while 10% cyclic stretch provides valvular cell quiescence. These findings impact the field by informing disease stages and by providing potential new drug targets to reverse or slow down valvular change before it affects cardiac function.
Collapse
Affiliation(s)
- Mir S Ali
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | - Xinmei Wang
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| | - Carla MR Lacerda
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409-3121, USA
| |
Collapse
|
268
|
Hu K, Wan Q. Biphasic influence of pravastatin on human cardiac microvascular endothelial cell functions under pathological and physiological conditions. Biochem Biophys Res Commun 2019; 511:476-481. [PMID: 30803760 DOI: 10.1016/j.bbrc.2019.02.090] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 02/17/2019] [Indexed: 01/24/2023]
Abstract
HMG-CoA reductase inhibitor statins are used to treat patients with hypercholesterolemia. The pleiotropic effects of statins have been recently extended to the regulation of angiogenesis. However, the observations on the effects of statins on endothelial cells seem to be contradictory. In this work, we systematically analysed the effects of pravastatin at concentrations covering 10,000-fold range on the functions of human cardiac microvascular endothelial cells (HMVEC-C) under H2O2-induced oxidative stress and normal physiological conditions. We observed the biphasic effects of pravastatin in protecting HMVEC-C dysfunctions induced by H2O2: pravastatin at low concentrations significantly enhanced vascular network formation, growth, migration and survival under H2O2-induced oxidative stress condition whereas this effect disappeared at higher concentrations. Interestingly, pravastatin at low concentrations did not affect HMVEC-C functions but at high concentrations significantly inhibited HMVEC-C vascular network formation, growth, migration and survival in a dose-dependent manner. We further demonstrated the different molecular mechanisms of the action of pravastatin at low and high concentrations on HMVEC-C: pravastatin at low concentrations alleviates H2O2-induced oxidative stress and damage and at high concentrations inhibits prenylation. Our work provides better understanding on the multiple differential effects and the underlying mechanisms of pravastatin on HMVEC-C, which may be of relevance to the influence of statins in cardiovascular system.
Collapse
Affiliation(s)
- Kun Hu
- Department of Vascular Surgery, The Second Clinical Medical College, Yangtze University, Jingzhou Central Hospital, Jingzhou, People's Republic of China.
| | - Qian Wan
- Department of Thoracic Surgery, Xiantao First People's Hospital, Xiantao, People's Republic of China
| |
Collapse
|
269
|
Fu W, Xu P, Feng B, Lu Y, Bai J, Zhang J, Zhang W, Yin M. A hydrogel derived from acellular blood vessel extracellular matrix to promote angiogenesis. J Biomater Appl 2019; 33:1301-1313. [DOI: 10.1177/0885328219831055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The biocompatibility and bioactivity of injectable acellular extracellular matrix nominates its use as an optimal candidate for cell delivery, serving as a reconstructive scaffold. In this study, we investigated the feasibility of preparing a blood vessel matrix (BVM) hydrogel, which revealed its pro-angiogenic effects in vitro and its therapeutic effects in an in vivo skin flap model. Aortic and abdominal aortic arteries from pigs were acellularized by Triton-X 100 and confirmed by hematoxylin and eosin and 4,6-diamidino-2-phenylindole staining. Different concentrations of blood vessel matrix hydrogel were generated successfully through enzymatic digestion, neutralization, and gelation. Hematoxylin and eosin staining, Masson’s trichrome staining, collagen type I immunohistochemistry staining, and enzyme-linked immunosorbent assays showed that type I collagen and some growth factors were retained in the hydrogel. Scanning electron microscopy demonstrated the different diametric fibrils in blood vessel matrix hydrogels. A blood vessel matrix hydrogel-coated plate promoted the tube formation of human umbilical vein endothelial cells in vitro. After injection into skin flaps, the hydrogel improved the flap survival rate and increased blood perfusion and capillary density. These results indicated that we successfully prepared a blood vessel matrix hydrogel and demonstrated its general characteristics and angiogenic effects in vitro and in vivo.
Collapse
Affiliation(s)
- Wei Fu
- Department of Pediatric Cardiothoracic Surgery, Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Peng Xu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, Shanghai 200011, China
| | - Bei Feng
- Department of Pediatric Cardiothoracic Surgery, Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yang Lu
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, Shanghai 200011, China
| | - Jie Bai
- Department of Pediatric Cardiothoracic Surgery, Institute of Pediatric Translational Medicine, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jialiang Zhang
- Department of Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China*These authors contributed equally to this work
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Key Laboratory of Tissue Engineering, National Tissue Engineering Center of China, Shanghai 200011, China
| | - Meng Yin
- Department of Cardiothoracic Surgery, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China*These authors contributed equally to this work
| |
Collapse
|
270
|
Cell population balance of cardiovascular spheroids derived from human induced pluripotent stem cells. Sci Rep 2019; 9:1295. [PMID: 30718597 PMCID: PMC6362271 DOI: 10.1038/s41598-018-37686-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 12/11/2018] [Indexed: 02/06/2023] Open
Abstract
Stem cell-derived cardiomyocytes and vascular cells can be used for a variety of applications such as studying human heart development and modelling human disease in culture. In particular, protocols based on modulation of Wnt signaling were able to produce high quality of cardiomyocytes or vascular cells from human pluripotent stem cells (hPSCs). However, the mechanism behind the development of 3D cardiovascular spheroids into either vascular or cardiac cells has not been well explored. Hippo/Yes-associated protein (YAP) signaling plays important roles in the regulation of organogenesis, but its impact on cardiovascular differentiation has been less evaluated. In this study, the effects of seeding density and a change in YAP signaling on 3D cardiovascular spheroids patterning from hPSCs were evaluated. Compared to 2D culture, 3D cardiovascular spheroids exhibited higher levels of sarcomeric striations and higher length-to-width ratios of α-actinin+ cells. The spheroids with high seeding density exhibited more α-actinin+ cells and less nuclear YAP expression. The 3D cardiovascular spheroids were also treated with different small molecules, including Rho kinase inhibitor (Y27632), Cytochalasin D, Dasatinib, and Lysophosphatidic acid to modulate YAP localization. Nuclear YAP inhibition resulted in lower expression of active β-catenin, vascular marker, and MRTF, the transcription factor mediated by RhoGTPases. Y27632 also promoted the gene expression of MMP-2/-3 (matrix remodeling) and Notch-1 (Notch signaling). These results should help our understanding of the underlying effects for the efficient patterning of cardiovascular spheroids after mesoderm formation from hPSCs.
Collapse
|
271
|
Salavati H, Soltani M. The impact of endothelial cells proliferation in a multiscale realistic reproduction of angiogenesis. Biochem Eng J 2019. [DOI: 10.1016/j.bej.2018.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
272
|
Shin BH, Kim BH, Kim S, Lee K, Choy YB, Heo CY. Silicone breast implant modification review: overcoming capsular contracture. Biomater Res 2018; 22:37. [PMID: 30598837 PMCID: PMC6302391 DOI: 10.1186/s40824-018-0147-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 12/07/2018] [Indexed: 12/25/2022] Open
Abstract
Background Silicone implants are biomaterials that are frequently used in the medical industry due to their physiological inertness and low toxicity. However, capsular contracture remains a concern in long-term transplantation. To date, several studies have been conducted to overcome this problem. This review summarizes and explores these trends. Main body First, we examined the overall foreign body response from initial inflammation to fibrosis capsule formation in detail and introduced various studies to overcome capsular contracture. Secondly, we introduced that the main research approaches are to inhibit fibrosis with anti-inflammatory drugs or antibiotics, to control the topography of the surface of silicone implants, and to administer plasma treatment. Each study examined aspects of the various mechanisms by which capsular contracture could occur, and addressed the effects of inhibiting fibrosis. Conclusion This review introduces various silicone surface modification methods to date and examines their limitations. This review will help identify new directions in inhibiting the fibrosis of silicone implants.
