1
|
Guo W, Xu H, Liu D, Dong L, Liang T, Li B, Meng B, Chen S. 3D-Printed lattice-inspired composites for bone reconstruction. J Mater Chem B 2023; 11:7353-7363. [PMID: 37522170 DOI: 10.1039/d3tb01053h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Mechanical performance is crucial for biomedical applications of scaffolds. In this study, the stress distribution of six lattice-inspired structures was investigated using finite element simulations, and scaffolds with pre-designed structures were prepared using selective laser sintering (SLS) technology. The results showed that scaffolds with face-centered cubic (FCC) structures exhibited the highest compressive strength. Moreover, scaffolds composed of polylactic acid/anhydrous calcium hydrogen phosphate (PLA/DCPA) showed good mechanical properties and bioactivity. An in vitro study showed that these scaffolds promoted cell proliferation significantly and showed excellent osteogenic performance. Composite scaffolds with FCC structures are promising for bone tissue engineering.
Collapse
Affiliation(s)
- Wenmin Guo
- Mechanical and Energy Engineering College, Shaoyang University, Shaoyang 422000, Hunan, China
| | - Huanhuan Xu
- Mechanical and Energy Engineering College, Shaoyang University, Shaoyang 422000, Hunan, China
| | - Dachuan Liu
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Li Dong
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Ting Liang
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Bin Li
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Bin Meng
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215006, Jiangsu, China.
| | - Song Chen
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
2
|
Suryawan IGR, Ratri AK, Andrianto A, Ardiana M, Nugraha RA. Fibronectin enhances attachment of human adipose-derived mesenchymal stem cells into polytetrafluoroethylene patch during surgical closure of the atrial and ventricular septal defect. Ann Pediatr Cardiol 2023; 16:189-193. [PMID: 37876958 PMCID: PMC10593271 DOI: 10.4103/apc.apc_9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/19/2023] [Accepted: 07/03/2023] [Indexed: 10/26/2023] Open
Abstract
Background Polytetrafluoroethylene (PTFE) patch is commonly used during surgical closure for atrial septal defect (ASD) and ventricular septal defect (VSD). However, this patch has several limitations such as its inability to grow or remodel, especially in children and young adults. To tackle these limitations, we have tried to use fibronectin and human adipose-derived mesenchymal stem cells (hAMSCs) in the PTFE patch. Objective To understand the impact of fibronectin to enhance hAMSCs cell-to-cell adherence and cell-to-patch surface attachment into PTFE patches used in the surgical closure of ASD or VSD. Materials and Methods The hAMSCs were plated and fixated with 15 mL methanol and cluster of differentiation (CD) 90+, CD105+, and CD45 - antibodies were labeled with fluorescein isothiocyanate, rinsed with phosphate-buffered saline, and analyzed under a fluorescence microscope. Fibronectin solution (0.1%) was used to soak patch scaffolds for approximately 2-h duration and then dried for 20 min in the treatment group. The samples were examined with a scanning electron microscope (SEM). Results SEM examination showed incomplete attachment of the cells even after 10 days in the control group at 1.14 ± 1.13. In contrast, the treatment group showed more cells attached to the patch surface at 31.25 ± 13.28 (P ≤ 0.0001). The observation at 5 days was 17.67 ± 20.21, at 7 days was 12.11 ± 10.94, and at 10 days was 18.83 ± 23.25. There was no significant statistical difference in mean cell per view among each treatment group (P = 0.802). Conclusion Our work demonstrates that fibronectin has a positive impact on hAMSC attachment seeded onto the PTFE patch. These properties, in combination with their developmental plasticity, have generated tremendous interest in regenerative medicine.
