1
|
Krymchenko R, Coşar Kutluoğlu G, van Hout N, Manikowski D, Doberenz C, van Kuppevelt TH, Daamen WF. Elastogenesis in Focus: Navigating Elastic Fibers Synthesis for Advanced Dermal Biomaterial Formulation. Adv Healthc Mater 2024; 13:e2400484. [PMID: 38989717 DOI: 10.1002/adhm.202400484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/31/2024] [Indexed: 07/12/2024]
Abstract
Elastin, a fibrous extracellular matrix (ECM) protein, is the main component of elastic fibers that are involved in tissues' elasticity and resilience, enabling them to undergo reversible extensibility and to endure repetitive mechanical stress. After wounding, it is challenging to regenerate elastic fibers and biomaterials developed thus far have struggled to induce its biosynthesis. This review provides a comprehensive summary of elastic fibers synthesis at the cellular level and its implications for biomaterial formulation, with a particular focus on dermal substitutes. The review delves into the intricate process of elastogenesis by cells and investigates potential triggers for elastogenesis encompassing elastin-related compounds, ECM components, and other molecules for their potential role in inducing elastin formation. Understanding of the elastogenic processes is essential for developing biomaterials that trigger not only the synthesis of the elastin protein, but also the formation of a functional and branched elastic fiber network.
Collapse
Affiliation(s)
- Roman Krymchenko
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Gizem Coşar Kutluoğlu
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
- MedSkin Solutions Dr. Suwelack AG, 48727, Billerbeck, Germany
| | - Noor van Hout
- Department of Dermatology, Radboud university medical center, Nijmegen, 6525 GA, The Netherlands
| | | | | | - Toin H van Kuppevelt
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Willeke F Daamen
- Department of Medical BioSciences, Research Institute for Medical Innovation, Radboud university medical center, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| |
Collapse
|
2
|
Fakhri N, Khalili A, Sachlos T, Rezai P. Fabrication of Porous Collagen Scaffolds Containing Embedded Channels with Collagen Membrane Linings. MICROMACHINES 2024; 15:1031. [PMID: 39203682 PMCID: PMC11356104 DOI: 10.3390/mi15081031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024]
Abstract
Tissues and organs contain an extracellular matrix (ECM). In the case of blood vessels, endothelium cells are anchored to a specialized basement membrane (BM) embedded inside the interstitial matrix (IM). We introduce a multi-structural collagen-based scaffold with embedded microchannels that mimics in vivo structures within vessels. Our scaffold consists of two parts, each containing two collagen layers, i.e., a 3D porous collagen layer analogous to IM lined with a thin 2D collagen film resembling the BM. Enclosed microchannels were fabricated using contact microprinting. Microchannel test structures with different sizes ranging from 300 to 800 µm were examined for their fabrication reproducibility. The heights and perimeters of the fabricated microchannels were ~20% less than their corresponding values in the replication PDMS mold; however, microchannel widths were significantly closer to their replica dimensions. The stiffness, permeability, and pore size properties of the 2D and 3D collagen layers were measured. The permeability of the 2D collagen film was negligible, making it suitable for mimicking the BM of large blood vessels. A leakage test at various volumetric flow rates applied to the microchannels showed no discharge, thereby verifying the reliability of the proposed integrated 2D/3D collagen parts and the contact printing method used for bonding them in the scaffold. In the future, multi-cell culturing will be performed within the 3D porous collagen and against the 2D membrane inside the microchannel, hence preparing this scaffold for studying a variety of blood vessel-tissue interfaces. Also, thicker collagen scaffold tissues will be fabricated by stacking several layers of the proposed scaffold.
Collapse
Affiliation(s)
| | | | - Terry Sachlos
- Department of Mechanical Engineering, York University, Toronto, ON M3J 1P3, Canada
| | - Pouya Rezai
- Department of Mechanical Engineering, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
3
|
Rosellini E, Giordano C, Guidi L, Cascone MG. Biomimetic Approaches in Scaffold-Based Blood Vessel Tissue Engineering. Biomimetics (Basel) 2024; 9:377. [PMID: 39056818 PMCID: PMC11274842 DOI: 10.3390/biomimetics9070377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/15/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Cardiovascular diseases remain a leading cause of mortality globally, with atherosclerosis representing a significant pathological means, often leading to myocardial infarction. Coronary artery bypass surgery, a common procedure used to treat coronary artery disease, presents challenges due to the limited autologous tissue availability or the shortcomings of synthetic grafts. Consequently, there is a growing interest in tissue engineering approaches to develop vascular substitutes. This review offers an updated picture of the state of the art in vascular tissue engineering, emphasising the design of scaffolds and dynamic culture conditions following a biomimetic approach. By emulating native vessel properties and, in particular, by mimicking the three-layer structure of the vascular wall, tissue-engineered grafts can improve long-term patency and clinical outcomes. Furthermore, ongoing research focuses on enhancing biomimicry through innovative scaffold materials, surface functionalisation strategies, and the use of bioreactors mimicking the physiological microenvironment. Through a multidisciplinary lens, this review provides insight into the latest advancements and future directions of vascular tissue engineering, with particular reference to employing biomimicry to create systems capable of reproducing the structure-function relationships present in the arterial wall. Despite the existence of a gap between benchtop innovation and clinical translation, it appears that the biomimetic technologies developed to date demonstrate promising results in preventing vascular occlusion due to blood clotting under laboratory conditions and in preclinical studies. Therefore, a multifaceted biomimetic approach could represent a winning strategy to ensure the translation of vascular tissue engineering into clinical practice.
Collapse
Affiliation(s)
- Elisabetta Rosellini
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy; (C.G.); (L.G.)
| | | | | | - Maria Grazia Cascone
- Department of Civil and Industrial Engineering, University of Pisa, Largo Lucio Lazzarino 1, 56122 Pisa, Italy; (C.G.); (L.G.)
| |
Collapse
|
4
|
Fu Y, Zhou Y, Wang K, Li Z, Kong W. Extracellular Matrix Interactome in Modulating Vascular Homeostasis and Remodeling. Circ Res 2024; 134:931-949. [PMID: 38547250 DOI: 10.1161/circresaha.123.324055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Abstract
The ECM (extracellular matrix) is a major component of the vascular microenvironment that modulates vascular homeostasis. ECM proteins include collagens, elastin, noncollagen glycoproteins, and proteoglycans/glycosaminoglycans. ECM proteins form complex matrix structures, such as the basal lamina and collagen and elastin fibers, through direct interactions or lysyl oxidase-mediated cross-linking. Moreover, ECM proteins directly interact with cell surface receptors or extracellular secreted molecules, exerting matricellular and matricrine modulation, respectively. In addition, extracellular proteases degrade or cleave matrix proteins, thereby contributing to ECM turnover. These interactions constitute the ECM interactome network, which is essential for maintaining vascular homeostasis and preventing pathological vascular remodeling. The current review mainly focuses on endogenous matrix proteins in blood vessels and discusses the interaction of these matrix proteins with other ECM proteins, cell surface receptors, cytokines, complement and coagulation factors, and their potential roles in maintaining vascular homeostasis and preventing pathological remodeling.
Collapse
Affiliation(s)
- Yi Fu
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Yuan Zhou
- Department of Biomedical Informatics (Y.Z.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Kai Wang
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Zhuofan Li
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology (Y.F., K.W., Z.L., W.K.), School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
5
|
Baldwin CS, Iyer S, Rao RR. The challenges and prospects of smooth muscle tissue engineering. Regen Med 2024; 19:135-143. [PMID: 38440898 PMCID: PMC10941056 DOI: 10.2217/rme-2023-0230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/16/2024] [Indexed: 03/06/2024] Open
Abstract
Many vascular disorders arise as a result of dysfunctional smooth muscle cells. Tissue engineering strategies have evolved as key approaches to generate functional vascular smooth muscle cells for use in cell-based precision and personalized regenerative medicine approaches. This article highlights some of the challenges that exist in the field and presents some of the prospects for translating research advancements into therapeutic modalities. The article emphasizes the need for better developing synergetic intracellular and extracellular cues in the processes to generate functional vascular smooth muscle cells from different stem cell sources for use in tissue engineering strategies.
Collapse
Affiliation(s)
- Christofer S Baldwin
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| | - Shilpa Iyer
- Department of Biological Sciences, Fulbright College of Arts & Sciences, University of Arkansas, Fayetteville, AR 72701, USA
| | - Raj R Rao
- Department of Biomedical Engineering, College of Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
6
|
Bhattacharjee A, Savargaonkar AV, Tahir M, Sionkowska A, Popat KC. Surface modification strategies for improved hemocompatibility of polymeric materials: a comprehensive review. RSC Adv 2024; 14:7440-7458. [PMID: 38433935 PMCID: PMC10906639 DOI: 10.1039/d3ra08738g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Abstract
Polymeric biomaterials are a widely used class of materials due to their versatile properties. However, as with all other types of materials used for biomaterials, polymers also have to interact with blood. When blood comes into contact with any foreign body, it initiates a cascade which leads to platelet activation and blood coagulation. The implant surface also has to encounter a thromboinflammatory response which makes the implant integrity vulnerable, this leads to blood coagulation on the implant and obstructs it from performing its function. Hence, the surface plays a pivotal role in the design and application of biomaterials. In particular, the surface properties of biomaterials are responsible for biocompatibility with biological systems and hemocompatibility. This review provides a report on recent advances in the field of surface modification approaches for improved hemocompatibility. We focus on the surface properties of polysaccharides, proteins, and synthetic polymers. The blood coagulation cascade has been discussed and blood - material surface interactions have also been explained. The interactions of blood proteins and cells with polymeric material surfaces have been discussed. Moreover, the benefits as well as drawbacks of blood coagulation on the implant surface for wound healing purposes have also been studied. Surface modifications implemented by other researchers to enhance as well as prevent blood coagulation have also been analyzed.
Collapse
Affiliation(s)
- Abhishek Bhattacharjee
- School of Advanced Material Discovery, Colorado State University Fort Collins CO 80523 USA
| | | | - Muhammad Tahir
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University Gagarina 7 87-100 Torun Poland
| | - Alina Sionkowska
- Department of Biomaterials and Cosmetic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University Gagarina 7 87-100 Torun Poland
| | - Ketul C Popat
- School of Advanced Material Discovery, Colorado State University Fort Collins CO 80523 USA
- Department of Mechanical Engineering, Colorado State University Fort Collins CO 80523 USA
- Department of Bioengineering, George Mason University Fairfax VA 22030 USA
| |
Collapse
|
7
|
Galindo JM, San-Millán MI, Castillo-Sarmiento CA, Ballesteros-Yáñez I, Vázquez E, Merino S, Herrero MA. Optimization of 3D Synthetic Scaffolds for Neuronal Tissue Engineering Applications. Chemistry 2024; 30:e202302481. [PMID: 37823243 DOI: 10.1002/chem.202302481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/13/2023]
Abstract
The increasing prevalence of neurodegenerative diseases has spurred researchers to develop advanced 3D models that accurately mimic neural tissues. Hydrogels stand out as ideal candidates as their properties closely resemble those of the extracellular matrix. A critical challenge in this regard is to comprehend the influence of the scaffold's mechanical properties on cell growth and differentiation, thus enabling targeted modifications. In light of this, a synthesis and comprehensive analysis of acrylamide-based hydrogels incorporating a peptide has been conducted. Adequate cell adhesion and development is achieved due to their bioactive nature and specific interactions with cellular receptors. The integration of a precisely controlled physicochemical hydrogel matrix and inclusion of the arginine-glycine-aspartic acid peptide sequence has endowed this system with an optimal structure, thus providing a unique ability to interact effectively with biomolecules. The analysis fully examined essential properties governing cell behavior, including pore size, mechanical characteristics, and swelling ability. Cell-viability experiments were performed to assess the hydrogel's biocompatibility, while the incorporation of grow factors aimed to promote the differentiation of neuroblastoma cells. The results underscore the hydrogel's ability to stimulate cell viability and differentiation in the presence of the peptide within the matrix.