Collapse
Affiliation(s)
- Byung Ho Shin
- 1Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Byung Hwi Kim
- 1Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea
| | - Sujin Kim
- 2Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826 Republic of Korea
| | - Kangwon Lee
- 2Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, 08826 Republic of Korea.,7Advanced Institutes of Convergence Technology, Suwon, Gyeonggi-do 16229 South Korea
| | - Young Bin Choy
- 1Department of Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080 Republic of Korea.,3Interdisciplinary Program for Bioengineering, College of Engineering, Seoul National University, Seoul, 08826 Republic of Korea.,6Institute of Medical & Biological Engineering, Medical Research Center, Seoul National University, Seoul, 03080 Republic of Korea
| | - Chan Yeong Heo
- 3Interdisciplinary Program for Bioengineering, College of Engineering, Seoul National University, Seoul, 08826 Republic of Korea.,4Department of Plastic and Reconstructive Surgery, College of Medicine, Seoul National University, Seoul, 03080 Republic of Korea.,5Department of Plastic and Reconstructive Surgery, Seoul National University Bundang Hospital, Seongnam, 13620 Republic of Korea
| |
Collapse
|
273
|
Witjas FMR, van den Berg BM, van den Berg CW, Engelse MA, Rabelink TJ. Concise Review: The Endothelial Cell Extracellular Matrix Regulates Tissue Homeostasis and Repair. Stem Cells Transl Med 2018; 8:375-382. [PMID: 30537441 PMCID: PMC6431685 DOI: 10.1002/sctm.18-0155] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/26/2018] [Indexed: 12/14/2022] Open
Abstract
All tissues are surrounded by a mixture of noncellular matrix components, that not only provide physical and mechanical support to cells, but also mediate biochemical signaling between cells. The extracellular matrix (ECM) of endothelial cells, also known as the perivascular matrix, forms an organ specific vascular niche that orchestrates mechano‐, growth factor, and angiocrine signaling required for tissue homeostasis and organ repair. This concise review describes how this perivascular ECM functions as a signaling platform and how this knowledge can impact the field of regenerative medicine, for example, when designing artificial matrices or using decellularized scaffolds from organs. stem cells translational medicine2019;8:375–382
Collapse
Affiliation(s)
- Franca M R Witjas
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bernard M van den Berg
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Cathelijne W van den Berg
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marten A Engelse
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ton J Rabelink
- The Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
274
|
Yu Y, Situ Q, Jia W, Li J, Wu Q, Lei J. Data driven mathematical modeling reveals the dynamic mechanism of MSC-induced neovascularization. FASEB J 2018; 33:3496-3509. [PMID: 30517036 DOI: 10.1096/fj.201801652r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Coculture of mesenchymal stem cells (MSCs) and vascular endothelial cells (ECs) in vitro leads to the formation of a capillary-like reticular structure by ECs, which has great potential as a better substitute for artificial blood vessels in terms of stability and functionality. To investigate the mechanisms of the early neovascularization induced by MSCs, we analyzed the kinematic features of the motion of ECs and concluded that the dynamic interaction between cells and the extracellular matrix would reveal the capillary-like structure formation. Based on this hypothesis, we proposed a mathematical model to simulate the vascular-like migration pattern of ECs in silico, which was confirmed by in vitro studies. These in vitro studies validated that the dynamic secretion and degradation of collagen I is the critical factor for capillary structure formation. The model proposed based on cell tracking, single cell sequencing, and mathematical simulation provides a better understanding of the neovascularization process induced by MSCs and a possible simple explanation guiding this important cellular behavior.-Yu, Y., Situ, Q., Jia, W., Li, J., Wu, Q., Lei, J. Data driven mathematical modeling reveals the dynamic mechanism of MSC-induced neovascularization.
Collapse
Affiliation(s)
- Yingting Yu
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China; and
| | - Qiaojun Situ
- Zhou Pei-Yuan Center for Applied Mathematics, Tsinghua University, Beijing, China
| | - Wangyue Jia
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China; and
| | - Junxiang Li
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China; and
| | - Qiong Wu
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, China.,Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China; and
| | - Jinzhi Lei
- MOE Key Laboratory of Bioinformatics, Tsinghua University, Beijing, China.,Zhou Pei-Yuan Center for Applied Mathematics, Tsinghua University, Beijing, China
| |
Collapse
|
275
|
Wei Z, Gerecht S. A self-healing hydrogel as an injectable instructive carrier for cellular morphogenesis. Biomaterials 2018; 185:86-96. [PMID: 30236839 PMCID: PMC6432635 DOI: 10.1016/j.biomaterials.2018.09.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/06/2018] [Accepted: 09/02/2018] [Indexed: 12/24/2022]
Abstract
Transplantation of progenitor cells can accelerate tissue healing and regenerative processes. Nonetheless, direct cell delivery fails to support survival of transplanted cells or long-term treatment of vascular related diseases due to compromised vasculature and tissue conditions. Using injectable hydrogels that cross-link in situ, could protect cells in vivo, but their sol-gel transition is time-dependent and difficult to precisely control. Hydrogels with self-healing properties are proposed to address these limitations, yet current self-healing hydrogels lack bio-functionality, hindering the morphogenesis of delivered cells into a tissue structure. Here we establish a gelatin (Gtn)-based self-healing hydrogel cross-linked by oxidized dextran (Odex) as an injectable carrier for delivery of endothelial progenitors. The dynamic imine cross-links between Gtn and Odex confer the self-healing ability to the Gtn-l-Odex hydrogels following syringe injection. The self-healing Gtn-l-Odex not only protects the progenitors from injected shear force but it also allows controllable spatial/temporal placement of the cells. Moreover, owing to the cell-adhesive and proteolytic sites of Gtn, the Gtn-l-Odex hydrogels support complex vascular network formation from the endothelial progenitors, both in vitro and in vivo. This is the first report of injectable, self-healing hydrogels with biological properties promoting vascular morphogenesis, which holds great promise for accelerating the success of regenerative therapies.
Collapse
Affiliation(s)
- Zhao Wei
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, The Institute for NanoBioTechnology Physical-Sciences Oncology Center, Johns Hopkins University, Baltimore, MD 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| |
Collapse
|
276
|
Post A, Wang E, Cosgriff-Hernandez E. A Review of Integrin-Mediated Endothelial Cell Phenotype in the Design of Cardiovascular Devices. Ann Biomed Eng 2018; 47:366-380. [PMID: 30488311 DOI: 10.1007/s10439-018-02171-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 11/20/2018] [Indexed: 12/14/2022]
Abstract
Sustained biomaterial thromboresistance has long been a goal and challenge in blood-contacting device design. Endothelialization is one of the most successful strategies to achieve long-term thromboresistance of blood-contacting devices, with the endothelial cell layer providing dynamic hemostatic regulation. It is well established that endothelial cell behavior is influenced by interactions with the underlying extracellular matrix (ECM). Numerous researchers have sought to exploit these interactions to generate improved blood-contacting devices by investigating the expression of hemostatic regulators in endothelial cells on various ECM coatings. The ability to select substrates that promote endothelial cell-mediated thromboresistance is crucial to advancing material design strategies to improve cardiovascular device outcomes. This review provides an overview of endothelial cell regulation of hemostasis, the major components found within the cardiovascular basal lamina, and the interactions of endothelial cells with prominent ECM components of the basement membrane. A summary of ECM-mimetic strategies used in cardiovascular devices is provided with a focus on the effects of key adhesion modalities on endothelial cell regulators of hemostasis.