Collapse
Affiliation(s)
- I. Gde Rurus Suryawan
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Airlangga, Dr. Soetomo General Hospital, Surabaya, Indonesia
| | - Anudya Kartika Ratri
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Airlangga, Dr. Soetomo General Hospital, Surabaya, Indonesia
| | - Andrianto Andrianto
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Airlangga, Dr. Soetomo General Hospital, Surabaya, Indonesia
| | - Meity Ardiana
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Airlangga, Dr. Soetomo General Hospital, Surabaya, Indonesia
| | - Ricardo Adrian Nugraha
- Department of Cardiology and Vascular Medicine, Faculty of Medicine Universitas Airlangga, Dr. Soetomo General Hospital, Surabaya, Indonesia
| |
Collapse
|
3
|
Bakhshandeh B, Ranjbar N, Abbasi A, Amiri E, Abedi A, Mehrabi M, Dehghani Z, Pennisi CP. Recent progress in the manipulation of biochemical and biophysical cues for engineering functional tissues. Bioeng Transl Med 2023; 8:e10383. [PMID: 36925674 PMCID: PMC10013802 DOI: 10.1002/btm2.10383] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 06/28/2022] [Accepted: 07/16/2022] [Indexed: 11/11/2022] Open
Abstract
Tissue engineering (TE) is currently considered a cutting-edge discipline that offers the potential for developing treatments for health conditions that negatively affect the quality of life. This interdisciplinary field typically involves the combination of cells, scaffolds, and appropriate induction factors for the regeneration and repair of damaged tissue. Cell fate decisions, such as survival, proliferation, or differentiation, critically depend on various biochemical and biophysical factors provided by the extracellular environment during developmental, physiological, and pathological processes. Therefore, understanding the mechanisms of action of these factors is critical to accurately mimic the complex architecture of the extracellular environment of living tissues and improve the efficiency of TE approaches. In this review, we recapitulate the effects that biochemical and biophysical induction factors have on various aspects of cell fate. While the role of biochemical factors, such as growth factors, small molecules, extracellular matrix (ECM) components, and cytokines, has been extensively studied in the context of TE applications, it is only recently that we have begun to understand the effects of biophysical signals such as surface topography, mechanical, and electrical signals. These biophysical cues could provide a more robust set of stimuli to manipulate cell signaling pathways during the formation of the engineered tissue. Furthermore, the simultaneous application of different types of signals appears to elicit synergistic responses that are likely to improve functional outcomes, which could help translate results into successful clinical therapies in the future.
Collapse
Affiliation(s)
- Behnaz Bakhshandeh
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Nika Ranjbar
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Ardeshir Abbasi
- Department of Immunology, Faculty of Medical SciencesTarbiat Modares UniversityTehranIran
| | - Elahe Amiri
- Department of Life Science Engineering, Faculty of New Sciences and TechnologyUniversity of TehranTehranIran
| | - Ali Abedi
- Department of Life Science Engineering, Faculty of New Sciences and TechnologyUniversity of TehranTehranIran
| | - Mohammad‐Reza Mehrabi
- Department of Microbial Biotechnology, School of Biology, College of ScienceUniversity of TehranTehranIran
| | - Zahra Dehghani
- Department of Biotechnology, College of ScienceUniversity of TehranTehranIran
| | - Cristian Pablo Pennisi
- Regenerative Medicine Group, Department of Health Science and TechnologyAalborg UniversityAalborgDenmark
| |
Collapse
|
4
|
Bayareh M. Active cell capturing for organ-on-a-chip systems: a review. BIOMED ENG-BIOMED TE 2022; 67:443-459. [PMID: 36062551 DOI: 10.1515/bmt-2022-0232] [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: 06/16/2022] [Accepted: 08/25/2022] [Indexed: 11/15/2022]
Abstract
Organ-on-a-chip (OOC) is an emerging technology that has been proposed as a new powerful cell-based tool to imitate the pathophysiological environment of human organs. For most OOC systems, a pivotal step is to culture cells in microfluidic devices. In active cell capturing techniques, external actuators, such as electrokinetic, magnetic, acoustic, and optical forces, or a combination of these forces, can be applied to trap cells after ejecting cell suspension into the microchannel inlet. This review paper distinguishes the characteristics of biomaterials and evaluates microfluidic technology. Besides, various types of OOC and their fabrication techniques are reported and various active cell capture microstructures are analyzed. Furthermore, their constraints, challenges, and future perspectives are provided.