Collapse
Affiliation(s)
- Josué M Galindo
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Ms Irene San-Millán
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | | | | | - Ester Vázquez
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Sonia Merino
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - M Antonia Herrero
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| |
Collapse
|
8
|
Bhattacharya A, Alam K, Roy NS, Kaur K, Kaity S, Ravichandiran V, Roy S. Exploring the interaction between extracellular matrix components in a 3D organoid disease model to replicate the pathophysiology of breast cancer. J Exp Clin Cancer Res 2023; 42:343. [PMID: 38102637 PMCID: PMC10724947 DOI: 10.1186/s13046-023-02926-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
In vitro models are necessary to study the pathophysiology of the disease and the development of effective, tailored treatment methods owing to the complexity and heterogeneity of breast cancer and the large population affected by it. The cellular connections and tumor microenvironments observed in vivo are often not recapitulated in conventional two-dimensional (2D) cell cultures. Therefore, developing 3D in vitro models that mimic the complex architecture and physiological circumstances of breast tumors is crucial for advancing our understanding of the illness. A 3D scaffold-free in vitro disease model mimics breast cancer pathophysiology by allowing cells to self-assemble/pattern into 3D structures, in contrast with other 3D models that rely on artificial scaffolds. It is possible that this model, whether applied to breast tumors using patient-derived primary cells (fibroblasts, endothelial cells, and cancer cells), can accurately replicate the observed heterogeneity. The complicated interactions between different cell types are modelled by integrating critical components of the tumor microenvironment, such as the extracellular matrix, vascular endothelial cells, and tumor growth factors. Tissue interactions, immune cell infiltration, and the effects of the milieu on drug resistance can be studied using this scaffold-free 3D model. The scaffold-free 3D in vitro disease model for mimicking tumor pathophysiology in breast cancer is a useful tool for studying the molecular basis of the disease, identifying new therapeutic targets, and evaluating treatment modalities. It provides a more physiologically appropriate high-throughput platform for screening large compound library in a 96-384 well format. We critically discussed the rapid development of personalized treatment strategies and accelerated drug screening platforms to close the gap between traditional 2D cell culture and in vivo investigations.
Collapse
Affiliation(s)
- Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
9
|
Kang L, Zhou Y, Chen X, Yue Z, Liu X, Baker C, Wallace GG. Fabrication and Characterization of an Electro-Compacted Collagen/Elastin/Hyaluronic Acid Sheet as a Potential Skin Scaffold. Macromol Biosci 2023; 23:e2300220. [PMID: 37589999 DOI: 10.1002/mabi.202300220] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/14/2023] [Indexed: 08/18/2023]
Abstract
The development of biomimetic structures with integrated extracellular matrix (ECM) components represents a promising approach to biomaterial fabrication. Here, an artificial ECM, comprising the structural protein collagen I and elastin (ELN), as well as the glycosaminoglycan hyaluronan (HA), is reported. Specifically, collagen and ELN are electrochemically aligned to mimic the compositional characteristics of the dermal matrix. HA is incorporated into the electro-compacted collagen-ELN matrices via adsorption and chemical immobilization, to give a final composition of collagen/ELN/HA of 7:2:1. This produces a final collagen/ELN/hyaluronic acid scaffold (CEH) that recapitulates the compositional feature of the native skin ECM. This study analyzes the effect of CEH composition on the cultivation of human dermal fibroblast cells (HDFs) and immortalized human keratinocytes (HaCaTs). It is shown that the CEH scaffold supports dermal regeneration by promoting HDFs proliferation, ECM deposition, and differentiation into myofibroblasts. The CEH scaffolds are also shown to support epidermis growth by supporting HaCaTs proliferation, differentiation, and stratification. A double-layered epidermal-dermal structure is constructed on the CEH scaffold, further demonstrating its ability in supporting skin cell function and skin regeneration.
Collapse
Affiliation(s)
- Lingzhi Kang
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Ying Zhou
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Xifang Chen
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Zhilian Yue
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Xiao Liu
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Wollongong, NSW, 2522, Australia
| | - Chris Baker
- Department of Dermatology, St Vincent's Hospital Melbourne, Melbourne, VIC, 3065, Australia
- Department of Medicine (Dermatology), University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Gordon G Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM Facility, Innovation Campus, University of Wollongong, Wollongong, NSW, 2522, Australia
| |
Collapse
|
10
|
Do NT, Lee SY, Lee YS, Shin C, Kim D, Lee TG, Son JG, Kim SH. Time-sequential fibroblast-to-myofibroblast transition in elastin-variable 3D hydrogel environments by collagen networks. Biomater Res 2023; 27:103. [PMID: 37848974 PMCID: PMC10583321 DOI: 10.1186/s40824-023-00439-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/25/2023] [Indexed: 10/19/2023] Open
Abstract
BACKGROUND Fibrosis plays an important role in both normal physiological and pathological phenomena as fibroblasts differentiate to myofibroblasts. The activation of fibroblasts is determined through interactions with the surrounding extracellular matrix (ECM). However, how this fibroblast-to-myofibroblast transition (FMT) is regulated and affected by elastin concentration in a three-dimensional (3D) microenvironment has not been investigated. METHODS We developed an insoluble elastin-gradient 3D hydrogel system for long-lasting cell culture and studied the molecular mechanisms of the FMT in embedded cells by nanoflow LC-MS/MS analysis along with validation through real-time PCR and immunofluorescence staining. RESULTS By optimizing pH and temperature, four 3D hydrogels containing fibroblasts were successfully fabricated having elastin concentrations of 0, 20, 50, and 80% in collagen. At the low elastin level (20%), fibroblast proliferation was significantly increased compared to others, and in particular, the FMT was clearly observed in this condition. Moreover, through mass spectrometry of the hydrogel environment, it was confirmed that differentiation proceeded in two stages. In the early stage, calcium-dependent proteins including calmodulin and S100A4 were highly associated. On the other hand, in the late stage after several passages of cells, distinct markers of myofibroblasts were presented such as morphological changes, increased production of ECM, and increased α-SMA expression. We also demonstrated that the low level of elastin concentration induced some cancer-associated fibroblast (CAF) markers, including PDGFR-β, and fibrosis-related disease markers, including THY-1. CONCLUSION Using our developed 3D elastin-gradient hydrogel system, we evaluated the effect of different elastin concentrations on the FMT. The FMT was induced even at a low concentration of elastin with increasing CAF level via calcium signaling. With this system, we were able to analyze varying protein expressions in the overall FMT process over several cellular passages. Our results suggest that the elastin-gradient system employing nonlinear optics imaging provides a good platform to study activated fibroblasts interacting with the microenvironment, where the ECM plays a pivotal role.
Collapse
Affiliation(s)
- Nhuan T Do
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
- BioMedical Measurement, University of Science and Technology, 217 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Sun Young Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Yoon Seo Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - ChaeHo Shin
- Interdisciplinary Materials Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
- Nanoconvergence Measurement, University of Science and Technology, 217 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Daeho Kim
- Bruker Nano Surface & Metrology, Bruker Korea, Seongnam, 13493, Republic of Korea
| | - Tae Geol Lee
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
- Nanoconvergence Measurement, University of Science and Technology, 217 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea
| | - Jin Gyeong Son
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea.
| | - Se-Hwa Kim
- Safety Measurement Institute, Korea Research Institute of Standards and Science, 267 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea.
- BioMedical Measurement, University of Science and Technology, 217 Gajeong-Ro, Yuseong-Gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
11
|
Ando Y, Chang FC, James M, Zhou Y, Zhang M. Chitosan Scaffolds as Microcarriers for Dynamic Culture of Human Neural Stem Cells. Pharmaceutics 2023; 15:1957. [PMID: 37514142 PMCID: PMC10384976 DOI: 10.3390/pharmaceutics15071957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Human neural stem cells (hNSCs) possess remarkable potential for regenerative medicine in the treatment of presently incurable diseases. However, a key challenge lies in producing sufficient quantities of hNSCs, which is necessary for effective treatment. Dynamic culture systems are recognized as a powerful approach to producing large quantities of hNSCs required, where microcarriers play a critical role in supporting cell expansion. Nevertheless, the currently available microcarriers have limitations, including a lack of appropriate surface chemistry to promote cell adhesion, inadequate mechanical properties to protect cells from dynamic forces, and poor suitability for mass production. Here, we present the development of three-dimensional (3D) chitosan scaffolds as microcarriers for hNSC expansion under defined conditions in bioreactors. We demonstrate that chitosan scaffolds with a concentration of 4 wt% (4CS scaffolds) exhibit desirable microstructural characteristics and mechanical properties suited for hNSC expansion. Furthermore, they could also withstand degradation in dynamic conditions. The 4CS scaffold condition yields optimal metabolic activity, cell adhesion, and protein expression, enabling sustained hNSC expansion for up to three weeks in a dynamic culture. Our study introduces an effective microcarrier approach for prolonged expansion of hNSCs, which has the potential for mass production in a three-dimensional setting.
Collapse
Affiliation(s)
- Yoshiki Ando
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Materials Department, Medical R&D Center, Corporate R&D Group, KYOCERA Corporation, Yasu 520-2362, Shiga, Japan
| | - Fei-Chien Chang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Matthew James
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Yang Zhou
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
12
|
Karimi A, Khan S, Razaghi R, Rahmati SM, Gathara M, Tudisco E, Aga M, Kelley MJ, Jian Y, Acott TS. Developing an experimental-computational workflow to study the biomechanics of the human conventional aqueous outflow pathway. Acta Biomater 2023; 164:346-362. [PMID: 37072067 PMCID: PMC10226761 DOI: 10.1016/j.actbio.2023.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/29/2023] [Accepted: 04/06/2023] [Indexed: 04/20/2023]
Abstract
The aqueous humor actively interacts with the trabecular meshwork (TM), juxtacanalicular tissue (JCT), and Schlemm's canal (SC) through a dynamic fluid-structure interaction (FSI) coupling. Despite the fact that intraocular pressure (IOP) undergoes significant fluctuations, our understanding of the hyperviscoelastic biomechanical properties of the aqueous outflow tissues is limited. In this study, a quadrant of the anterior segment from a normal human donor eye was dynamically pressurized in the SC lumen, and imaged using a customized optical coherence tomography (OCT). The TM/JCT/SC complex finite element (FE) with embedded collagen fibrils was reconstructed based on the segmented boundary nodes in the OCT images. The hyperviscoelastic mechanical properties of the outflow tissues' extracellular matrix with embedded viscoelastic collagen fibrils were calculated using an inverse FE-optimization method. Thereafter, the 3D microstructural FE model of the TM, with adjacent JCT and SC inner wall, from the same donor eye was constructed using optical coherence microscopy and subjected to a flow load-boundary from the SC lumen. The resultant deformation/strain in the outflow tissues was calculated using the FSI method, and compared to the digital volume correlation (DVC) data. TM showed larger shear modulus (0.92 MPa) compared to the JCT (0.47 MPa) and SC inner wall (0.85 MPa). Shear modulus (viscoelastic) was larger in the SC inner wall (97.65 MPa) compared to the TM (84.38 MPa) and JCT (56.30 MPa). The conventional aqueous outflow pathway is subjected to a rate-dependent IOP load-boundary with large fluctuations. This necessitates addressing the biomechanics of the outflow tissues using hyperviscoelastic material-model. STATEMENT OF SIGNIFICANCE: While the human conventional aqueous outflow pathway is subjected to a large-deformation and time-dependent IOP load-boundary, we are not aware of any studies that have calculated the hyperviscoelastic mechanical properties of the outflow tissues with embedded viscoelastic collagen fibrils. A quadrant of the anterior segment of a normal humor donor eye was dynamically pressurized from the SC lumen with relatively large fluctuations. The TM/JCT/SC complex were OCT imaged and the mechanical properties of the tissues with embedded collagen fibrils were calculated using the inverse FE-optimization algorithm. The resultant displacement/strain in the FSI outflow model was validated versus the DVC data. The proposed experimental-computational workflow may significantly contribute to understanding of the effects of different drugs on the biomechanics of the conventional aqueous outflow pathway.