Collapse
Affiliation(s)
- Allison Post
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Ellen Wang
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Elizabeth Cosgriff-Hernandez
- Department of Biomedical Engineering, University of Texas, 107 W. Dean Keaton, BME 3.503D, 1 University Station, C0800, Austin, TX, 78712, USA.
| |
Collapse
|
277
|
Monte F, Cebe T, Ripperger D, Ighani F, Kojouharov HV, Chen BM, Kim HKW, Aswath PB, Varanasi VG. Ionic silicon improves endothelial cells' survival under toxic oxidative stress by overexpressing angiogenic markers and antioxidant enzymes. J Tissue Eng Regen Med 2018; 12:2203-2220. [PMID: 30062712 PMCID: PMC6508967 DOI: 10.1002/term.2744] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 11/10/2022]
Abstract
Oxidative stress, induced by harmful levels of reactive oxygen species, is a common occurrence that impairs proper bone defect vascular healing through the impairment of endothelial cell function. Ionic silicon released from silica-based biomaterials, can upregulate hypoxia-inducible factor-1α (HIF-1α). Yet it is unclear whether ionic Si can restore endothelial cell function under oxidative stress conditions. Therefore, we hypothesized that ionic silicon can help improve human umbilical vein endothelial cells' (HUVECs') survival under toxic oxidative stress. In this study, we evaluated the ionic jsilicon effect on HUVECs viability, proliferation, migration, gene expression, and capillary tube formation under normal conditions and under harmful hydrogen peroxide levels. We demonstrated that 0.5-mM Si4+ significantly enhanced angiogenesis in HUVECs under normal condition (p < 0.05). HUVECs exposed to 0.5-mM Si4+ presented a morphological change, even without the bed of Matrigel, and formed significantly more tube-like structures than the control (p < 0.001). In addition, 0.5-mM Si4+ enhanced cell viability in HUVECs under harmful H2 O2 levels. HIF-1α, vascular endothelial growth factor-A, and vascular endothelial growth factor receptor-2 were overexpressed more than twofold in silicon-treated HUVECs, under normal and toxic H2 O2 conditions. Moreover, the HUVECs were treated with 0.5-mM Si4+ overexpressed superoxide dismutase-1 (SOD-1), catalase-1 (Cat-1), and nitric oxide synthase-3 (NOS3) under normal and oxidative stress environment (p < 0.01). A computational model was used for explaining the antioxidant effect of Si4+ in endothelial cells and human periosteum cells by SOD-1 enhancement. In conclusion, we demonstrated that 0.5-mM Si4+ can recover the HUVECs' viability under oxidative stress conditions by reducing cell death and upregulating expression of angiogenic and antioxidant factors.
Collapse
Affiliation(s)
- Felipe Monte
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital, Dallas, Texas
| | - Tugba Cebe
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | | | - Fareed Ighani
- Texas A&M University College of Dentistry, Dallas, Texas
| | | | - Benito M. Chen
- Department of Mathematics, University of Texas at Arlington, Arlington, Texas
| | - Harry K. W. Kim
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital, Dallas, Texas
- Department of Orthopedic Surgery, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas
| | - Pranesh B. Aswath
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas
| | - Venu G. Varanasi
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas
- Department of College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| |
Collapse
|
278
|
Sweeney M, Foldes G. It Takes Two: Endothelial-Perivascular Cell Cross-Talk in Vascular Development and Disease. Front Cardiovasc Med 2018; 5:154. [PMID: 30425990 PMCID: PMC6218412 DOI: 10.3389/fcvm.2018.00154] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 10/10/2018] [Indexed: 12/26/2022] Open
Abstract
The formation of new blood vessels is a crucial step in the development of any new tissue both during embryogenesis and in vitro models as without sufficient perfusion the tissue will be unable to grow beyond the size where nutrition and oxygenation can be managed by diffusion alone. Endothelial cells are the primary building block of blood vessels and are capable of forming tube like structures independently however they are unable to independently form functional vasculature which is capable of conducting blood flow. This requires support from other structures including supporting perivascular cells and the extracellular matrix. The crosstalk between endothelial cells and perivascular cells is vital in regulating vasculogenesis and angiogenesis and the consequences when this is disrupted can be seen in a variety of congenital and acquired disease states. This review details the mechanisms of vasculogenesis in vivo during embryogenesis and compares this to currently employed in vitro techniques. It also highlights clinical consequences of defects in the endothelial cell-pericyte cross-talk and highlights therapies which are being developed to target this pathway. Improving the understanding of the intricacies of endothelial-pericyte signaling will inform pathophysiology of multiple vascular diseases and allow the development of effective in vitro models to guide drug development and assist with approaches in tissue engineering to develop functional vasculature for regenerative medicine applications.
Collapse
Affiliation(s)
- Mark Sweeney
- Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Gabor Foldes
- Cardiovascular Division, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| |
Collapse
|
279
|
Li M, Qian M, Kyler K, Xu J. Endothelial-Vascular Smooth Muscle Cells Interactions in Atherosclerosis. Front Cardiovasc Med 2018; 5:151. [PMID: 30406116 PMCID: PMC6207093 DOI: 10.3389/fcvm.2018.00151] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory process that can eventually lead to cardiovascular disease (CVD). Despite available treatment, the prevalence of atherosclerotic CVD, which has become the leading cause of death worldwide, persists. Identification of new mechanisms of atherogenesis are highly needed in order to develop an effective therapeutic treatment. The blood vessels contain two primary major cell types: endothelial cells (EC) and vascular smooth muscle cells (VSMC). Each of these performs an essential function in sustaining vascular homeostasis. EC-VSMC communication is essential not only to development, but also to the homeostasis of mature blood vessels. Aberrant EC-VSMC interaction could promote atherogenesis. Identification of the mode of EC-VSMC crosstalk that regulates vascular functionality and sustains homeostasis may offer strategic insights for prevention and treatment of atherosclerotic CVD. Here we will review the molecular mechanisms underlying the interplay between EC and VSMC that could contribute to atherosclerosis. We also highlight open questions for future research directions.
Collapse
Affiliation(s)
- Manna Li
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Ming Qian
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Kathy Kyler
- Office of Research Administration, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Jian Xu
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| |
Collapse
|
280
|
Herrera A, Herrera M, Guerra-Perez N, Galindo-Pumariño C, Larriba MJ, García-Barberán V, Gil B, Giménez-Moyano S, Ferreiro-Monteagudo R, Veguillas P, Candia A, Peña R, Pinto J, García-Bermejo ML, Muñoz A, García de Herreros A, Bonilla F, Carrato A, Peña C. Endothelial cell activation on 3D-matrices derived from PDGF-BB-stimulated fibroblasts is mediated by Snail1. Oncogenesis 2018; 7:76. [PMID: 30250018 PMCID: PMC6155204 DOI: 10.1038/s41389-018-0085-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 08/26/2018] [Indexed: 01/26/2023] Open
Abstract
Carcinomas, such as colon cancer, initiate their invasion by rescuing the innate plasticity of both epithelial cells and stromal cells. Although Snail is a transcriptional factor involved in the Epithelial-Mesenchymal Transition, in recent years, many studies have also identified the major role of Snail in the activation of Cancer-Associated Fibroblast (CAF) cells and the remodeling of the extracellular matrix. In CAFs, Platelet-derived growth factor (PDGF) receptor signaling is a major functional determinant. High expression of both SNAI1 and PDGF receptors is associated with poor prognosis in cancer patients, but the mechanism(s) that underlie these connections are not understood. In this study, we demonstrate that PDGF-activated fibroblasts stimulate extracellular matrix (ECM) fiber remodeling and deposition. Furthermore, we describe how SNAI1, through the FAK pathway, is a necessary factor for ECM fiber organization. The parallel-oriented fibers are used by endothelial cells as “tracks”, facilitating their activation and the creation of tubular structures mimicking in vivo capillary formation. Accordingly, Snail1 expression in fibroblasts was required for the co-adjuvant effect of these cells on matrix remodeling and neoangiogenesis when co-xenografted in nude mice. Finally, in tumor samples from colorectal cancer patients a direct association between stromal SNAI1 expression and the endothelial marker CD34 was observed. In summary, our results advance the understanding of PDGF/SNAI1-activated CAFs in matrix remodeling and angiogenesis stimulation.