Collapse
Affiliation(s)
- Morteza Bayareh
- Department of Mechanical Engineering, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
5
|
Alamdari SG, Alibakhshi A, de la Guardia M, Baradaran B, Mohammadzadeh R, Amini M, Kesharwani P, Mokhtarzadeh A, Oroojalian F, Sahebkar A. Conductive and Semiconductive Nanocomposite-Based Hydrogels for Cardiac Tissue Engineering. Adv Healthc Mater 2022; 11:e2200526. [PMID: 35822350 DOI: 10.1002/adhm.202200526] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/26/2022] [Indexed: 01/27/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide and the most common cause is myocardial infarction. Therefore, appropriate approaches should be used to repair damaged heart tissue. Recently, cardiac tissue engineering approaches have been extensively studied. Since the creation of the nature of cardiovascular tissue engineering, many advances have been made in cellular and scaffolding technologies. Due to the hydrated and porous structures of the hydrogel, they are used as a support matrix to deliver cells to the infarct tissue. In heart tissue regeneration, bioactive and biodegradable hydrogels are required by simulating native tissue microenvironments to support myocardial wall stress in addition to preserving cells. Recently, the use of nanostructured hydrogels has increased the use of nanocomposite hydrogels and has revolutionized the field of cardiac tissue engineering. Therefore, to overcome the limitation of the use of hydrogels due to their mechanical fragility, various nanoparticles of polymers, metal, and carbon are used in tissue engineering and create a new opportunity to provide hydrogels with excellent properties. Here, the types of synthetic and natural polymer hydrogels, nanocarbon-based hydrogels, and other nanoparticle-based materials used for cardiac tissue engineering with emphasis on conductive nanostructured hydrogels are briefly introduced.
Collapse
Affiliation(s)
- Sania Ghobadi Alamdari
- Department of Cell and Molecular Biology, Faculty of Basic Science, University of Maragheh, Maragheh, 83111-55181, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Abbas Alibakhshi
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, Burjassot, Valencia, 46100, Spain
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Reza Mohammadzadeh
- Department of Cell and Molecular Biology, Faculty of Basic Science, University of Maragheh, Maragheh, 83111-55181, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 94149-75516, Iran.,Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94149-75516, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran
| |
Collapse
|
6
|
Lee SO, Xie Q, Fried SD. Optimized Loopable Translation as a Platform for the Synthesis of Repetitive Proteins. ACS CENTRAL SCIENCE 2021; 7:1736-1750. [PMID: 34729417 PMCID: PMC8554844 DOI: 10.1021/acscentsci.1c00574] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Indexed: 06/13/2023]
Abstract
The expression of long proteins with repetitive amino acid sequences often presents a challenge in recombinant systems. To overcome this obstacle, we report a genetic construct that circularizes mRNA in vivo by rearranging the topology of a group I self-splicing intron from T4 bacteriophage, thereby enabling "loopable" translation. Using a fluorescence-based assay to probe the translational efficiency of circularized mRNAs, we identify several conditions that optimize protein expression from this system. Our data suggested that translation of circularized mRNAs could be limited primarily by the rate of ribosomal initiation; therefore, using a modified error-prone PCR method, we generated a library that concentrated mutations into the initiation region of circularized mRNA and discovered mutants that generated markedly higher expression levels. Combining our rational improvements with those discovered through directed evolution, we report a loopable translator that achieves protein expression levels within 1.5-fold of the levels of standard vectorial translation. In summary, our work demonstrates loopable translation as a promising platform for the creation of large peptide chains, with potential utility in the development of novel protein materials.