Collapse
Affiliation(s)
- Alireza Karimi
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Shanjida Khan
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Reza Razaghi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Michael Gathara
- Department of Computer Science, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Erika Tudisco
- Division of Geotechnical Engineering, Lund University, Lund, Sweden
| | - Mini Aga
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Mary J Kelley
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA; Department Integrative Biosciences, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
| | - Yifan Jian
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA; Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, USA
| | - Ted S Acott
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA; Department Chemical Physiology & Biochemistry, School of Medicine, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
13
|
Liu W, LeBar K, Roth K, Pang J, Ayers J, Chicco AJ, Puttlitz CM, Wang Z. Alterations of biaxial viscoelastic properties of the right ventricle in pulmonary hypertension development in rest and acute stress conditions. Front Bioeng Biotechnol 2023; 11:1182703. [PMID: 37324443 PMCID: PMC10266205 DOI: 10.3389/fbioe.2023.1182703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction: The right ventricle (RV) mechanical property is an important determinant of its function. However, compared to its elasticity, RV viscoelasticity is much less studied, and it remains unclear how pulmonary hypertension (PH) alters RV viscoelasticity. Our goal was to characterize the changes in RV free wall (RVFW) anisotropic viscoelastic properties with PH development and at varied heart rates. Methods: PH was induced in rats by monocrotaline treatment, and the RV function was quantified by echocardiography. After euthanasia, equibiaxial stress relaxation tests were performed on RVFWs from healthy and PH rats at various strain-rates and strain levels, which recapitulate physiological deformations at varied heart rates (at rest and under acute stress) and diastole phases (at early and late filling), respectively. Results and Discussion: We observed that PH increased RVFW viscoelasticity in both longitudinal (outflow tract) and circumferential directions. The tissue anisotropy was pronounced for the diseased RVs, not healthy RVs. We also examined the relative change of viscosity to elasticity by the damping capacity (ratio of dissipated energy to total energy), and we found that PH decreased RVFW damping capacity in both directions. The RV viscoelasticity was also differently altered from resting to acute stress conditions between the groups-the damping capacity was decreased only in the circumferential direction for healthy RVs, but it was reduced in both directions for diseased RVs. Lastly, we found some correlations between the damping capacity and RV function indices and there was no correlation between elasticity or viscosity and RV function. Thus, the RV damping capacity may be a better indicator of RV function than elasticity or viscosity alone. These novel findings on RV dynamic mechanical properties offer deeper insights into the role of RV biomechanics in the adaptation of RV to chronic pressure overload and acute stress.
Collapse
Affiliation(s)
- Wenqiang Liu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Kristen LeBar
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Kellan Roth
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Jassia Pang
- Laboratory Animal Resources, Colorado State University, Fort Collins, CO, United States
| | - Jessica Ayers
- Laboratory Animal Resources, Colorado State University, Fort Collins, CO, United States
| | - Adam J. Chicco
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Christian M. Puttlitz
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO, United States
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
14
|
Baghersad S, Sathish Kumar A, Kipper MJ, Popat K, Wang Z. Recent Advances in Tissue-Engineered Cardiac Scaffolds-The Progress and Gap in Mimicking Native Myocardium Mechanical Behaviors. J Funct Biomater 2023; 14:jfb14050269. [PMID: 37233379 DOI: 10.3390/jfb14050269] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/27/2023] Open
Abstract
Heart failure is the leading cause of death in the US and worldwide. Despite modern therapy, challenges remain to rescue the damaged organ that contains cells with a very low proliferation rate after birth. Developments in tissue engineering and regeneration offer new tools to investigate the pathology of cardiac diseases and develop therapeutic strategies for heart failure patients. Tissue -engineered cardiac scaffolds should be designed to provide structural, biochemical, mechanical, and/or electrical properties similar to native myocardium tissues. This review primarily focuses on the mechanical behaviors of cardiac scaffolds and their significance in cardiac research. Specifically, we summarize the recent development of synthetic (including hydrogel) scaffolds that have achieved various types of mechanical behavior-nonlinear elasticity, anisotropy, and viscoelasticity-all of which are characteristic of the myocardium and heart valves. For each type of mechanical behavior, we review the current fabrication methods to enable the biomimetic mechanical behavior, the advantages and limitations of the existing scaffolds, and how the mechanical environment affects biological responses and/or treatment outcomes for cardiac diseases. Lastly, we discuss the remaining challenges in this field and suggestions for future directions to improve our understanding of mechanical control over cardiac function and inspire better regenerative therapies for myocardial restoration.
Collapse
Affiliation(s)
- Somayeh Baghersad
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Abinaya Sathish Kumar
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Matt J Kipper
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, Fort Collins, CO 80523, USA
- School of Materials Science and Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Ketul Popat
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
- School of Materials Science and Engineering, Colorado State University, Fort Collins, CO 80523, USA
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| | - Zhijie Wang
- School of Biomedical Engineering, Colorado State University, Fort Collins, CO 80523, USA
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
15
|
Goldman J, Liu SQ, Tefft BJ. Anti-Inflammatory and Anti-Thrombogenic Properties of Arterial Elastic Laminae. Bioengineering (Basel) 2023; 10:bioengineering10040424. [PMID: 37106611 PMCID: PMC10135563 DOI: 10.3390/bioengineering10040424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 03/07/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Elastic laminae, an elastin-based, layered extracellular matrix structure in the media of arteries, can inhibit leukocyte adhesion and vascular smooth muscle cell proliferation and migration, exhibiting anti-inflammatory and anti-thrombogenic properties. These properties prevent inflammatory and thrombogenic activities in the arterial media, constituting a mechanism for the maintenance of the structural integrity of the arterial wall in vascular disorders. The biological basis for these properties is the elastin-induced activation of inhibitory signaling pathways, involving the inhibitory cell receptor signal regulatory protein α (SIRPα) and Src homology 2 domain-containing protein tyrosine phosphatase 1 (SHP1). The activation of these molecules causes deactivation of cell adhesion- and proliferation-regulatory signaling mechanisms. Given such anti-inflammatory and anti-thrombogenic properties, elastic laminae and elastin-based materials have potential for use in vascular reconstruction.
Collapse
|
16
|
The Fabrication of Gelatin-Elastin-Nanocellulose Composite Bioscaffold as a Potential Acellular Skin Substitute. Polymers (Basel) 2023; 15:polym15030779. [PMID: 36772084 PMCID: PMC9920652 DOI: 10.3390/polym15030779] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Gelatin usage in scaffold fabrication is limited due to its lack of enzymatic and thermal resistance, as well as its mechanical weakness. Hence, gelatin requires crosslinking and reinforcement with other materials. This study aimed to fabricate and characterise composite scaffolds composed of gelatin, elastin, and cellulose nanocrystals (CNC) and crosslinked with genipin. The scaffolds were fabricated using the freeze-drying method. The composite scaffolds were composed of different concentrations of CNC, whereas scaffolds made of pure gelatin and a gelatin-elastin mixture served as controls. The physicochemical and mechanical properties of the scaffolds, and their cellular biocompatibility with human dermal fibroblasts (HDF), were evaluated. The composite scaffolds demonstrated higher porosity and swelling capacity and improved enzymatic resistance compared to the controls. Although the group with 0.5% (w/v) CNC recorded the highest pore size homogeneity, the diameters of most of the pores in the composite scaffolds ranged from 100 to 200 μm, which is sufficient for cell migration. Tensile strength analysis revealed that increasing the CNC concentration reduced the scaffolds' stiffness. Chemical analyses revealed that despite chemical and structural alterations, both elastin and CNC were integrated into the gelatin scaffold. HDF cultured on the scaffolds expressed collagen type I and α-SMA proteins, indicating the scaffolds' biocompatibility with HDF. Overall, the addition of elastin and CNC improved the properties of gelatin-based scaffolds. The composite scaffolds are promising candidates for an acellular skin substitute.
Collapse
|
17
|
Lysyl oxidase-like 1 deficiency alters ultrastructural and biomechanical properties of the peripapillary sclera in mice. Matrix Biol Plus 2022; 16:100120. [PMID: 36060791 PMCID: PMC9436796 DOI: 10.1016/j.mbplus.2022.100120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/17/2022] [Indexed: 11/30/2022] Open
Abstract
Lysyl oxidate-like 1 knockout (Loxl1-/-) mice have decreased vision without elevated intraocular pressure. Loxl1-/- mice exhibit biometric changes of the anterior segment of the eye. Loxl1-/- mice have altered elastin and collagen structure in peripapillary sclera. Structural alternations of peripapillary sclera correlate with its increased stiffness in Loxl1-/- mice.
Lysyl oxidase-like 1 encoded by the LOXL1 gene is a member of the lysyl oxidase family of enzymes that are important in the maintenance of extracellular matrix (ECM)-rich tissue. LOXL1 is important for proper elastic fiber formation and mice lacking LOXL1 (Loxl1−/−) exhibit systemic elastic fiber disorders, such as pelvic organ prolapse, a phenotype associated with exfoliation syndrome (XFS) in humans. Patients with XFS have a significant risk of developing exfoliation glaucoma (XFG), a severe form of glaucoma, which is a neurodegenerative condition leading to irreversible blindness if not detected and treated in a timely fashion. Although Loxl1−/− mice have been used extensively to investigate mechanisms of pelvic organ prolapse, studies of eyes in those mice are limited and some showed inconsistent ocular phenotypes. In this study we demonstrate that Loxl1−/− mice have significant anterior segment biometric abnormalities which recapitulate some human XFS features. We then focused on the peripapillary sclera (PPS), a critical structure for maintaining optic nerve health. We discovered quantitative and qualitive changes in ultrastructure of PPS, such as reduced elastic fibers, enlarged collagen fibrils, and transformed collagen lamella organization detected by transmission electron microscopy (TEM). Importantly, these changes corelate with altered tissue biomechanics detected by Atomic Force Microscopy (AFM) of PPS in mice. Together, our results support a crucial role for LOXL1 in ocular tissue structure and biomechanics, and Loxl1−/− mice could be a valuable resource for understanding the role of scleral tissue biomechanics in ocular disease.
Collapse
|
18
|
Karimi A, Razaghi R, Padilla S, Rahmati SM, Downs JC, Acott TS, Kelley MJ, Wang RK, Johnstone M. Viscoelastic Biomechanical Properties of the Conventional Aqueous Outflow Pathway Tissues in Healthy and Glaucoma Human Eyes. J Clin Med 2022; 11:6049. [PMID: 36294371 PMCID: PMC9605362 DOI: 10.3390/jcm11206049] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/11/2022] [Accepted: 10/12/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Although the tissues comprising the ocular conventional outflow pathway have shown strong viscoelastic mechanical response to aqueous humor pressure dynamics, the viscoelastic mechanical properties of the trabecular meshwork (TM), juxtacanalicular connective tissue (JCT), and Schlemm's canal (SC) inner wall are largely unknown. METHODS A quadrant of the anterior segment from two human donor eyes at low- and high-flow (LF and HF) outflow regions was pressurized and imaged using optical coherence tomography (OCT). A finite element (FE) model of the TM, the adjacent JCT, and the SC inner wall was constructed and viscoelastic beam elements were distributed in the extracellular matrix (ECM) of the TM and JCT to represent anisotropic collagen. An inverse FE-optimization algorithm was used to calculate the viscoelastic properties of the ECM/beam elements such that the TM/JCT/SC model and OCT imaging data best matched over time. RESULTS The ECM of the glaucoma tissues showed significantly larger time-dependent shear moduli compared to the heathy tissues. Significantly larger shear moduli were also observed in the LF regions of both the healthy and glaucoma eyes compared to the HF regions. CONCLUSIONS The outflow tissues in both glaucoma eyes and HF regions are stiffer and less able to respond to dynamic IOP.
Collapse
Affiliation(s)
- Alireza Karimi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Reza Razaghi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Steven Padilla
- Department of Ophthalmology, University of Washington, Seattle, WA 98109, USA
| | | | - J. Crawford Downs
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Ted S. Acott
- Departments of Ophthalmology and Biochemistry and Molecular Biology, Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Mary J. Kelley
- Departments of Ophthalmology and Integrative Biosciences, Casey Eye Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Ruikang K. Wang
- Department of Ophthalmology, University of Washington, Seattle, WA 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA 98105, USA
| | - Murray Johnstone
- Department of Ophthalmology, University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
19
|
Engineering Smooth Muscle to Understand Extracellular Matrix Remodeling and Vascular Disease. Bioengineering (Basel) 2022; 9:bioengineering9090449. [PMID: 36134994 PMCID: PMC9495899 DOI: 10.3390/bioengineering9090449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
The vascular smooth muscle is vital for regulating blood pressure and maintaining cardiovascular health, and the resident smooth muscle cells (SMCs) in blood vessel walls rely on specific mechanical and biochemical signals to carry out these functions. Any slight change in their surrounding environment causes swift changes in their phenotype and secretory profile, leading to changes in the structure and functionality of vessel walls that cause pathological conditions. To adequately treat vascular diseases, it is essential to understand how SMCs crosstalk with their surrounding extracellular matrix (ECM). Here, we summarize in vivo and traditional in vitro studies of pathological vessel wall remodeling due to the SMC phenotype and, conversely, the SMC behavior in response to key ECM properties. We then analyze how three-dimensional tissue engineering approaches provide opportunities to model SMCs’ response to specific stimuli in the human body. Additionally, we review how applying biomechanical forces and biochemical stimulation, such as pulsatile fluid flow and secreted factors from other cell types, allows us to study disease mechanisms. Overall, we propose that in vitro tissue engineering of human vascular smooth muscle can facilitate a better understanding of relevant cardiovascular diseases using high throughput experiments, thus potentially leading to therapeutics or treatments to be tested in the future.