Collapse
Affiliation(s)
- Alberto Herrera
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro de Majadahonda, Majadahonda, Madrid, Spain
| | - Mercedes Herrera
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro de Majadahonda, Majadahonda, Madrid, Spain.,Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Natalia Guerra-Perez
- Medical Oncology Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Cristina Galindo-Pumariño
- Medical Oncology Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - María Jesús Larriba
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, CIBERONC, Madrid, Spain
| | - Vanesa García-Barberán
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro de Majadahonda, Majadahonda, Madrid, Spain.,Laboratory of Molecular Oncology, IIS Hospital Clínico San Carlos, CIBERONC, Madrid, Spain
| | - Beatriz Gil
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro de Majadahonda, Majadahonda, Madrid, Spain.,Laboratorio de Oncología Traslacional y Nuevas Terapias. Instituto de Investigación i+12, Madrid, Spain
| | - Sara Giménez-Moyano
- Biomarkers and Therapeutic Targets Lab, Pathology Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Reyes Ferreiro-Monteagudo
- Medical Oncology Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Pilar Veguillas
- Surgery Department, Hospital Universitario de Guadalajara, Guadalajara, Spain
| | - Antonio Candia
- Pathology Department, Hospital Universitario de Guadalajara, Guadalajara, Spain
| | - Raúl Peña
- Programa de Recerca en Càncer, Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona, Spain
| | - Jesús Pinto
- Pathology Department, Virgen de la Concha Hospital, Zamora, Castilla y León, Spain
| | - Mª Laura García-Bermejo
- Laboratorio de Oncología Traslacional y Nuevas Terapias. Instituto de Investigación i+12, Madrid, Spain
| | - Alberto Muñoz
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, CIBERONC, Madrid, Spain
| | | | | | - Alfredo Carrato
- Medical Oncology Department, Ramon y Cajal University Hospital, IRYCIS, CIBERONC, Alcala University, Madrid, Spain
| | - Cristina Peña
- Department of Medical Oncology, Hospital Universitario Puerta de Hierro de Majadahonda, Majadahonda, Madrid, Spain. .,Medical Oncology Department, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), CIBERONC, Madrid, Spain.
| |
Collapse
|
281
|
Que RA, Crakes DR, Abdulhadi F, Niu C, Da Silva NA, Wang S. Tailoring Collagen to Engineer the Cellular Microenvironment. Biotechnol J 2018; 13:e1800140. [DOI: 10.1002/biot.201800140] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Revised: 08/14/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Richard A. Que
- Department of Biomedical EngineeringUniversity of CaliforniaIrvineCA92697USA
| | - Dale R. Crakes
- Department of Biomedical EngineeringUniversity of CaliforniaIrvineCA92697USA
| | - Faten Abdulhadi
- Department of Chemical and Biomolecular EngineeringUniversity of CaliforniaIrvineCA92697USA
| | - Chun‐Hao Niu
- Department of Chemical and Biomolecular EngineeringUniversity of CaliforniaIrvineCA92697USA
| | - Nancy A. Da Silva
- Department of Biomedical EngineeringUniversity of CaliforniaIrvineCA92697USA
- Department of Chemical and Biomolecular EngineeringUniversity of CaliforniaIrvineCA92697USA
| | - Szu‐Wen Wang
- Department of Biomedical EngineeringUniversity of CaliforniaIrvineCA92697USA
- Department of Chemical and Biomolecular EngineeringUniversity of CaliforniaIrvineCA92697USA
| |
Collapse
|
282
|
Marchand M, Monnot C, Muller L, Germain S. Extracellular matrix scaffolding in angiogenesis and capillary homeostasis. Semin Cell Dev Biol 2018; 89:147-156. [PMID: 30165150 DOI: 10.1016/j.semcdb.2018.08.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 07/31/2018] [Accepted: 08/14/2018] [Indexed: 01/03/2023]
Abstract
The extracellular matrix (ECM) of blood vessels, which is composed of both the vascular basement membrane (BM) and the interstitial ECM is identified as a crucial component of the vasculature. We here focus on the unique molecular composition and scaffolding of the capillary ECM, which provides structural support to blood vessels and regulates properties of endothelial cells and pericytes. The major components of the BM are collagen IV, laminins, heparan sulfate proteoglycans and nidogen and also associated proteins such as collagen XVIII and fibronectin. Their organization and scaffolding in the BM is required for proper capillary morphogenesis and maintenance of vascular homeostasis. The BM also regulates vascular mechanosensing. A better understanding of the mechanical and structural properties of the vascular BM and interstitial ECM therefore opens new perspectives to control physiological and pathological angiogenesis and vascular homeostasis. The overall aim of this review is to explain how ECM scaffolding influences angiogenesis and capillary integrity.
Collapse
Affiliation(s)
- Marion Marchand
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France; Sorbonne Université, Collège Doctoral, F-75005 Paris, France
| | - Catherine Monnot
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France
| | - Laurent Muller
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, 11 Place Marcelin Berthelot, 75005, Paris, France.
| |
Collapse
|
283
|
Alfonso-Garcia A, Shklover J, Sherlock BE, Panitch A, Griffiths LG, Marcu L. Fiber-based fluorescence lifetime imaging of recellularization processes on vascular tissue constructs. JOURNAL OF BIOPHOTONICS 2018; 11:e201700391. [PMID: 29781171 PMCID: PMC7700018 DOI: 10.1002/jbio.201700391] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/16/2018] [Indexed: 05/22/2023]
Abstract
New techniques able to monitor the maturation of tissue engineered constructs over time are needed for a more efficient control of developmental parameters. Here, a label-free fluorescence lifetime imaging (FLIm) approach implemented through a single fiber-optic interface is reported for nondestructive in situ assessment of vascular biomaterials. Recellularization processes of antigen removed bovine pericardium scaffolds with endothelial cells and mesenchymal stem cells were evaluated on the serous and the fibrous sides of the scaffolds, 2 distinct extracellular matrix niches, over the course of a 7 day culture period. Results indicated that fluorescence lifetime successfully report cell presence resolved from extracellular matrix fluorescence. The recellularization process was more rapid on the serous side than on the fibrous side for both cell types, and endothelial cells expanded faster than mesenchymal stem cells on antigen-removed bovine pericardium. Fiber-based FLIm has the potential to become a nondestructive tool for the assessment of tissue maturation by allowing in situ imaging of intraluminal vascular biomaterials.