Collapse
|
7
|
Omer A, Al-Sharabi N, Qiu Y, Xue Y, Li Y, Fujio M, Mustafa K, Xing Z. Biological responses of dental pulp cells to surfaces modified by collagen 1 and fibronectin. J Biomed Mater Res A 2020; 108:1369-1379. [PMID: 32107841 DOI: 10.1002/jbm.a.36908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/18/2020] [Accepted: 02/24/2020] [Indexed: 01/09/2023]
Abstract
Collagen 1 (COL1) and fibronectin (FN) are extracellular matrix proteins that contribute in cell activity and involve in regulating dental pulp cells (DPCs). The purpose of this study was to investigate the effect of COL1 and FN on the behavior of DPCs. Here, DPCs were grown under three different conditions: COL1 coating, FN coating, and control group without coating. The proliferation and differentiation of DPCs were investigated. DPCs in osteogenic media were able to differentiate into osteoblastic phenotype. The morphological analysis revealed no obvious difference on the shape of cells. Cells had spread well on both coated and noncoated culture plates with slightly more spreading in the coated plates after 24 hr. The MTT analysis did not demonstrate a significant difference at 1 and 3 hr among the groups, but interestingly, the analysis disclosed more cells on the coated plates after longer cultures, which indicated a higher proliferative capacity in response to COL1 and FN. RT-PCR, Western Blotting and mineralization assays did not reveal significant differences between the coated and noncoated surfaces in relation to osteogenic differential potential. Our data suggested that the surface coating of COL1 and FN were able to promote cellular proliferation and the osteogenic differentiation tendency of DPCs was also observed in vitro.
Collapse
Affiliation(s)
- Abedelfattah Omer
- School of Stomatology, Lanzhou University, Lanzhou, People's Republic of China.,Centre for Clinical Dental Research, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Niyaz Al-Sharabi
- Centre for Clinical Dental Research, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Yingfei Qiu
- School of Stomatology, Lanzhou University, Lanzhou, People's Republic of China
| | - Ying Xue
- Centre for Clinical Dental Research, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Yi Li
- School of Stomatology, Lanzhou University, Lanzhou, People's Republic of China
| | - Masahito Fujio
- Centre for Clinical Dental Research, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway.,Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kamal Mustafa
- Centre for Clinical Dental Research, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| | - Zhe Xing
- School of Stomatology, Lanzhou University, Lanzhou, People's Republic of China.,Centre for Clinical Dental Research, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
8
|
Ramaswamy AK, Vorp DA, Weinbaum JS. Functional Vascular Tissue Engineering Inspired by Matricellular Proteins. Front Cardiovasc Med 2019; 6:74. [PMID: 31214600 PMCID: PMC6554335 DOI: 10.3389/fcvm.2019.00074] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/15/2019] [Indexed: 12/17/2022] Open
Abstract
Modern regenerative medicine, and tissue engineering specifically, has benefited from a greater appreciation of the native extracellular matrix (ECM). Fibronectin, collagen, and elastin have entered the tissue engineer's toolkit; however, as fully decellularized biomaterials have come to the forefront in vascular engineering it has become apparent that the ECM is comprised of more than just fibronectin, collagen, and elastin, and that cell-instructive molecules known as matricellular proteins are critical for desired outcomes. In brief, matricellular proteins are ECM constituents that contrast with the canonical structural proteins of the ECM in that their primary role is to interact with the cell. Of late, matricellular genes have been linked to diseases including connective tissue disorders, cardiovascular disease, and cancer. Despite the range of biological activities, this class of biomolecules has not been actively used in the field of regenerative medicine. The intent of this review is to bring matricellular proteins into wider use in the context of vascular tissue engineering. Matricellular proteins orchestrate the formation of new collagen and elastin fibers that have proper mechanical properties-these will be essential components for a fully biological small diameter tissue engineered vascular graft (TEVG). Matricellular proteins also regulate the initiation of thrombosis via fibrin deposition and platelet activation, and the clearance of thrombus when it is no longer needed-proper regulation of thrombosis will be critical for maintaining patency of a TEVG after implantation. Matricellular proteins regulate the adhesion, migration, and proliferation of endothelial cells-all are biological functions that will be critical for formation of a thrombus-resistant endothelium within a TEVG. Lastly, matricellular proteins regulate the adhesion, migration, proliferation, and activation of smooth muscle cells-proper control of these biological activities will be critical for a TEVG that recellularizes and resists neointimal formation/stenosis. We review all of these functions for matricellular proteins here, in addition to reviewing the few studies that have been performed at the intersection of matricellular protein biology and vascular tissue engineering.