Collapse
|
20
|
Yi B, Xu Q, Liu W. An overview of substrate stiffness guided cellular response and its applications in tissue regeneration. Bioact Mater 2022; 15:82-102. [PMID: 35386347 PMCID: PMC8940767 DOI: 10.1016/j.bioactmat.2021.12.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 02/06/2023] Open
Abstract
Cell-matrix interactions play a critical role in tissue repair and regeneration. With gradual uncovering of substrate mechanical characteristics that can affect cell-matrix interactions, much progress has been made to unravel substrate stiffness-mediated cellular response as well as its underlying mechanisms. Yet, as a part of cell-matrix interaction biology, this field remains in its infancy, and the detailed molecular mechanisms are still elusive regarding scaffold-modulated tissue regeneration. This review provides an overview of recent progress in the area of the substrate stiffness-mediated cellular responses, including 1) the physical determination of substrate stiffness on cell fate and tissue development; 2) the current exploited approaches to manipulate the stiffness of scaffolds; 3) the progress of recent researches to reveal the role of substrate stiffness in cellular responses in some representative tissue-engineered regeneration varying from stiff tissue to soft tissue. This article aims to provide an up-to-date overview of cell mechanobiology research in substrate stiffness mediated cellular response and tissue regeneration with insightful information to facilitate interdisciplinary knowledge transfer and enable the establishment of prognostic markers for the design of suitable biomaterials. Substrate stiffness physically determines cell fate and tissue development. Rational design of scaffolds requires the understanding of cell-matrix interactions. Substrate stiffness depends on scaffold molecular-constituent-structure interaction. Substrate stiffness-mediated cellular responses vary in different tissues.
Collapse
|
21
|
Guimarães CF, Marques AP, Reis RL. Pushing the Natural Frontier: Progress on the Integration of Biomaterial Cues toward Combinatorial Biofabrication and Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105645. [PMID: 35419887 DOI: 10.1002/adma.202105645] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/14/2022] [Indexed: 06/14/2023]
Abstract
The engineering of fully functional, biological-like tissues requires biomaterials to direct cellular events to a near-native, 3D niche extent. Natural biomaterials are generally seen as a safe option for cell support, but their biocompatibility and biodegradability can be just as limited as their bioactive/biomimetic performance. Furthermore, integrating different biomaterial cues and their final impact on cellular behavior is a complex equation where the outcome might be very different from the sum of individual parts. This review critically analyses recent progress on biomaterial-induced cellular responses, from simple adhesion to more complex stem cell differentiation, looking at the ever-growing possibilities of natural materials modification. Starting with a discussion on native material formulation and the inclusion of cell-instructive cues, the roles of shape and mechanical stimuli, the susceptibility to cellular remodeling, and the often-overlooked impact of cellular density and cell-cell interactions within constructs, are delved into. Along the way, synergistic and antagonistic combinations reported in vitro and in vivo are singled out, identifying needs and current lessons on the development of natural biomaterial libraries to solve the cell-material puzzle efficiently. This review brings together knowledge from different fields envisioning next-generation, combinatorial biomaterial development toward complex tissue engineering.
Collapse
Affiliation(s)
- Carlos F Guimarães
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra P Marques
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
22
|
Karimi A, Rahmati SM, Razaghi R, Crawford Downs J, Acott TS, Wang RK, Johnstone M. Biomechanics of human trabecular meshwork in healthy and glaucoma eyes via dynamic Schlemm's canal pressurization. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 221:106921. [PMID: 35660943 PMCID: PMC10424782 DOI: 10.1016/j.cmpb.2022.106921] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/17/2022] [Accepted: 05/26/2022] [Indexed: 05/27/2023]
Abstract
BACKGROUND AND OBJECTIVE The trabecular meshwork (TM) consists of extracellular matrix (ECM) with embedded collagen and elastin fibers providing its mechanical support. TM stiffness is considerably higher in glaucoma eyes. Emerging data indicates that the TM moves dynamically with transient intraocular pressure (IOP) fluctuations, implying the viscoelastic mechanical behavior of the TM. However, little is known about TM viscoelastic behavior. We calculated the viscoelastic mechanical properties of the TM in n = 2 healthy and n = 2 glaucoma eyes. METHODS A quadrant of the anterior segment was submerged in a saline bath, and a cannula connected to an adjustable saline reservoir was inserted into Schlemm's canal (SC). A spectral domain-OCT (SD-OCT) provided continuous cross-sectional B-scans of the TM/JCT/SC complex during pressure oscillation from 0 to 30 mmHg at two locations. The TM/JCT/SC complex boundaries were delineated to construct a 20-µm-thick volume finite element (FE) mesh. Pre-tensioned collagen and elastin fibrils were embedded in the model using a mesh-free penalty-based cable-in-solid algorithm. SC pressure was represented by a position- and time-dependent pressure boundary; floating boundary conditions were applied to the other cut edges of the model. An FE-optimization algorithm was used to adjust the ECM/fiber mechanical properties such that the TM/JCT/SC model and SD-OCT imaging data best matched over time. RESULTS Significantly larger short- and long-time ECM shear moduli (p = 0.0032), and collagen (1.82x) and elastin (2.72x) fibril elastic moduli (p = 0.0001), were found in the TM of glaucoma eyes compared to healthy controls. CONCLUSIONS These findings provide additional clarity on the mechanical property differences in healthy and glaucomatous outflow pathway under dynamic loading. Understanding the viscoelastic properties of the TM may serve as a new biomarker in early diagnosis of glaucoma.
Collapse
Affiliation(s)
- Alireza Karimi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA.
| | | | - Reza Razaghi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Crawford Downs
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Ted S Acott
- Ophthalmology and Biochemistry and Molecular Biology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon, USA.
| | - Ruikang K Wang
- Department of Ophthalmology, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA.
| | - Murray Johnstone
- Department of Ophthalmology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
23
|
Karimi A, Razaghi R, Rahmati SM, Downs JC, Acott TS, Wang RK, Johnstone M. Modeling the biomechanics of the conventional aqueous outflow pathway microstructure in the human eye. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 221:106922. [PMID: 35660940 PMCID: PMC10424784 DOI: 10.1016/j.cmpb.2022.106922] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/16/2022] [Accepted: 05/26/2022] [Indexed: 05/27/2023]
Abstract
BACKGROUND AND OBJECTIVE Intraocular pressure (IOP) is determined by aqueous humor outflow resistance, which is a function of the combined resistance of Schlemm's canal (SC) endothelium and the trabecular meshwork (TM) and their interactions in the juxtacanalicular connective tissue (JCT) region. Aqueous outflow in the conventional outflow pathway results in pressure gradient across the TM, JCT, and SC inner wall, and induces mechanical stresses and strains that influence the geometry and homeostasis of the outflow system. The outflow resistance is affected by alteration in tissues' geometry, so there is potential for active, two-way, fluid-structure interaction (FSI) coupling between the aqueous humor (fluid) and the TM, JCT, and SC inner wall (structure). However, our understanding of the biomechanical interactions of the aqueous humor with the outflow connective tissues and its contribution to the outflow resistance regulation is incomplete. METHODS In this study, a microstructural finite element (FE) model of a human eye TM, JCT, and SC inner wall was constructed from a segmented, high-resolution histologic 3D reconstruction of the human outflow system. Three different elastic moduli (0.004, 0.128, and 51.5 MPa based on prior reports) were assigned to the TM/JCT complex while the elastic modulus of the SC inner wall was kept constant at 0.00748 MPa. The hydraulic conductivity was programmed separately for the TM, JCT, and SC inner wall using a custom subroutine. Cable elements were embedded into the TM and JCT extracellular matrix to represent the directional stiffness imparted by anisotropic collagen fibril orientation. The resultant stresses and strains in the outflow system were calculated using fluid-structure interaction method. RESULTS The higher TM/JCT stiffness resulted in larger stresses, but smaller strains in the outflow connective tissues, and resulted in a 4- and 5-fold larger pressure drop across the SC inner wall, respectively, compared to the most compliant model. Funneling through µm-sized SC endothelial pores was evident in the models at lower tissue stiffness, but aqueous flow was more turbulent in models with higher TM/JCT stiffness. CONCLUSIONS The mechanical properties of the outflow tissues play a crucial role in the hydrodynamics of the aqueous humor in the conventional outflow system.
Collapse
Affiliation(s)
- Alireza Karimi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University Boulevard, VH 372B, Birmingham, AL 35294, USA.
| | - Reza Razaghi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University Boulevard, VH 372B, Birmingham, AL 35294, USA
| | | | - J Crawford Downs
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, 1670 University Boulevard, VH 372B, Birmingham, AL 35294, USA
| | - Ted S Acott
- Ophthalmology and Biochemistry and Molecular Biology, Casey Eye Institute, Oregon Health & Science University, Portland, OR, USA
| | - Ruikang K Wang
- Department of Ophthalmology, University of Washington, Seattle, WA, USA; Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Murray Johnstone
- Department of Ophthalmology, University of Washington, Seattle, WA, USA
| |
Collapse
|
24
|
Fell CA, Brooks-Richards TL, Woodruff M, Allenby MC. Soft pneumatic actuators for mimicking multi-axial femoropopliteal artery mechanobiology. Biofabrication 2022; 14. [PMID: 35378520 DOI: 10.1088/1758-5090/ac63ef] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/04/2022] [Indexed: 11/12/2022]
Abstract
Tissue biomanufacturing aims to produce lab-grown stem cell grafts and biomimetic drug testing platforms but remains limited in its ability to recapitulate native tissue mechanics. The emerging field of soft robotics aims to emulate dynamic physiological locomotion, representing an ideal approach to recapitulate physiologically complex mechanical stimuli and enhance patient-specific tissue maturation. The kneecap's femoropopliteal artery (FPA) represents a highly flexible tissue across multiple axes during blood flow, walking, standing, and crouching positions, and these complex biomechanics are implicated in the FPA's frequent presentation of peripheral artery disease. We developed a soft pneumatically actuated (SPA) cell culture platform to investigate how patient-specific FPA mechanics affect lab-grown arterial tissues. Silicone hyperelastomers were screened for flexibility and biocompatibility, then additively manufactured into SPAs using a simulation-based design workflow to mimic normal and diseased FPA extensions in radial, angular, and longitudinal dimensions. SPA culture platforms were seeded with mesenchymal stem cells, connected to a pneumatic controller, and provided with 24-hour multi-axial exercise schedules to demonstrate the effect of dynamic conditioning on cell alignment, collagen production, and muscle differentiation without additional growth factors. Soft robotic bioreactors are promising platforms for recapitulating patient-, disease-, and lifestyle-specific mechanobiology for understanding disease, treatment simulations, and lab-grown tissue grafts.