Collapse
Affiliation(s)
- Alba Alfonso-Garcia
- Department of Biomedical Engineering, University of California Davis, Davis, California
| | - Jeny Shklover
- Department of Biomedical Engineering, University of California Davis, Davis, California
| | - Benjamin E. Sherlock
- Department of Biomedical Engineering, University of California Davis, Davis, California
| | - Alyssa Panitch
- Department of Biomedical Engineering, University of California Davis, Davis, California
| | - Leigh G. Griffiths
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Laura Marcu
- Department of Biomedical Engineering, University of California Davis, Davis, California
| |
Collapse
|
284
|
Lee Y, Chakraborty S, Meininger CJ, Muthuchamy M. Insulin resistance disrupts cell integrity, mitochondrial function, and inflammatory signaling in lymphatic endothelium. Microcirculation 2018; 25:e12492. [PMID: 30025187 DOI: 10.1111/micc.12492] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Lymphatic vessel dysfunction and increased lymph leakage have been directly associated with several metabolic diseases. However, the underlying cellular mechanisms causing lymphatic dysfunction have not been determined. Aberrant insulin signaling affects the metabolic function of cells and consequently impairs tissue function. We hypothesized that insulin resistance in LECs decreases eNOS activity, disrupts barrier integrity increases permeability, and activates mitochondrial dysfunction and pro-inflammatory signaling pathways. METHODS LECs were treated with insulin and/or glucose to determine the mechanisms leading to insulin resistance. RESULTS Acute insulin treatment increased eNOS phosphorylation and NO production in LECs via activation of the PI3K/Akt signaling pathway. Prolonged hyperglycemia and hyperinsulinemia induced insulin resistance in LECs. Insulin-resistant LECs produced less NO due to a decrease in eNOS phosphorylation and showed a significant decrease in impedance across an LEC monolayer that was associated with disruption in the adherence junctional proteins. Additionally, insulin resistance in LECs impaired mitochondrial function by decreasing basal-, maximal-, and ATP-linked OCRs and activated NF-κB nuclear translocation coupled with increased pro-inflammatory signaling. CONCLUSION Our data provide the first evidence that insulin resistance disrupts endothelial barrier integrity, decreases eNOS phosphorylation and mitochondrial function, and activates inflammation in LECs.
Collapse
Affiliation(s)
- Yang Lee
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| | - Sanjukta Chakraborty
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| | - Cynthia J Meininger
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| | - Mariappan Muthuchamy
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| |
Collapse
|
285
|
Lin CH, Su JJM, Lee SY, Lin YM. Stiffness modification of photopolymerizable gelatin-methacrylate hydrogels influences endothelial differentiation of human mesenchymal stem cells. J Tissue Eng Regen Med 2018; 12:2099-2111. [PMID: 30058281 DOI: 10.1002/term.2745] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 06/09/2018] [Accepted: 07/13/2018] [Indexed: 01/19/2023]
Abstract
For stem cell differentiation, the microenvironment can play an important role, and hydrogels can provide a three-dimensional microenvironment to allow native cell growth in vitro. A challenge is that the stem cell's differentiation can be influenced by the matrix stiffness. We demonstrate a low-toxicity method to create different stiffness matrices, by using a photopolymerizable gelatin methacrylate (GelMA) hydrogel cross-linked by blue light (440 nm). The stiffness and porosity of GelMA hydrogel is easily modified by altering its concentration. We used human bone marrow mesenchymal stem cells (MSCs) as a cell source and cultured the GelMA-encapsulated cells with EGM-2 medium to induce endothelial differentiation. In our GelMA blue light hydrogel system, we found that MSCs can be differentiated into both endothelial-like and osteogenic-like cells. The mRNA expressions of endothelial cell markers CD31, von Willebrand factor, vascular endothelial growth factor receptor-2, and CD34 were significantly increased in softer GelMA hydrogels (7.5% and 10%) compared with stiffer matrices (15% GelMA). On the other hand, the enhancements of osteogenic markers mRNA expressions (Alkaline phosphatase (ALP), Runx2, osteocalcin, and osteopontin) were highest in 10% GelMA. We also found that 10% GelMA hydrogel offered optimal conditions for MSCs to form capillary-like structures. These results suggest that the mechanical properties of the GelMA hydrogel can influence both endothelial and osteogenic differentiation of MSCs and sequent capillary-like formation.
Collapse
Affiliation(s)
- Chih-Hsin Lin
- Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,The David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | | | - Shyh-Yuan Lee
- Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yuan-Min Lin
- Department of Dentistry, National Yang-Ming University, Taipei, Taiwan.,Department of Stomatology, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
286
|
Abstract
Ligaments serve as compliant connectors between hard tissues. In that role, they function under various load regimes and directions. The 3D structure of ligaments is considered to form as a uniform entity that changes due to function. The periodontal ligament (PDL) connects the tooth to the bone and sustains different types of loads in various directions. Using the PDL as a model, employing a fabricated motorized setup in a microCT, we demonstrate that the fibrous network structure within the PDL is not uniform, even before the tooth becomes functional. Utilizing morphological automated segmentation methods, directionality analysis, as well as second harmonic generation imaging, we find high correlation between blood vessel distribution and fiber density. We also show a structural feature in a form of a dense collar around the neck of the tooth as well as a preferred direction of the fibrous network. Finally, we show that the PDL develops as a nonuniform structure, with an architecture designed to sustain specific types of load in designated areas. Based on these findings, we propose that ligaments in general should be regarded as nonuniform entities, structured already at developmental stages for optimal functioning under variable load regimes.
Collapse
|
287
|
Nguyen EH, Murphy WL. Customizable biomaterials as tools for advanced anti-angiogenic drug discovery. Biomaterials 2018; 181:53-66. [PMID: 30077137 DOI: 10.1016/j.biomaterials.2018.07.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/17/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
The inhibition of angiogenesis is a critical element of cancer therapy, as cancer vasculature contributes to tumor expansion. While numerous drugs have proven to be effective at disrupting cancer vasculature, patient survival has not significantly improved as a result of anti-angiogenic drug treatment. Emerging evidence suggests that this is due to a combination of unintended side effects resulting from the application of anti-angiogenic compounds, including angiogenic rebound after treatment and the activation of metastasis in the tumor. There is currently a need to better understand the far-reaching effects of anti-angiogenic drug treatments in the context of cancer. Numerous innovations and discoveries in biomaterials design and tissue engineering techniques are providing investigators with tools to develop physiologically relevant vascular models and gain insights into the holistic impact of drug treatments on tumors. This review examines recent advances in the design of pro-angiogenic biomaterials, specifically in controlling integrin-mediated cell adhesion, growth factor signaling, mechanical properties and oxygen tension, as well as the implementation of pro-angiogenic materials into sophisticated co-culture models of cancer vasculature.
Collapse
Affiliation(s)
- Eric H Nguyen
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Human Models for Analysis of Pathways (Human MAPs) Center, University of Wisconsin, Madison, WI, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Human Models for Analysis of Pathways (Human MAPs) Center, University of Wisconsin, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
288
|
Zhao Q, Cui H, Wang J, Chen H, Wang Y, Zhang L, Du X, Wang M. Regulation Effects of Biomimetic Hybrid Scaffolds on Vascular Endothelium Remodeling. ACS APPLIED MATERIALS & INTERFACES 2018; 10:23583-23594. [PMID: 29943973 DOI: 10.1021/acsami.8b06205] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The formation of complete and well-functioning endothelium is critical for the success of tissue-engineered vascular grafts yet remaining a fundamental challenge. Endothelium remodeling onto the lumen of tissue-engineered vascular grafts is affected by their topographical, mechanical, and biochemical characteristics. For meeting multiple requirements, composite strategies have recently emerged for fabricating hybrid scaffolds, where the integrated properties are tuned by varying their compositions. However, the underlying principle how the integrated properties of hybrid scaffolds regulate vascular endothelium remodeling remains unclear. To uncover the regulation effects of hybrid scaffolds on vascular endothelium remodeling, we prepared different biomimetic hybrid scaffolds using gelatin methacrylamide (GelMA) and poly-ε-caprolactone (PCL) and then investigated vascular endothelial cell responses on them. GelMA and PCL, respectively, conferred the resulting scaffolds with biomimetic bioactivity and mechanical properties, which were tuned by varying GelMA/PCL mass ratios (3:1, 1:1, or 1:3). On different GelMA/PCL hybrid scaffolds, distinct vascular endothelial cell responses were observed. Firm cell-scaffold/cell-cell interactions were rapidly established on the hybrid scaffolds with the highest mass ratio of bioactive GelMA. However, they were mechanically insufficient as vascular grafts. On the contrary, the scaffolds with the highest mass ratio of PCL showed significantly reinforced mechanical properties but poor biological performance. Between the two extremes, the scaffolds with the same GelMA/PCL mass ratio balanced the pros and cons of two materials. Therefore, they could meet the mechanical requirements of vascular grafts and support the early-stage vascular endothelial cell remodeling by appropriate biological signaling and mechanotransduction. This investigation experimentally proves that scaffold bioactivity is the dominant factor affecting vascular endothelial cell adhesion and remodeling, whereas mechanical properties are crucial factors for the integrity of endothelium. This work offers a universal design strategy for desirable vascular grafts for improved endothelium remodeling.