Collapse
Affiliation(s)
- Aneesh K Ramaswamy
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - David A Vorp
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Justin S Weinbaum
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
9
|
Islami M, Payandeh Z, Dalir Abdolahinia E, Saburi E, Soleimanifar F, Kehtari M, Mortazavi Y, Nadri S, Darvish M. Fucosylated umbilical cord blood hematopoietic stem cell expansion on selectin-coated scaffolds. J Cell Physiol 2019; 234:22593-22603. [PMID: 31102280 DOI: 10.1002/jcp.28825] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 12/17/2022]
Abstract
Despite the advantages of transplantation of umbilical cord blood's (UCB's) hematopoietic stem cells (uHSCs) for hematologic malignancy treatment, there are two major challenges in using them: (a) Insufficient amount of uHSCs in a UCB unit; (b) a defect in uHSCs homing to bone marrow (BM) due to loose binding of their surface glycan ligands to BM's endothelium selectin receptors. To overcome these limitations, after poly l-lactic acid (PLLA) scaffold establishment and incubation of uHSCs with fucosyltransferase-VI and GDP-fucose, ex vivo expansion of these cells on selectin-coated scaffold was done. The characteristics of the cultured fucosylated and nonfucosylated cells on a two-dimensional culture system, PLLA, and a selectin-coated scaffold were evaluated by flow cytometry, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, colony-forming unit (CFU) assay, and CXCR4 expression at the messenger RNA and protein levels. According to the findings of this study, optimized attachment to the scaffold in scanning electron microscopy micrograph, maximum count of CFU, and the highest 570 nm absorption were observed in fucosylated cells expanded on selectin-coated scaffolds. Furthermore, real-time polymerase chain reaction showed the highest expression of the CXCR4 gene, and immunocytochemistry data confirmed that the CXCR4 protein was functional in this group compared with the other groups. Considered together, the results showed that selectin-coated scaffold could be a supportive structure for fucosylated uHSC expansion and homing by nanotopography. Fucosylated cells placed on the selectin-coated scaffold serve as a basal surface for cell-cell interaction and more homing potential of uHSCs. Accordingly, this procedure can also be considered as a promising technique for the hematological disorder treatment and tissue engineering applications.
Collapse
Affiliation(s)
- Maryam Islami
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Science, Karaj, Iran
| | - Zahra Payandeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Department of Medical Biotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Ehsan Saburi
- Immunogenetic and Cell Culture Department, Immunology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Soleimanifar
- Dietary Supplements and Probiotic Research Center, Alborz University of Medical Science, Karaj, Iran
| | - Mousa Kehtari
- Department of Stem Cell Biology, Stem Cell Technology Research Center, Tehran, Iran
| | - Yousef Mortazavi
- Department of Medical Biotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran.,Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Samad Nadri
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran.,Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Maryam Darvish
- Department of Medical Biotechnology, Faculty of Medicine, Arak University of Medical Science, Arāk, Iran
| |
Collapse
|
10
|
Willerth SM, Sakiyama-Elbert SE. Combining Stem Cells and Biomaterial Scaffolds for Constructing Tissues and Cell Delivery. ACTA ACUST UNITED AC 2019. [DOI: 10.3233/stj-180001] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Combining stem cells with biomaterial scaffolds serves as a promising strategy for engineering tissues for both in vitro and in vivo applications. This updated review details commonly used biomaterial scaffolds for engineering tissues from stem cells. We first define the different types of stem cells and their relevant properties and commonly used scaffold formulations. Next, we discuss natural and synthetic scaffold materials typically used when engineering tissues, along with their associated advantages and drawbacks and gives examples of target applications. New approaches to engineering tissues, such as 3D bioprinting, are described as they provide exciting opportunities for future work along with current challenges that must be addressed. Thus, this review provides an overview of the available biomaterials for directing stem cell differentiation as a means of producing replacements for diseased or damaged tissues.