Collapse
Affiliation(s)
- Cody A Fell
- School of Mechanical, Medical and Process Engineering; Centre for Biomedical Technologies, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Queensland, 4001, AUSTRALIA
| | - Trent L Brooks-Richards
- School of Mechanical, Medical and Process Engineering; Centre for Biomedical Technologies, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, Queensland, 4001, AUSTRALIA
| | - Mia Woodruff
- School of Mechanical, Medical and Process Engineering; Centre for Biomedical Technologies, Queensland University of Technology, 60 Musk Avenue, Brisbane, Queensland, 4001, AUSTRALIA
| | - Mark Colin Allenby
- School of Chemical Engineering, The University of Queensland, Andrew N. Liveris Building, St Lucia, Queensland, 4072, AUSTRALIA
| |
Collapse
|
25
|
Zhi D, Cheng Q, Midgley AC, Zhang Q, Wei T, Li Y, Wang T, Ma T, Rafique M, Xia S, Cao Y, Li Y, Li J, Che Y, Zhu M, Wang K, Kong D. Mechanically reinforced biotubes for arterial replacement and arteriovenous grafting inspired by architectural engineering. SCIENCE ADVANCES 2022; 8:eabl3888. [PMID: 35294246 PMCID: PMC8926343 DOI: 10.1126/sciadv.abl3888] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
There is a lack in clinically-suitable vascular grafts. Biotubes, prepared using in vivo tissue engineering, show potential for vascular regeneration. However, their mechanical strength is typically poor. Inspired by architectural design of steel fiber reinforcement of concrete for tunnel construction, poly(ε-caprolactone) (PCL) fiber skeletons (PSs) were fabricated by melt-spinning and heat treatment. The PSs were subcutaneously embedded to induce the assembly of host cells and extracellular matrix to obtain PS-reinforced biotubes (PBs). Heat-treated medium-fiber-angle PB (hMPB) demonstrated superior performance when evaluated by in vitro mechanical testing and following implantation in rat abdominal artery replacement models. hMPBs were further evaluated in canine peripheral arterial replacement and sheep arteriovenous graft models. Overall, hMPB demonstrated appropriate mechanics, puncture resistance, rapid hemostasis, vascular regeneration, and long-term patency, without incidence of luminal expansion or intimal hyperplasia. These optimized hMPB properties show promise as an alternatives to autologous vessels in clinical applications.
Collapse
Affiliation(s)
- Dengke Zhi
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Quhan Cheng
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Adam C. Midgley
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Qiuying Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Tingting Wei
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Yi Li
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Ting Wang
- Urban Transport Emission Control Research Centre, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Tengzhi Ma
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Muhammad Rafique
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Shuang Xia
- Department of Radiology, Tianjin Key Disciplines of Radiology, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Yuejuan Cao
- Department of Vascular Surgery, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Yangchun Li
- Department of Vascular Surgery, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Jing Li
- Department of Ultrasound, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Yongzhe Che
- Department of Pathology and Anatomy, School of Medicine, Nankai University, Tianjin 300071, China
| | - Meifeng Zhu
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- Corresponding author. (D.K.); (K.W.); (M.Z.)
| | - Kai Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- Corresponding author. (D.K.); (K.W.); (M.Z.)
| | - Deling Kong
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- Institute of Transplant Medicine, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
- Corresponding author. (D.K.); (K.W.); (M.Z.)
| |
Collapse
|
26
|
Ellis MW, Riaz M, Huang Y, Anderson CW, Luo J, Park J, Lopez CA, Batty LD, Gibson KH, Qyang Y. Epigallocatechin gallate facilitates extracellular elastin fiber formation in induced pluripotent stem cell derived vascular smooth muscle cells for tissue engineering. J Mol Cell Cardiol 2022; 163:167-174. [PMID: 34979103 PMCID: PMC8920537 DOI: 10.1016/j.yjmcc.2021.12.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 02/03/2023]
Abstract
Tissue engineered vascular grafts possess several advantages over synthetic or autologous grafts, including increased availability and reduced rates of infection and thrombosis. Engineered grafts constructed from human induced pluripotent stem cell derivatives further offer enhanced reproducibility in graft production. One notable obstacle to clinical application of these grafts is the lack of elastin in the vessel wall, which would serve to endow compliance in addition to mechanical strength. This study establishes the ability of the polyphenol compound epigallocatechin gallate, a principal component of green tea, to facilitate the extracellular formation of elastin fibers in vascular smooth muscle cells derived from human induced pluripotent stem cells. Further, this study describes the creation of a doxycycline-inducible elastin expression system to uncouple elastin production from vascular smooth muscle cell proliferative capacity to permit fiber formation in conditions conducive to robust tissue engineering.
Collapse
Affiliation(s)
- Matthew W Ellis
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT 06519, USA
| | - Muhammad Riaz
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Yan Huang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Christopher W Anderson
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Pathology, Yale University, New Haven, CT 06520, USA
| | - Jiesi Luo
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Jinkyu Park
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Colleen A Lopez
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA
| | - Luke D Batty
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Pathology, Yale University, New Haven, CT 06520, USA
| | - Kimberley H Gibson
- Center for Cellular and Molecular Imaging: Electron Microscopy, Department of Cell Biology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yibing Qyang
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06511, USA; Yale Stem Cell Center, New Haven, CT 06520, USA; Department of Pathology, Yale University, New Haven, CT 06520, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
27
|
Wei Q, Wang S, Han F, Wang H, Zhang W, Yu Q, Liu C, Ding L, Wang J, Yu L, Zhu C, Li B, Bl, Cz, Cz, Cz, Qw, Sw, Fh, Hw, Wz, Qy, Cl, Ld, Jw, Ly, Cz, Qw. Cellular modulation by the mechanical cues from biomaterials for tissue engineering. BIOMATERIALS TRANSLATIONAL 2021; 2:323-342. [PMID: 35837415 PMCID: PMC9255801 DOI: 10.12336/biomatertransl.2021.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/13/2021] [Accepted: 07/10/2021] [Indexed: 01/17/2023]
Abstract
Mechanical cues from the extracellular matrix (ECM) microenvironment are known to be significant in modulating the fate of stem cells to guide developmental processes and maintain bodily homeostasis. Tissue engineering has provided a promising approach to the repair or regeneration of damaged tissues. Scaffolds are fundamental in cell-based regenerative therapies. Developing artificial ECM that mimics the mechanical properties of native ECM would greatly help to guide cell functions and thus promote tissue regeneration. In this review, we introduce various mechanical cues provided by the ECM including elasticity, viscoelasticity, topography, and external stimuli, and their effects on cell behaviours. Meanwhile, we discuss the underlying principles and strategies to develop natural or synthetic biomaterials with different mechanical properties for cellular modulation, and explore the mechanism by which the mechanical cues from biomaterials regulate cell function toward tissue regeneration. We also discuss the challenges in multimodal mechanical modulation of cell behaviours and the interplay between mechanical cues and other microenvironmental factors.
Collapse
Affiliation(s)
- Qiang Wei
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Shenghao Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Feng Han
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Huan Wang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Weidong Zhang
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Qifan Yu
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Changjiang Liu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Luguang Ding
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Jiayuan Wang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Lili Yu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China
| | - Caihong Zhu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China,Corresponding authors: Caihong Zhu, ; Bin Li,
| | - Bin Li
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China,College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, Jiangsu Province, China,China Orthopaedic Regenerative Medicine Group (CORMed), Hangzhou, Zhejiang Province, China,Corresponding authors: Caihong Zhu, ; Bin Li,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
|
29
|
Bax DV, Nair M, Weiss AS, Farndale RW, Best SM, Cameron RE. Tailoring the biofunctionality of collagen biomaterials via tropoelastin incorporation and EDC-crosslinking. Acta Biomater 2021; 135:150-163. [PMID: 34454082 DOI: 10.1016/j.actbio.2021.08.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/13/2021] [Accepted: 08/18/2021] [Indexed: 11/17/2022]
Abstract
Recreating the cell niche of virtually all tissues requires composite materials fabricated from multiple extracellular matrix (ECM) macromolecules. Due to their wide tissue distribution, physical attributes and purity, collagen, and more recently, tropoelastin, represent two appealing ECM components for biomaterials development. Here we blend tropoelastin and collagen, harnessing the cell-modulatory properties of each biomolecule. Tropoelastin was stably co-blended into collagen biomaterials and was retained after EDC-crosslinking. We found that human dermal fibroblasts (HDF), rat glial cells (Rugli) and HT1080 fibrosarcoma cells ligate to tropoelastin via EDTA-sensitive and EDTA-insensitive receptors or do not ligate with tropoelastin, respectively. These differing elastin-binding properties allowed us to probe the cellular response to the tropoelastin-collagen composites assigning specific bioactivity to the collagen and tropoelastin component of the composite material. Tropoelastin addition to collagen increased total Rugli cell adhesion, spreading and proliferation. This persisted with EDC-crosslinking of the tropoelastin-collagen composite. Tropoelastin addition did not affect total HDF and HT1080 cell adhesion; however, it increased the contribution of cation-independent adhesion, without affecting the cell morphology or, for HT1080 cells, proliferation. Instead, EDC-crosslinking dictated the HDF and HT1080 cellular response. These data show that a tropoelastin component dominates the response of cells that possess non-integrin based tropoelastin receptors. EDC modification of the collagen component directs cell function when non-integrin tropoelastin receptors are not crucial for cell activity. Using this approach, we have assigned the biological contribution of each component of tropoelastin-collagen composites, allowing informed biomaterial design for directed cell function via more physiologically relevant mechanisms. STATEMENT OF SIGNIFICANCE: Biomaterials fabricated from multiple extracellular matrix (ECM) macromolecules are required to fully recreate the native tissue niche where each ECM macromolecule engages with a specific repertoire of cell-surface receptors. Here we investigate combining tropoelastin with collagen as they interact with cells via different receptors. We identified specific cell lines, which associate with tropoelastin via distinct classes of cell-surface receptor. These showed that tropoelastin, when combined with collagen, altered the cell behaviour in a receptor-usage dependent manner. Integrin-mediated tropoelastin interactions influenced cell proliferation and non-integrin receptors influenced cell spreading and proliferation. These data shed light on the interplay between biomaterial macromolecular composition, cell surface receptors and cell behaviour, advancing bespoke materials design and providing functionality to specific cell populations.
Collapse
Affiliation(s)
- Daniel V Bax
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge, CB3 0FS, United Kingdom; Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, United Kingdom.
| | - Malavika Nair
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge, CB3 0FS, United Kingdom
| | - Anthony S Weiss
- Life and Environmental Sciences, University of Sydney, NSW, 2006, Australia; Charles Perkins Centre, University of Sydney, NSW, 2006, Australia; Sydney Nano Institute, University of Sydney, NSW, 2006, Australia
| | - Richard W Farndale
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, United Kingdom
| | - Serena M Best
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge, CB3 0FS, United Kingdom
| | - Ruth E Cameron
- Department of Materials Science and Metallurgy, University of Cambridge, 27 Charles Babbage Road, Cambridge, CB3 0FS, United Kingdom
| |
Collapse
|
30
|
Elastin-Plasma Hybrid Hydrogels for Skin Tissue Engineering. Polymers (Basel) 2021; 13:polym13132114. [PMID: 34203144 PMCID: PMC8271496 DOI: 10.3390/polym13132114] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/20/2021] [Accepted: 06/22/2021] [Indexed: 11/23/2022] Open
Abstract
Dermo-epidermal equivalents based on plasma-derived fibrin hydrogels have been extensively studied for skin engineering. However, they showed rapid degradation and contraction over time and low mechanical properties which limit their reproducibility and lifespan. In order to achieve better mechanical properties, elasticity and biological properties, we incorporated a elastin-like recombinamer (ELR) network, based on two types of ELR, one modified with azide (SKS-N3) and other with cyclooctyne (SKS-Cyclo) chemical groups at molar ratio 1:1 at three different SKS (serine-lysine-serine sequence) concentrations (1, 3, and 5 wt.%), into plasma-derived fibrin hydrogels. Our results showed a decrease in gelation time and contraction, both in the absence and presence of the encapsulated human primary fibroblasts (hFBs), higher mechanical properties and increase in elasticity when SKSs content is equal or higher than 3%. However, hFBs proliferation showed an improvement when the lowest SKS content (1 wt.%) was used but started decreasing when increasing SKS concentration at day 14 with respect to the plasma control. Proliferation of human primary keratinocytes (hKCs) seeded on top of the hybrid-plasma hydrogels containing 1 and 3% of SKS showed no differences to plasma control and an increase in hKCs proliferation was observed for hybrid-plasma hydrogels containing 5 wt.% of SKS. These promising results showed the need to achieve a balance between the reduced contraction, the better mechanical properties and biological properties and indicate the potential of using this type of hydrogel as a testing platform for pharmaceutical products and cosmetics, and future work will elucidate their potential.