Collapse
Affiliation(s)
- Qilong Zhao
- Institute of Biomedical & Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT) , Chinese Academy of Sciences (CAS) , Shenzhen 518055 , China
| | - Huanqing Cui
- Institute of Biomedical & Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT) , Chinese Academy of Sciences (CAS) , Shenzhen 518055 , China
| | - Juan Wang
- Institute of Biomedical & Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT) , Chinese Academy of Sciences (CAS) , Shenzhen 518055 , China
| | - Hongxu Chen
- Institute of Biomedical & Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT) , Chinese Academy of Sciences (CAS) , Shenzhen 518055 , China
| | - Yunlong Wang
- Institute of Biomedical & Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT) , Chinese Academy of Sciences (CAS) , Shenzhen 518055 , China
| | - Lidong Zhang
- Department of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200062 , China
| | - Xuemin Du
- Institute of Biomedical & Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT) , Chinese Academy of Sciences (CAS) , Shenzhen 518055 , China
| | - Min Wang
- Department of Mechanical Engineering , The University of Hong Kong , Hong Kong , China
| |
Collapse
|
289
|
Molecular profiling of reticular gigantocellularis neurons indicates that eNOS modulates environmentally dependent levels of arousal. Proc Natl Acad Sci U S A 2018; 115:E6900-E6909. [PMID: 29967172 DOI: 10.1073/pnas.1806123115] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neurons of the medullary reticular nucleus gigantocellularis (NGC) and their targets have recently been a focus of research on mechanisms supporting generalized CNS arousal (GA) required for proper cognitive functions. Using the retro-TRAP method, we characterized transcripts enriched in NGC neurons which have projections to the thalamus. The unique expression and activation of the endothelial nitric oxide (eNOS) signaling pathway in these cells and their intimate connections with blood vessels indicate that these neurons exert direct neurovascular coupling. Production of nitric oxide (NO) within eNOS-positive NGC neurons increases after environmental perturbations, indicating a role for eNOS/NO in modulating environmentally appropriate levels of GA. Inhibition of NO production causes dysregulated behavioral arousal after exposure to environmental perturbation. Further, our findings suggest interpretations for associations between psychiatric disorders and mutations in the eNOS locus.
Collapse
|
290
|
Kim DH, Cui LH, Seo HR, Joo HJ, Choi SC, Lim DS, Lee KB. Fabrication of Gradient Nanopattern by Thermal Nanoimprinting Technique and Screening of the Response of Human Endothelial Colony-forming Cells. J Vis Exp 2018:57661. [PMID: 30010660 PMCID: PMC6102024 DOI: 10.3791/57661] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nanotopography can be found in various extracellular matrices (ECMs) around the body and is known to have important regulatory actions upon cellular reactions. However, it is difficult to determine the relation between the size of a nanostructure and the responses of cells owing to the lack of proper screening tools. Here, we show the development of reproducible and cost-effective gradient nanopattern plates for the manipulation of cellular responses. Using anodic aluminum oxide (AAO) as a master mold, gradient nanopattern plates with nanopillars of increasing diameter ranges [120-200 nm (GP 120/200), 200-280 nm (GP 200/280), and 280-360 nm (GP 280/360)] were fabricated by a thermal imprinting technique. These gradient nanopattern plates were designed to mimic the various sizes of nanotopography in the ECM and were used to screen the responses of human endothelial colony-forming cells (hECFCs). In this protocol, we describe the step-by-step procedure of fabricating gradient nanopattern plates for cell engineering, techniques of cultivating hECFCs from human peripheral blood, and culturing hECFCs on nanopattern plates.
Collapse
Affiliation(s)
- Dae Hwan Kim
- School of Biomedical Engineering, Korea University
| | - Long-Hui Cui
- Department of Cardiology, Korea University Anam Hospital
| | - Ha-Rim Seo
- Department of Cardiology, Korea University Anam Hospital
| | - Hyung Joon Joo
- Department of Cardiology, Korea University Anam Hospital
| | | | - Do-Sun Lim
- Department of Cardiology, Korea University Anam Hospital;
| | - Kyu Back Lee
- School of Biomedical Engineering, Korea University;
| |
Collapse
|
291
|
Blache U, Ehrbar M. Inspired by Nature: Hydrogels as Versatile Tools for Vascular Engineering. Adv Wound Care (New Rochelle) 2018; 7:232-246. [PMID: 29984113 PMCID: PMC6032659 DOI: 10.1089/wound.2017.0760] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 10/22/2017] [Indexed: 12/21/2022] Open
Abstract
Significance: Diseases related to vascular malfunction, hyper-vascularization, or lack of vascularization are among the leading causes of morbidity and mortality. Engineered, vascularized tissues as well as angiogenic growth factor-releasing hydrogels could replace defective tissues. Further, treatments and testing of novel vascular therapeutics will benefit significantly from models that allow for the study of vascularized tissues under physiological relevant in vitro conditions. Recent Advances: Inspired by fibrin, the provisional matrix during wound healing, naturally derived and synthetic hydrogel scaffolds have been developed for vascular engineering. Today, engineers and biologists use commercially available hydrogels to pre-vascularize tissues, to control the delivery of angiogenic growth factors, and to establish vascular diseases models. Critical Issue: For clinical translation, pre-vascularized tissue constructs must be sufficiently large and stable to substitute function-relevant tissue defects and integrate with host vascular perfusion. Moreover, the continuous integration of knowhow from basic vascular biology with innovative, tailorable materials and advanced manufacturing technologies is key to achieving near-physiological tissue models and new treatments to control vascularization. Future Directions: For transplantation, engineered tissues must comprise hierarchically organized vascular trees of different caliber and function. The development of novel vascularization-promoting or -inhibiting therapeutics will benefit from physiologically relevant vessel models. In addition, tissue models representing treatment-relevant vascular tissue functions will increase the capacity to screen for therapeutic compounds and will significantly reduce the need for animals for their validation.
Collapse
Affiliation(s)
- Ulrich Blache
- Department of Obstetrics, University and University Hospital Zurich, Zurich, Switzerland
- Department of Health Sciences and Technology, Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Martin Ehrbar
- Department of Obstetrics, University and University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
292
|
He C, Yang Z, Jin Y, Qi X, Chu J, Deng X. ADM Scaffolds Generate a Pro-regenerative Microenvironment During Full-Thickness Cutaneous Wound Healing Through M2 Macrophage Polarization via Lamtor1. Front Physiol 2018; 9:657. [PMID: 29915541 PMCID: PMC5994424 DOI: 10.3389/fphys.2018.00657] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 05/14/2018] [Indexed: 12/21/2022] Open
Abstract
Adult mammalian skin has a defective regenerative capacity following full-thickness cutaneous injury; this defect overshadows the complete physiological functions of the skin. Immune-mediated skin reconstruction driven by biological scaffolds is a recently developed innovative repair strategy to support regenerative wound healing. However, to date, little is known about how biological scaffolds orchestrate the immune response to promote regeneration. Here, using acellular dermal matrix (ADM) scaffolds, we discovered that the default pro-inflammatory response was altered in response to a pro-regenerative response characterized by specific M2 polarization. M2 macrophages subsequently produced a series of wound healing factors, including matrix metalloproteinases (Mmps), and growth factors which promoted cell proliferation, stabilized angiogenesis, and remodeled the extracellular matrix. Our investigations further revealed that the M2 polarization of macrophages arose from an ADM scaffold-derived amino acid sufficiency signal by collagen degradation via macrophage phagocytosis, which activated the acid-sensing pathway (v-ATPase, Lamtor1, and mTORC1). Lamtor1, the acid-sensing pathway-associated lysosomal adaptor protein was critical for inducing M2 polarization, while with the presence of extracellular interleukin 4 (IL4). Our results suggest that ADM scaffolds generate a pro-regenerative microenvironment during full-thickness cutaneous wound healing through M2 macrophage polarization via Lamtor1.