Collapse
|
31
|
Ye L, Takagi T, Tu C, Hagiwara A, Geng X, Feng Z. The performance of heparin modified poly(ε-caprolactone) small diameter tissue engineering vascular graft in canine-A long-term pilot experiment in vivo. J Biomed Mater Res A 2021; 109:2493-2505. [PMID: 34096176 DOI: 10.1002/jbm.a.37243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 05/12/2021] [Accepted: 05/28/2021] [Indexed: 01/22/2023]
Abstract
Long-term in vivo observation in large animal model is critical for evaluating the potential of small diameter tissue engineering vascular graft (SDTEVG) in clinical application, but is rarely reported. In this study, a SDTEVG is fabricated by the electrospinning of poly(ε-caprolactone) and subsequent heparin modification. SDTEVG is implanted into canine's abdominal aorta for 511 days in order to investigate its clinical feasibility. An active and robust remodeling process was characterized by a confluent endothelium, macrophage infiltrate, extracellular matrix deposition and remodeling on the explanted graft. The immunohistochemical and immunofluorescence analysis further exhibit the regeneration of endothelium and smooth muscle layer on tunica intima and tunica media, respectively. Thus, long-term follow-up reveals viable neovessel formation beyond graft degradation. Furthermore, the von Kossa staining exhibits no occurrence of calcification. However, although no TEVG failure or rupture happens during the follow-up, the aneurysm is found by both Doppler ultrasonic and gross observation. Consequently, as-prepared TEVG shows promising potential in vascular tissue engineering if it can be appropriately strengthened to prevent the occurrence of aneurysm.
Collapse
Affiliation(s)
- Lin Ye
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China.,Department of Medical Life System, Doshisha University, Kyoto, Japan
| | - Toshitaka Takagi
- Department of Medical Life System, Doshisha University, Kyoto, Japan
| | - Chengzhao Tu
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China
| | - Akeo Hagiwara
- Department of Medical Life System, Doshisha University, Kyoto, Japan
| | - Xue Geng
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China.,Beijing Key Laboratory of Construction Tailorable Advanced Functional Materials and Green Applications, Beijing, China
| | - Zengguo Feng
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, China.,Beijing Key Laboratory of Construction Tailorable Advanced Functional Materials and Green Applications, Beijing, China
| |
Collapse
|
32
|
Extracellular Vesicles Derived from Primary Adipose Stromal Cells Induce Elastin and Collagen Deposition by Smooth Muscle Cells within 3D Fibrin Gel Culture. Bioengineering (Basel) 2021; 8:bioengineering8050051. [PMID: 33925413 PMCID: PMC8145221 DOI: 10.3390/bioengineering8050051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/13/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023] Open
Abstract
Macromolecular components of the vascular extracellular matrix (ECM), particularly elastic fibers and collagen fibers, are critical for the proper physiological function of arteries. When the unique biomechanical combination of these fibers is disrupted, or in the ultimate extreme where fibers are completely lost, arterial disease can emerge. Bioengineers in the realms of vascular tissue engineering and regenerative medicine must therefore ideally consider how to create tissue engineered vascular grafts containing the right balance of these fibers and how to develop regenerative treatments for situations such as an aneurysm where fibers have been lost. Previous work has demonstrated that the primary cells responsible for vascular ECM production during development, arterial smooth muscle cells (SMCs), can be induced to make new elastic fibers when exposed to secreted factors from adipose-derived stromal cells. To further dissect how this signal is transmitted, in this study, the factors were partitioned into extracellular vesicle (EV)-rich and EV-depleted fractions as well as unseparated controls. EVs were validated using electron microscopy, dynamic light scattering, and protein quantification before testing for biological effects on SMCs. In 2D culture, EVs promoted SMC proliferation and migration. After 30 days of 3D fibrin construct culture, EVs promoted SMC transcription of the elastic microfibril gene FBN1 as well as SMC deposition of insoluble elastin and collagen. Uniaxial biomechanical properties of strand fibrin constructs were no different after 30 days of EV treatment versus controls. In summary, it is apparent that some of the positive effects of adipose-derived stromal cells on SMC elastogenesis are mediated by EVs, indicating a potential use for these EVs in a regenerative therapy to restore the biomechanical function of vascular ECM in arterial disease.
Collapse
|
33
|
Abstract
Significance: The vascular extracellular matrix (ECM) not only provides mechanical stability but also manipulates vascular cell behaviors, which are crucial for vascular function and homeostasis. ECM remodeling, which alters vascular wall mechanical properties and exposes vascular cells to bioactive molecules, is involved in the development and progression of hypertension. Recent Advances: This brief review summarized the dynamic changes in ECM components and their modification and degradation during hypertension and after antihypertensive treatment. We also discussed how alterations in the ECM amount, assembly, mechanical properties, and degradation fragment generation provide input into the pathological process of hypertension. Critical Issues: Although the relevance between ECM remodeling and hypertension has been recognized, the underlying mechanism by which ECM remodeling initiates the development of hypertension remains unclear. Therefore, the modulation of ECM remodeling on arterial stiffness and hypertension in genetically modified rodent models is summarized in this review. The circulating biomarkers based on ECM metabolism and therapeutic strategies targeting ECM disorders in hypertension are also introduced. Future Directions: Further research will provide more comprehensive understanding of ECM remodeling in hypertension by the application of matridomic and degradomic approaches. The better understanding of mechanisms underlying vascular ECM remodeling may provide novel potential therapeutic strategies for preventing and treating hypertension. Antioxid. Redox Signal. 34, 765-783.
Collapse
Affiliation(s)
- Zeyu Cai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Ze Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Zhiqing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Li Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China
| |
Collapse
|
34
|
Sharma A, Sharma P, Roy S. Elastin-inspired supramolecular hydrogels: a multifaceted extracellular matrix protein in biomedical engineering. SOFT MATTER 2021; 17:3266-3290. [PMID: 33730140 DOI: 10.1039/d0sm02202k] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The phenomenal advancement in regenerative medicines has led to the development of bioinspired materials to fabricate a biomimetic artificial extracellular matrix (ECM) to support cellular survival, proliferation, and differentiation. Researchers have diligently developed protein polymers consisting of functional sequences of amino acids evolved in nature. Nowadays, certain repetitive bioinspired polymers are treated as an alternative to synthetic polymers due to their unique properties like biodegradability, easy scale-up, biocompatibility, and non-covalent molecular associations which imparts tunable supramolecular architecture to these materials. In this direction, elastin has been identified as a potential scaffold that renders extensibility and elasticity to the tissues. Elastin-like polypeptides (ELPs) are artificial repetitive polymers that exhibit lower critical solution temperature (LCST) behavior in a particular environment than synthetic polymers and hence have gained extensive interest in the fabrication of stimuli-responsive biomaterials. This review discusses in detail the unique structural aspects of the elastin and its soluble precursor, tropoelastin. Furthermore, the versatility of elastin-like peptides is discussed through numerous examples that bolster the significance of elastin in the field of regenerative medicines such as wound care, cardiac tissue engineering, ocular disorders, bone tissue regeneration, etc. Finally, the review highlights the importance of exploring short elastin-mimetic peptides to recapitulate the structural and functional aspects of elastin for advanced healthcare applications.
Collapse
Affiliation(s)
- Archita Sharma
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, 140306, Punjab, India.
| | | | | |
Collapse
|
35
|
Almeida-González FR, González-Vázquez A, Mithieux SM, O'Brien FJ, Weiss AS, Brougham CM. A step closer to elastogenesis on demand; Inducing mature elastic fibre deposition in a natural biomaterial scaffold. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111788. [PMID: 33545914 DOI: 10.1016/j.msec.2020.111788] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/20/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022]
Abstract
Elastic fibres play a key role in bodily functions where fatigue resistance and elastic recovery are necessary while regulating phenotype, proliferation and migration in cells. While in vivo elastic fibres are created at a late foetal stage, a major obstacle in the development of engineered tissue is that human vascular smooth muscle cells (hVSMCs), one of the principal elastogenic cells, are unable to spontaneously promote elastogenesis in vitro. Therefore, the overall aim of this study was to activate elastogenesis in vitro by hVSMCs seeded in fibrin, collagen, glycosaminoglycan (FCG) scaffolds, following the addition of recombinant human tropoelastin. This combination of scaffold, tropoelastin and cells induced the deposition of elastin and formation of lamellar maturing elastic fibres, similar to those found in skin, blood vessels and heart valves. Furthermore, higher numbers of maturing branched elastic fibres were synthesised when a higher cell density was used and by drop-loading tropoelastin onto cell-seeded FCG scaffolds prior to adding growth medium. The addition of tropoelastin showed no effect on cell proliferation or mechanical properties of the scaffold which remained dimensionally stable throughout. With these results, we have established a natural biomaterial scaffold that can undergo controlled elastogenesis on demand, suitable for tissue engineering applications.
Collapse
Affiliation(s)
- Francisco R Almeida-González
- Biomedical Research Group, School of Mechanical and Design Engineering, Technological University Dublin, Bolton St, Dublin 1, Ireland; Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Ireland
| | - Arlyng González-Vázquez
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Ireland; Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI, Ireland
| | - Suzanne M Mithieux
- Charles Perkins Centre, University of Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, University of Sydney, NSW 2006, Australia; Bosch Institute, University of Sydney, NSW 2006, Australia
| | - Fergal J O'Brien
- Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Ireland; Advanced Materials and Bioengineering Research (AMBER) Centre, RCSI, Ireland
| | - Anthony S Weiss
- Charles Perkins Centre, University of Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, University of Sydney, NSW 2006, Australia; Bosch Institute, University of Sydney, NSW 2006, Australia
| | - Claire M Brougham
- Biomedical Research Group, School of Mechanical and Design Engineering, Technological University Dublin, Bolton St, Dublin 1, Ireland; Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, RCSI University of Medicine and Health Sciences, 123 St. Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
36
|
Zamprogno P, Wüthrich S, Achenbach S, Thoma G, Stucki JD, Hobi N, Schneider-Daum N, Lehr CM, Huwer H, Geiser T, Schmid RA, Guenat OT. Second-generation lung-on-a-chip with an array of stretchable alveoli made with a biological membrane. Commun Biol 2021; 4:168. [PMID: 33547387 PMCID: PMC7864995 DOI: 10.1038/s42003-021-01695-0] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
The air-blood barrier with its complex architecture and dynamic environment is difficult to mimic in vitro. Lung-on-a-chips enable mimicking the breathing movements using a thin, stretchable PDMS membrane. However, they fail to reproduce the characteristic alveoli network as well as the biochemical and physical properties of the alveolar basal membrane. Here, we present a lung-on-a-chip, based on a biological, stretchable and biodegradable membrane made of collagen and elastin, that emulates an array of tiny alveoli with in vivo-like dimensions. This membrane outperforms PDMS in many ways: it does not absorb rhodamine-B, is biodegradable, is created by a simple method, and can easily be tuned to modify its thickness, composition and stiffness. The air-blood barrier is reconstituted using primary lung alveolar epithelial cells from patients and primary lung endothelial cells. Typical alveolar epithelial cell markers are expressed, while the barrier properties are preserved for up to 3 weeks.
Collapse
Affiliation(s)
- Pauline Zamprogno
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Simon Wüthrich
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Sven Achenbach
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Giuditta Thoma
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
| | - Janick D Stucki
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- AlveoliX AG, Bern, Switzerland
| | - Nina Hobi
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland
- AlveoliX AG, Bern, Switzerland
| | - Nicole Schneider-Daum
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Claus-Michael Lehr
- Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Saarbrücken, Germany
| | - Hanno Huwer
- SHG Clinics, Department of Cardiothoracic Surgery, Völklingen Heart Center, Völklingen, Germany
| | - Thomas Geiser
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland
| | - Ralph A Schmid
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland
| | - Olivier T Guenat
- Organs-on-Chip Technologies Laboratory, ARTORG Center, University of Bern, Bern, Switzerland.
- Department of Pulmonary Medicine, University Hospital of Bern, Bern, Switzerland.
- Department of General Thoracic Surgery, University Hospital of Bern, Bern, Switzerland.
| |
Collapse
|
37
|
Moysidou CM, Barberio C, Owens RM. Advances in Engineering Human Tissue Models. Front Bioeng Biotechnol 2021; 8:620962. [PMID: 33585419 PMCID: PMC7877542 DOI: 10.3389/fbioe.2020.620962] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
Research in cell biology greatly relies on cell-based in vitro assays and models that facilitate the investigation and understanding of specific biological events and processes under different conditions. The quality of such experimental models and particularly the level at which they represent cell behavior in the native tissue, is of critical importance for our understanding of cell interactions within tissues and organs. Conventionally, in vitro models are based on experimental manipulation of mammalian cells, grown as monolayers on flat, two-dimensional (2D) substrates. Despite the amazing progress and discoveries achieved with flat biology models, our ability to translate biological insights has been limited, since the 2D environment does not reflect the physiological behavior of cells in real tissues. Advances in 3D cell biology and engineering have led to the development of a new generation of cell culture formats that can better recapitulate the in vivo microenvironment, allowing us to examine cells and their interactions in a more biomimetic context. Modern biomedical research has at its disposal novel technological approaches that promote development of more sophisticated and robust tissue engineering in vitro models, including scaffold- or hydrogel-based formats, organotypic cultures, and organs-on-chips. Even though such systems are necessarily simplified to capture a particular range of physiology, their ability to model specific processes of human biology is greatly valued for their potential to close the gap between conventional animal studies and human (patho-) physiology. Here, we review recent advances in 3D biomimetic cultures, focusing on the technological bricks available to develop more physiologically relevant in vitro models of human tissues. By highlighting applications and examples of several physiological and disease models, we identify the limitations and challenges which the field needs to address in order to more effectively incorporate synthetic biomimetic culture platforms into biomedical research.
Collapse
Affiliation(s)
| | | | - Róisín Meabh Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
38
|
Gong J, Meng T, Yang J, Hu N, Zhao H, Tian T. Three-dimensional in vitro tissue culture models of brain organoids. Exp Neurol 2021; 339:113619. [PMID: 33497645 DOI: 10.1016/j.expneurol.2021.113619] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/03/2021] [Accepted: 01/12/2021] [Indexed: 12/18/2022]
Abstract
Brain organoids are three-dimensional self-assembled structures that are derived from human induced pluripotent stem cells (hiPSCs). They can recapitulate the spatiotemporal organization and function of the brain, presenting a robust system for in vitro modeling of brain development, evolution, and diseases. Significant advances in biomaterials, microscale technologies, gene editing technologies, and stem cell biology have enabled the construction of human specific brain structures in vitro. However, the limitations of long-term culture, necrosis, and hypoxic cores in different culture models obstruct brain organoid growth and survival. The in vitro models should facilitate oxygen and nutrient absorption, which is essential to generate complex organoids and provides a biomimetic microenvironment for modeling human brain organogenesis and human diseases. This review aims to highlight the progress in the culture devices of brain organoids, including dish, bioreactor, and organ-on-a-chip models. With the modulation of bioactive molecules and biomaterials, the generated organoids recapitulate the key features of the human brain in a more reproducible and hyperoxic fashion. Furthermore, an outlook for future preclinical studies and the genetic modifications of brain organoids is presented.
Collapse
Affiliation(s)
- Jing Gong
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Tianyue Meng
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jun Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Ning Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Hezhao Zhao
- Gastrointestinal Cancer Center, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Tian Tian
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
39
|
Mechanical, compositional and morphological characterisation of the human male urethra for the development of a biomimetic tissue engineered urethral scaffold. Biomaterials 2021; 269:120651. [PMID: 33476892 DOI: 10.1016/j.biomaterials.2021.120651] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/31/2020] [Accepted: 12/31/2020] [Indexed: 12/11/2022]
Abstract
This study addresses a crucial gap in the literature by characterising the relationship between urethral tissue mechanics, composition and gross structure. We then utilise these data to develop a biomimetic urethral scaffold with physical properties that more accurately mimic the native tissue than existing gold standard scaffolds; small intestinal submucosa (SIS) and urinary bladder matrix (UBM). Nine human urethra samples were mechanically characterised using pressure-diameter and uniaxial extension testing. The composition and gross structure of the tissue was determined using immunohistological staining. A pressure stiffening response is observed during the application of intraluminal pressure. The elastic and viscous tissue responses to extension are free of regional or directional variance. The elastin and collagen content of the tissue correlates significantly with tissue mechanics. Building on these data, a biomimetic urethral scaffold was fabricated from collagen and elastin in a ratio that mimics the composition of the native tissue. The resultant scaffold is comprised of a dense inner layer and a porous outer layer that structurally mimic the submucosa and corpus spongiosum layers of the native tissue, respectively. The porous outer layer facilitated more uniform cell infiltration relative to SIS and UBM when implanted subcutaneously (p < 0.05). The mechanical properties of the biomimetic scaffold better mimic the native tissue compared to SIS and UBM. The tissue characterisation data presented herein paves the way for the development of biomimetic urethral grafts, and the novel scaffold we develop demonstrates positive findings that warrant further in vivo evaluation.
Collapse
|
40
|
Gonçalves RC, Banfi A, Oliveira MB, Mano JF. Strategies for re-vascularization and promotion of angiogenesis in trauma and disease. Biomaterials 2020; 269:120628. [PMID: 33412374 DOI: 10.1016/j.biomaterials.2020.120628] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/14/2020] [Accepted: 12/18/2020] [Indexed: 12/18/2022]
Abstract
The maintenance of a healthy vascular system is essential to ensure the proper function of all organs of the human body. While macrovessels have the main role of blood transportation from the heart to all tissues, microvessels, in particular capillaries, are responsible for maintaining tissues' functionality by providing oxygen, nutrients and waste exchanges. Occlusion of blood vessels due to atherosclerotic plaque accumulation remains the leading cause of mortality across the world. Autologous vein and artery grafts bypassing are the current gold standard surgical procedures to substitute primarily obstructed vascular structures. Ischemic scenarios that condition blood supply in downstream tissues may arise from blockage phenomena, as well as from other disease or events leading to trauma. The (i) great demand for new vascular substitutes, arising from both the limited availability of healthy autologous vessels, as well as the shortcomings associated with small-diameter synthetic vascular grafts, and (ii) the challenging induction of the formation of adequate and stable microvasculature are current driving forces for the growing interest in the development of bioinspired strategies to ensure the proper function of vasculature in all its dimensional scales. Here, a critical review of well-established technologies and recent biotechnological advances to substitute or regenerate the vascular system is provided.
Collapse
Affiliation(s)
- Raquel C Gonçalves
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Andrea Banfi
- Department of Biomedicine, University of Basel, Basel, 4056, Switzerland; Department of Surgery, University Hospital Basel, Basel, 4056, Switzerland
| | - Mariana B Oliveira
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
41
|
Rodrigues ICP, Pereira KD, Woigt LF, Jardini AL, Luchessi AD, Lopes ÉSN, Webster TJ, Gabriel LP. A novel technique to produce tubular scaffolds based on collagen and elastin. Artif Organs 2020; 45:E113-E122. [PMID: 33169400 DOI: 10.1111/aor.13857] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/27/2020] [Accepted: 11/03/2020] [Indexed: 12/14/2022]
Abstract
Tubular polymer scaffolds based on tissue engineering techniques have been studied as potential alternatives for vascular regeneration implants. The blood vessels of the cardiovascular system are mainly fibrous, composed of collagen (Col) and elastin (El), and its inner layer consists of endothelial cells. In this work, Col and El were combined with polyurethane (PU), a biocompatible synthetic polymer, and rotary jet spinning, a new and highly productive technique, to produce fibrous scaffolds. The scaffolds produced at 18 000 rpm presented homogeneous, bead-free, and solvent-free fibers. The blend formation between PU-Col-El was identified by chemical composition analysis and enhanced the thermal stability up to 324°C. The hydrophilic nature of the scaffold was revealed by its low contact angle. Cell viability of human umbilical vein endothelial cells with the scaffold was proven for 72 hours. The combined strategy of rotary jet spinning with a polymer blend containing Col and El was verified as an effective and promising alternative to obtain tubular scaffolds for tissue engineering on a large-scale production.
Collapse
Affiliation(s)
- Isabella C P Rodrigues
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,School of Mechanical Engineering, University of Campinas, Campinas, Brazil
| | - Karina D Pereira
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,Institute of Biosciences, São Paulo State University, Rio Claro, Brazil
| | - Luiza F Woigt
- School of Applied Sciences, University of Campinas, Limeira, Brazil
| | | | - Augusto D Luchessi
- School of Applied Sciences, University of Campinas, Limeira, Brazil.,Institute of Biosciences, São Paulo State University, Rio Claro, Brazil
| | - Éder S N Lopes
- School of Mechanical Engineering, University of Campinas, Campinas, Brazil
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Laís P Gabriel
- School of Applied Sciences, University of Campinas, Limeira, Brazil
| |
Collapse
|
42
|
Wang Z, Liu L, Mithieux SM, Weiss AS. Fabricating Organized Elastin in Vascular Grafts. Trends Biotechnol 2020; 39:505-518. [PMID: 33019966 DOI: 10.1016/j.tibtech.2020.09.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/07/2020] [Accepted: 09/08/2020] [Indexed: 01/01/2023]
Abstract
Surgically bypassing or replacing a severely damaged artery using a biodegradable synthetic vascular graft is a promising treatment that allows for the remodeling and regeneration of the graft to form a neoartery. Elastin-based structures, such as elastic fibers, elastic lamellae, and laminae, are key functional components in the arterial extracellular matrix. In this review, we identify the lack of elastin in vascular grafts as a key factor that prevents their long-term success. We further summarize advances in vascular tissue engineering that are focused on either de novo production of organized elastin or incorporation of elastin-based biomaterials within vascular grafts to mitigate failure and enhance enduring in vivo performance.
Collapse
Affiliation(s)
- Ziyu Wang
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Linyang Liu
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Suzanne M Mithieux
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Anthony S Weiss
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia; School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia; Sydney Nano Institute, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
43
|
Hyaluronan promotes the regeneration of vascular smooth muscle with potent contractile function in rapidly biodegradable vascular grafts. Biomaterials 2020; 257:120226. [DOI: 10.1016/j.biomaterials.2020.120226] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 06/30/2020] [Accepted: 07/04/2020] [Indexed: 12/15/2022]
|
44
|
Yong U, Lee S, Jung S, Jang J. Interdisciplinary approaches to advanced cardiovascular tissue engineering: ECM-based biomaterials, 3D bioprinting, and its assessment. ACTA ACUST UNITED AC 2020. [DOI: 10.1088/2516-1091/abb211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
45
|
Vazquez-Portalatin N, Alfonso-Garcia A, Liu JC, Marcu L, Panitch A. Physical, Biomechanical, and Optical Characterization of Collagen and Elastin Blend Hydrogels. Ann Biomed Eng 2020; 48:2924-2935. [PMID: 32929559 DOI: 10.1007/s10439-020-02605-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/02/2020] [Indexed: 12/13/2022]
Abstract
Collagen and elastin proteins are major components of the extracellular matrix of many organs. The presence of collagen and elastin networks, and their associated properties, in different tissues have led scientists to study collagen and elastin composites for use in tissue engineering. In this study, we characterized physical, biochemical, and optical properties of gels composed of collagen and elastin blends. We demonstrated that the addition of varying amounts of elastin to the constructs alters collagen fibrillogenesis, D-banding pattern length, and storage modulus. However, the addition of elastin does not affect collagen fibril diameter. We also evaluated the autofluorescence properties of the different collagen and elastin blends with fluorescence lifetime imaging (FLIm). Autofluorescence emission showed a red shift with the addition of elastin to the hydrogels. The fluorescence lifetime values of the gels increased with the addition of elastin and were strongly correlated with the storage moduli measurements. These results suggest that FLIm can be used to monitor the gels' mechanical properties nondestructively. These collagen and elastin constructs, along with the FLIm capabilities, can be used to develop and study collagen and elastin composites for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Nelda Vazquez-Portalatin
- Biomedical Engineering Department, University of California, Davis, 451 Health Sciences Dr, Davis, CA, 95616, USA.,Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr, West Lafayette, IN, 47907, USA
| | - Alba Alfonso-Garcia
- Biomedical Engineering Department, University of California, Davis, 451 Health Sciences Dr, Davis, CA, 95616, USA
| | - Julie C Liu
- Weldon School of Biomedical Engineering, Purdue University, 206 S Martin Jischke Dr, West Lafayette, IN, 47907, USA.,Davidson School of Chemical Engineering, Purdue University, 480 Stadium Mall Dr, West Lafayette, IN, 47907, USA
| | - Laura Marcu
- Biomedical Engineering Department, University of California, Davis, 451 Health Sciences Dr, Davis, CA, 95616, USA
| | - Alyssa Panitch
- Biomedical Engineering Department, University of California, Davis, 451 Health Sciences Dr, Davis, CA, 95616, USA. .,Department of Surgery, University of California, Davis, 2335 Stockton Boulevard, Sacramento, CA, 95817, USA.
| |
Collapse
|
46
|
Nakatsuji H, Kitano S, Irie S, Matsusaki M. Preparation of Extracellular Matrix Paper and Construction of Multi-Layered 3D Tissue Model. ACTA ACUST UNITED AC 2020; 88:e112. [PMID: 32776707 DOI: 10.1002/cpcb.112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Construction of organized three-dimensional (3D) tissue with extracellular matrix (ECM) and multiple types of cells is important for tissue engineering to enable tissue function and enhance cellular function. However, the concentration of ECM and the thickness of the 3D tissue have been limited in previous methods due to a lack of permeability to nutrients and oxygen. Besides, it is difficult to use matured natural ECM as a cell scaffold without chemical modification due to its insolubility. In this article, we focus on multi-layered structure, which is commonly found in living tissue such as skin, blood vessels, and other organs. Here, we describe the preparation of a paper-like scaffold (ECM paper) from micro-fibered natural ECM and the construction of 3D multi-layered tissue composed of cell layers and ECM layers by stacking cell-seeded ECM papers. The thickness and components of the ECM layers are easily controllable by changing the composition of the ECM papers, and the fibrous structure of ECM paper shows high permeability and permits cell migration. Additionally, the ECM microfiber, which is physically defiberized from natural ECM, has a high ECM concentration equal to that of living tissue and high stability under physiological conditions. Therefore, this set of protocols enables construction of multi-layered 3D tissue composed of precisely controlled ECM layers and cell layers that may contribute to the assembly of tissue models. © 2020 by John Wiley & Sons, Inc. Basic Protocol 1: Preparation of extracellular matrix paper Basic Protocol 2: Evaluation of cellular function of cells on extracellular matrix paper Basic Protocol 3: Construction of multi-layered 3D tissue.