Collapse
Affiliation(s)
- Chengmin He
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Zhi Yang
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Ying Jin
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Xiaoyang Qi
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Jin Chu
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| | - Xiaoyuan Deng
- MOE Key Laboratory of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, China
| |
Collapse
|
293
|
Hielscher D, Kaebisch C, Braun BJV, Gray K, Tobiasch E. Stem Cell Sources and Graft Material for Vascular Tissue Engineering. Stem Cell Rev Rep 2018; 14:642-667. [DOI: 10.1007/s12015-018-9825-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
294
|
Kukumberg M, Yao Y, Goh SH, Neo DJ, Yao JY, Yim EK. Evaluation of the topographical influence on the cellular behavior of human umbilical vein endothelial cells. ADVANCED BIOSYSTEMS 2018; 2:1700217. [PMID: 30766915 PMCID: PMC6370334 DOI: 10.1002/adbi.201700217] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Indexed: 12/17/2022]
Abstract
Adhesion and proliferation of vascular endothelial cells are important parameters in the endothelialization of biomedical devices for vascular applications. Endothelialization is a complex process affected by endothelial cells and their interaction with the extracellular microenvironment. Although numerous approaches are taken to study the influence of the external environment, a systematic investigation of the impact of an engineered microenvironment on endothelial cell processes is needed. This study aims to investigate the influence of topography, initial cell seeding density, and collagen coating on human umbilical vein endothelial cells (HUVECs). Utilizing the MultiARChitecture (MARC) chamber, the effects of various topographies on HUVECs are identified, and those with more prominent effects were further evaluated individually using the MARC plate. Endothelial cell marker expression and monocyte adhesion assay are examined on the HUVEC monolayer. HUVECs on 1.8 μm convex and concave microlens topographies demonstrate the lowest cell adhesion and proliferation, regardless of initial cell seeding density and collagen I coating, and the HUVEC monolayer on the microlens shows the lowest monocyte adhesion. This property of lens topographies would potentially be a useful parameter in designing vascular biomedical devices. The MARC chamber and MARC plate show a great potential for faster and easy pattern identification for various cellular processes.
Collapse
Affiliation(s)
- Marek Kukumberg
- Mechanobiology Institute, National University of Singapore, #05-01 T-lab, 5A Engineering Drive 1, Singapore 117411
| | - Yuan Yao
- Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| | - Seok Hong Goh
- Institute of Materials Research and Engineering, Agency for Science, Technology and Research (ASTAR), 2 Fusionopolis Way, Innovis, 138634, Singapore, Department of Biomedical Engineering, National University of Singapore, E4, #04-10,4 Engineering Drive 3, Singapore 117583
| | - Dawn Jh Neo
- Mechanobiology Institute, National University of Singapore, #05-01 T-lab, 5A Engineering Drive 1, Singapore 117411
| | - Jia Yi Yao
- Department of Biomedical Engineering, National University of Singapore, E4, #04-10,4 Engineering Drive 3, Singapore 117583
| | - Evelyn Kf Yim
- Mechanobiology Institute, National University of Singapore, #05-01 T-lab, 5A Engineering Drive 1, Singapore 117411, Department of Biomedical Engineering, National University of Singapore, E4, #04-10,4 Engineering Drive 3, Singapore 117583, Department of Surgery, National University of Singapore, NUHS Tower Block, Level 8,1E Kent Ridge Road, Singapore 119228, Department of Chemical Engineering, University of Waterloo, 200 University Avenue West, Waterloo, Ontario, N2L 3G1, Canada
| |
Collapse
|
295
|
Song L, Ahmed MF, Li Y, Zeng C, Li Y. Vascular differentiation from pluripotent stem cells in 3-D auxetic scaffolds. J Tissue Eng Regen Med 2018; 12:1679-1689. [PMID: 29749038 DOI: 10.1002/term.2695] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 04/10/2018] [Accepted: 05/03/2018] [Indexed: 12/29/2022]
Abstract
Auxetic scaffolds, that is, scaffolds that can display negative Poisson's ratio, have unique physical properties and can expand transversally when axially strained or contract under compression. Auxetic materials have been used for bioprostheses and artery stents due to the enhanced compressive strength and shear stiffness. In vascular tissue engineering, auxetic scaffolds allow the widening of blood vessels when blood flows through (creating compressive stress) to prevent the blockage. However, the influence of auxetic materials on the cellular fate decision in local environment is unclear. In this study, auxetic polyurethane foams were used to support vascular differentiation from pluripotent stem cells. The expression of alkaline phosphatase, Oct-4, and Nanog was lower after 4 days of differentiation for the cells grown in auxetic scaffolds. Higher expression of vascular markers CD31 and VE-cadherin was observed for the cells from auxetic scaffolds compared with those from the scaffolds before auxetic conversion. Little influence on the expression of cardiac marker α-actinin was observed. The vascular cells secreted extracellular matrix proteins vitronectin and laminin and expressed membrane-bound matrix metalloproteinase 9. The examination of Yes-associated protein expression indicated more cytoplasmic retention in the cells from auxetic scaffolds compared with those from regular scaffolds, suggesting that the auxetic scaffolds may affect cellular contraction. This study demonstrates a novel 3-D culture based on auxetic scaffolds for vascular differentiation and provides a platform to study the influence of biophysical microenvironments on differentiation of pluripotent stem cells. The outcome of this study has implications for regenerative medicine and drug discovery.
Collapse
Affiliation(s)
- Liqing Song
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| | - Mohammad Faisal Ahmed
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA.,High-Performance Materials Institute, Florida State University, Tallahassee, FL, USA
| | - Yan Li
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA.,High-Performance Materials Institute, Florida State University, Tallahassee, FL, USA
| | - Changchun Zeng
- Department of Industrial and Manufacturing Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA.,High-Performance Materials Institute, Florida State University, Tallahassee, FL, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL, USA
| |
Collapse
|
296
|
Importance of extracellular matrix and growth state for the EA.hy926 endothelial cell response to polyunsaturated fatty acids. PLoS One 2018; 13:e0197613. [PMID: 29763471 PMCID: PMC5953484 DOI: 10.1371/journal.pone.0197613] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 05/04/2018] [Indexed: 12/31/2022] Open
Abstract
Consumption of different PUFAs (polyunsaturated fatty acids) can induce functional changes in blood vessels via endothelial cells, which interact with dietary factors in the circulation. The basement membrane that separates the endothelium from the smooth muscle cells of the medial layer can also influence the functional state of endothelial cells. However, the effect of basement membrane on the endothelial response to dietary PUFAs in relation to growth state (e.g. proliferation versus quiescence) has never been investigated. We therefore compared the viability (CCK kit) and proliferation (bromodeoxyuridine incorporation) of EA.hy926 endothelial cells grown on Matrigel or collagen versus non-coated plates. EA.hy926 viability and proliferation were also assessed after treatment with 0–150 μM of PUFAs [linoleic acid (LA), arachidonic acid (AA), α-linolenic acid (ALA), eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)]. Our study showed that only cells grown on Matrigel-coated plates reached quiescence after becoming confluent with a decreased level of MCM2 and p-cyclin D1 (T286), increased levels of p27kip1 and a low level of apoptosis and senescence. AA, EPA and DHA decreased the viability and proliferation of subconfluent cells grown on plastic dishes in a dose-dependent manner, while the presence of Matrigel made the cells resistant to these adverse effects. Confluent cell viability was less sensitive to higher concentrations of AA, EPA and DHA than subconfluent cells, and a significant increase in caspase-3 cleavage was only observed in confluent cells treated with DHA. Higher concentrations of AA, EPA and DHA suppressed DNA synthesis by both subconfluent and confluent cells, while precursor C18 PUFAs (LA and ALA) had no negative effects on viability and proliferation. Our study is the first to show that extracellular matrix and growth state are important factors in the EA.hy926 cell response to PUFAs, and that the mechanisms by which individual PUFAs operate may be growth state-dependent.