Collapse
Affiliation(s)
- Hirotaka Nakatsuji
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Shiro Kitano
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan.,Toppan Printing Co., Ltd., Tokyo, Japan
| | - Shinji Irie
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan.,Toppan Printing Co., Ltd., Tokyo, Japan
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan.,Division of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| |
Collapse
|
47
|
Santarella F, Sridharan R, Marinkovic M, Do Amaral RJFC, Cavanagh B, Smith A, Kashpur O, Gerami‐Naini B, Garlick JA, O'Brien FJ, Kearney CJ. Scaffolds Functionalized with Matrix from Induced Pluripotent Stem Cell Fibroblasts for Diabetic Wound Healing. Adv Healthc Mater 2020; 9:e2000307. [PMID: 32597577 DOI: 10.1002/adhm.202000307] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/12/2020] [Indexed: 12/15/2022]
Abstract
Diabetic foot ulcers (DFUs) are chronic wounds, with 20% of cases resulting in amputation, despite intervention. A recently approved tissue engineering product-a cell-free collagen-glycosaminoglycan (GAG) scaffold-demonstrates 50% success, motivating its functionalization with extracellular matrix (ECM). Induced pluripotent stem cell (iPSC) technology reprograms somatic cells into an embryonic-like state. Recent findings describe how iPSCs-derived fibroblasts ("post-iPSF") are proangiogenic, produce more ECM than their somatic precursors ("pre-iPSF"), and their ECM has characteristics of foetal ECM (a wound regeneration advantage, as fetuses heal scar-free). ECM production is 45% higher from post-iPSF and has favorable components (e.g., Collagen I and III, and fibronectin). Herein, a freeze-dried scaffold using ECM grown by post-iPSF cells (Post-iPSF Coll) is developed and tested vs precursors ECM-activated scaffolds (Pre-iPSF Coll). When seeded with healthy or DFU fibroblasts, both ECM-derived scaffolds have more diverse ECM and more robust immune responses to cues. Post-iPSF-Coll had higher GAG, higher cell content, higher Vascular Endothelial Growth Factor (VEGF) in DFUs, and higher Interleukin-1-receptor antagonist (IL-1ra) vs. pre-iPSF Coll. This work constitutes the first step in exploiting ECM from iPSF for tissue engineering scaffolds.
Collapse
Affiliation(s)
- Francesco Santarella
- Royal College of Surgeons in Ireland 123 St Stephen's Green, Saint Peter's Dublin D02 YN77 Ireland
| | - Rukmani Sridharan
- Royal College of Surgeons in Ireland 123 St Stephen's Green, Saint Peter's Dublin D02 YN77 Ireland
| | - Milica Marinkovic
- Royal College of Surgeons in Ireland 123 St Stephen's Green, Saint Peter's Dublin D02 YN77 Ireland
| | - Ronaldo Jose Farias Correa Do Amaral
- Royal College of Surgeons in Ireland 123 St Stephen's Green, Saint Peter's Dublin D02 YN77 Ireland
- Biomedical Sciences, National University of Ireland Galway Newcastle Road Galway H91 W2TY Ireland
| | - Brenton Cavanagh
- Royal College of Surgeons in Ireland 123 St Stephen's Green, Saint Peter's Dublin D02 YN77 Ireland
| | - Avi Smith
- Department of Diagnostic SciencesTufts University School of Dental Medicine Boston MA 02111 USA
| | - Olga Kashpur
- Department of Diagnostic SciencesTufts University School of Dental Medicine Boston MA 02111 USA
| | - Behzad Gerami‐Naini
- Department of Diagnostic SciencesTufts University School of Dental Medicine Boston MA 02111 USA
| | - Jonathan A. Garlick
- Department of Diagnostic SciencesTufts University School of Dental Medicine Boston MA 02111 USA
| | - Fergal J. O'Brien
- Royal College of Surgeons in Ireland 123 St Stephen's Green, Saint Peter's Dublin D02 YN77 Ireland
- The University of Dublin Trinity College, College Street Dublin Dublin 2, D02 R590 Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)RCSI and TCD Dublin D02 HP52 Ireland
| | - Cathal J. Kearney
- Royal College of Surgeons in Ireland 123 St Stephen's Green, Saint Peter's Dublin D02 YN77 Ireland
- The University of Dublin Trinity College, College Street Dublin Dublin 2, D02 R590 Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)RCSI and TCD Dublin D02 HP52 Ireland
- Department of Biomedical EngineeringUniversity of Massachusetts Amherst Amherst MA 01003‐9292 USA
| |
Collapse
|
48
|
Hierarchical biofabrication of biomimetic collagen-elastin vascular grafts with controllable properties via lyophilisation. Acta Biomater 2020; 112:52-61. [PMID: 32525053 DOI: 10.1016/j.actbio.2020.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 12/19/2022]
Abstract
This article describes the development of a hierarchical biofabrication technique suitable to create large but complex structures, such as vascular mimicking grafts, using facile lyophilisation technology amenable to multiple other biomaterial classes. The combination of three fabrication techniques together, namely solvent evaporation, lyophilisation, and crosslinking together allows highly tailorable structures from the microstructure up to the macrostructure, and with the ability to independently crosslink each layer it allows great flexibility to match desired native mechanical properties independently of the micro/macrostructure. We have demonstrated the flexibility of this biofabrication technique by independently optimising each of the layers to create a multi-layered arterial structure with tailored architectural and biophysical/biochemical properties using a collagen-elastin composite. Taken together, the facile biofabrication methodology developed has led to the development of a biomimetic bilayered scaffold suitable for use as a tissue engineered vascular graft (for haemodialysis access or peripheral/coronary bypass), or as an in vitro test platform to examine disease progression, pharmacological toxicity, or cardiovascular medical device testing. STATEMENT OF SIGNIFICANCE: The ability to grow large complex tissues such as blood vessels for transplantation is often hampered by the limitations of the selected biofabrication technique. Here, we sought to overcome some of the fabrication limitations for naturally occurring cardiovascular polymers (collagen/elastin) via a hierarchical approach to fabrication where each layer is built upon the previous. This approach enabled the flexibility to modify and tailor each layer's properties independently via control over polymer concentration, microstructure, and crosslinking. This simple approach facilitated us to fabricate multi-layered vascular grafts which were remodelled into high-density vascular tissue after 21-days. The fabrication approach could be translated to a myriad of other tissues while the engineered vascular graft could also be used as a test platform for drugs/medical devices or as a tissue engineering scaffold for vascular grafting for different indications.
Collapse
|
49
|
Klimek K, Ginalska G. Proteins and Peptides as Important Modifiers of the Polymer Scaffolds for Tissue Engineering Applications-A Review. Polymers (Basel) 2020; 12:E844. [PMID: 32268607 PMCID: PMC7240665 DOI: 10.3390/polym12040844] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/31/2020] [Accepted: 04/02/2020] [Indexed: 12/21/2022] Open
Abstract
Polymer scaffolds constitute a very interesting strategy for tissue engineering. Even though they are generally non-toxic, in some cases, they may not provide suitable support for cell adhesion, proliferation, and differentiation, which decelerates tissue regeneration. To improve biological properties, scaffolds are frequently enriched with bioactive molecules, inter alia extracellular matrix proteins, adhesive peptides, growth factors, hormones, and cytokines. Although there are many papers describing synthesis and properties of polymer scaffolds enriched with proteins or peptides, few reviews comprehensively summarize these bioactive molecules. Thus, this review presents the current knowledge about the most important proteins and peptides used for modification of polymer scaffolds for tissue engineering. This paper also describes the influence of addition of proteins and peptides on physicochemical, mechanical, and biological properties of polymer scaffolds. Moreover, this article sums up the major applications of some biodegradable natural and synthetic polymer scaffolds modified with proteins and peptides, which have been developed within the past five years.
Collapse
Affiliation(s)
- Katarzyna Klimek
- Chair and Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland;
| | | |
Collapse
|
50
|
Cao Y, Lee BH, Irvine SA, Wong YS, Bianco Peled H, Venkatraman S. Inclusion of Cross-Linked Elastin in Gelatin/PEG Hydrogels Favourably Influences Fibroblast Phenotype. Polymers (Basel) 2020; 12:polym12030670. [PMID: 32192137 PMCID: PMC7183321 DOI: 10.3390/polym12030670] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/16/2022] Open
Abstract
The capacity of a biomaterial to innately modulate cell behavior while meeting the mechanical property requirements of the implant is a much sought-after goal within bioengineering. Here we covalently incorporate soluble elastin into a gelatin–poly (ethylene glycol) (PEG) hydrogel for three-dimensional (3D) cell encapsulation to achieve these properties. The inclusion of elastin into a previously optimized gelatin–PEG hydrogel was then evaluated for effects on entrapped fibroblasts, with the aim to assess the hydrogel as an extracellular matrix (ECM)-mimicking 3D microenvironment for cellular guidance. Soluble elastin was incorporated both physically and covalently into novel gelatin/elastin hybrid PEG hydrogels with the aim to harness the cellular interactivity and mechanical tunability of both elastin and gelatin. This design allowed us to assess the benefits of elastin-containing hydrogels in guiding fibroblast activity for evaluation as a potential dermal replacement. It was found that a gelatin–PEG hydrogel with covalently conjugated elastin, supported neonatal fibroblast viability, promoted their proliferation from 7.3% to 13.5% and guided their behavior. The expression of collagen alpha-1(COL1A1) and elastin in gelatin/elastin hybrid gels increased 16-fold and 6-fold compared to control sample at day 9, respectively. Moreover, cells can be loaded into the hydrogel precursor solution, deposited, and the matrix cross-linked without affecting the incorporated cells adversely, thus enabling a potential injectable system for dermal wound healing.
Collapse
Affiliation(s)
- Ye Cao
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.C.); (B.H.L.); (S.A.I.); (Y.S.W.)
- The Inter-Departmental Program for Biotechnology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Bae Hoon Lee
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.C.); (B.H.L.); (S.A.I.); (Y.S.W.)
| | - Scott Alexander Irvine
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.C.); (B.H.L.); (S.A.I.); (Y.S.W.)
| | - Yee Shan Wong
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; (Y.C.); (B.H.L.); (S.A.I.); (Y.S.W.)
| | - Havazelet Bianco Peled
- Department of Chemical Engineering, Technion-Israel Institute of Technology, Haifa 32000, Israel
- Correspondence: (H.B.P.); (S.V.)
| | - Subramanian Venkatraman
- Subramanian Venkatraman, Materials Science and Engineering, National University of Singapore, Singapore 119077, Singapore
- Correspondence: (H.B.P.); (S.V.)
| |
Collapse
|