Collapse
|
297
|
Kurokawa YK, Yin RT, Shang MR, Shirure VS, Moya ML, George SC. Human Induced Pluripotent Stem Cell-Derived Endothelial Cells for Three-Dimensional Microphysiological Systems. Tissue Eng Part C Methods 2018. [PMID: 28622076 DOI: 10.1089/ten.tec.2017.0133] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Microphysiological systems (MPS), or "organ-on-a-chip" platforms, aim to recapitulate in vivo physiology using small-scale in vitro tissue models of human physiology. While significant efforts have been made to create vascularized tissues, most reports utilize primary endothelial cells that hinder reproducibility. In this study, we report the use of human induced pluripotent stem cell-derived endothelial cells (iPS-ECs) in developing three-dimensional (3D) microvascular networks. We established a CDH5-mCherry reporter iPS cell line, which expresses the vascular endothelial (VE)-cadherin fused to mCherry. The iPS-ECs demonstrate physiological functions characteristic of primary endothelial cells in a series of in vitro assays, including permeability, response to shear stress, and the expression of endothelial markers (CD31, von Willibrand factor, and endothelial nitric oxide synthase). The iPS-ECs form stable, perfusable microvessels over the course of 14 days when cultured within 3D microfluidic devices. We also demonstrate that inhibition of TGF-β signaling improves vascular network formation by the iPS-ECs. We conclude that iPS-ECs can be a source of endothelial cells in MPS providing opportunities for human disease modeling and improving the reproducibility of 3D vascular networks.
Collapse
Affiliation(s)
- Yosuke K Kurokawa
- 1 Department of Biomedical Engineering, Washington University in St. Louis , St. Louis, Missouri
| | - Rose T Yin
- 1 Department of Biomedical Engineering, Washington University in St. Louis , St. Louis, Missouri
| | - Michael R Shang
- 1 Department of Biomedical Engineering, Washington University in St. Louis , St. Louis, Missouri
| | - Venktesh S Shirure
- 1 Department of Biomedical Engineering, Washington University in St. Louis , St. Louis, Missouri
| | - Monica L Moya
- 2 Center for Micro and Nano Technology, Materials Engineering Division, Lawrence Livermore National Laboratory, Livermore, California
| | - Steven C George
- 1 Department of Biomedical Engineering, Washington University in St. Louis , St. Louis, Missouri
- 3 Department of Energy, Environment, and Chemical Engineering, Washington University in St. Louis , St. Louis, Missouri
| |
Collapse
|
298
|
Haggerty AE, Maldonado-Lasunción I, Oudega M. Biomaterials for revascularization and immunomodulation after spinal cord injury. ACTA ACUST UNITED AC 2018; 13:044105. [PMID: 29359704 DOI: 10.1088/1748-605x/aaa9d8] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Spinal cord injury (SCI) causes immediate damage to the nervous tissue accompanied by loss of motor and sensory function. The limited self-repair competence of injured nervous tissue underscores the need for reparative interventions to recover function after SCI. The vasculature of the spinal cord plays a crucial role in SCI and repair. Ruptured and sheared blood vessels in the injury epicenter and blood vessels with a breached blood-spinal cord barrier (BSCB) in the surrounding tissue cause bleeding and inflammation, which contribute to the overall tissue damage. The insufficient formation of new functional vasculature in and near the injury impedes endogenous tissue repair and limits the prospect of repair approaches. Limiting the loss of blood vessels, stabilizing the BSCB, and promoting the formation of new blood vessels are therapeutic targets for spinal cord repair. Inflammation is an integral part of injury-mediated vascular damage, which has deleterious and reparative consequences. Inflammation and the formation of new blood vessels are intricately interwoven. Biomaterials can be effectively used for promoting and guiding blood vessel formation or modulating the inflammatory response after SCI, thereby governing the extent of damage and the success of reparative interventions. This review deals with the vasculature after SCI, the reciprocal interactions between inflammation and blood vessel formation, and the potential of biomaterials to support revascularization and immunomodulation in damaged spinal cord nervous tissue.
Collapse
Affiliation(s)
- Agnes E Haggerty
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | | | | |
Collapse
|
299
|
Osaki T, Serrano JC, Kamm RD. Cooperative Effects of Vascular Angiogenesis and Lymphangiogenesis. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2018; 4:120-132. [PMID: 30417074 DOI: 10.1007/s40883-018-0054-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
In this study, we modeled lymphangiogenesis and vascular angiogenesis in a microdevice using a tissue engineering approach. Lymphatic vessels (LV) and blood vessels (BV) were fabricated by sacrificial molding with seeding human lymphatic endothelial cells and human umbilical vein endothelial cells into molded microchannels (600 μm diameter). During subsequent perfusion culture, lymphangiogenesis and vascular angiogenesis were induced by addition of phorbol 12-myristate 13-acetate (PMA) and VEGF-C or VEGF-A characterized by podoplanin and Prox-1 expression. The lymphatic capillaries formed button-like junctions treated with dexamethasone. To test the potential for screening anti-angiogenic (vascular and lymphatic) factors, antagonists of VEGF were introduced. We found that an inhibitor of VEGF-R3 did not completely suppress lymphatic angiogenesis with BVs present, although lymphatic angiogenesis was selectively prevented by addition of a VEGF-R3 inhibitor without BVs. To probe the mechanism of action, we focus on matrix metalloproteinase (MMP) secretion by vascular endothelial cells and lymphatic endothelial cells under monoculture or co-culture conditions. We found that vascular angiogenesis facilitated lymphangiogenesis via remodeling of the local microenvironment by the increased secretion of MMP, mainly by endothelial cells. Applications of this model include a drug screening assay for corneal disease and models for tumorigenesis including lymphatic angiogenesis and vascular angiogenesis.
Collapse
Affiliation(s)
- Tatsuya Osaki
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jean C Serrano
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roger D Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,BioSystems and Micromechanics (BioSyM), Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| |
Collapse
|
300
|
Developmental Pathways Pervade Stem Cell Responses to Evolving Extracellular Matrices of 3D Bioprinted Microenvironments. Stem Cells Int 2018; 2018:4809673. [PMID: 29765414 PMCID: PMC5896227 DOI: 10.1155/2018/4809673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/10/2017] [Accepted: 12/10/2017] [Indexed: 12/17/2022] Open
Abstract
Developmental studies and 3D in vitro model systems show that the production and engagement of extracellular matrix (ECM) often precede stem cell differentiation. Yet, unclear is how the ECM triggers signaling events in sequence to accommodate multistep process characteristic of differentiation. Here, we employ transcriptome profiling and advanced imaging to delineate the specificity of ECM engagement to particular differentiation pathways and to determine whether specificity in this context is a function of long-term ECM remodeling. To this end, human mesenchymal stem cells (hMSCs) were cultured in 3D bioprinted prisms created from ECM proteins and associated controls. We found that exogenous ECM provided in 3D microenvironments at early time points impacts on the composition of microenvironments at later time points and that each evolving 3D microenvironment is uniquely poised to promote stem cell differentiation. Moreover, 2D cultures undergo minimal ECM remodeling and are ill-equipped to stimulate pathways associated with development.
Collapse
|