Collapse
Affiliation(s)
- Stephanie M. Willerth
- Department of Mechanical Engineering, University of Victoria, VIC, Canada
- Division of Medical Sciences, University of Victoria, VIC, Canada
- International Collaboration on Repair Discoveries, University of British Columbia, Vancouver, Canada
| | | |
Collapse
|
11
|
Future Research Directions in the Design of Versatile Extracellular Matrix in Tissue Engineering. Int Neurourol J 2018; 22:S66-75. [PMID: 30068068 PMCID: PMC6077942 DOI: 10.5213/inj.1836154.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/12/2018] [Indexed: 12/19/2022] Open
Abstract
Native and artificial extracellular matrices (ECMs) have been widely applied in biomedical fields as one of the most effective components in tissue regeneration. In particular, ECM-based drugs are expected to be applied to treat diseases in organs relevant to urology, because tissue regeneration is particularly important for preventing the recurrence of these diseases. Native ECMs provide a complex in vivo architecture and native physical and mechanical properties that support high biocompatibility. However, the applications of native ECMs are limited due to their tissue-specificity and chemical complexity. Artificial ECMs have been fabricated in an attempt to create a broadly applicable scaffold by using controllable components and a uniform formulation. On the other hands, artificial ECMs fail to mimic the properties of a native ECM; consequently, their applications in tissues are also limited. For that reason, the design of a versatile, hybrid ECM that can be universally applied to various tissues is an emerging area of interest in the biomedical field.
Collapse
|
12
|
Smithmyer ME, Spohn JB, Kloxin AM. Probing fibroblast activation in response to extracellular cues with whole protein- or peptide-functionalized step-growth hydrogels. ACS Biomater Sci Eng 2018; 4:3304-3316. [PMID: 32494587 DOI: 10.1021/acsbiomaterials.8b00491] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Synthetic hydrogels with well-defined mechanical properties have become invaluable tools for probing cell response to extracellular cues including matrix stiffness and integrin binding. These synthetic matrices are often decorated with either proteins or integrin-binding peptides to promote cell adhesion and to direct or probe cell behavior. For example, both collagen I-functionalized polyacrylamide and peptide-functionalized poly(ethylene glycol) hydrogels have been instrumental in elucidating the role of the elasticity or 'stiffness' of the matrix in promoting fibroblast activation in wound healing and fibrosis. However, the two methods of promoting integrin binding are not often directly compared in the same system, partly owing to differences in material designs, despite the potential differences in the way cells interact with whole proteins and protein mimetic peptides. We hypothesized that such a comparison could provide insight into the ways integrin binding affects fibroblast activation within commonly utilized in vitro cell culture models, and more broadly, to inform the design of materials to modulate fibroblast activation in studies of wound healing and disease. To enable this comparison, we developed a method to conjugate whole proteins to step-growth poly(ethylene glycol) (PEG) hydrogels and investigated fibroblast response to protein-peptide pairs: fibronectin and PHSRN(G)10RGDS or collagen I and (POG)3POGFOGER(POG)4, which are important in matrix remodeling and relevant to fibroblast activation. With this approach, we observed that human pulmonary fibroblasts adopted a similar morphology on fibronectin and PHSRN(G)10RGDS, although with a slight increase in the percentage of alpha smooth muscle actin (αSMA) expressing cells on PHSRN(G)10RGDS. Interestingly, we observed that fibroblasts formed activated clusters on the collagen mimic (POG)3POGFOGER(POG)4 while exhibiting less activation on collagen I. This cell activation and clustering is reminiscent of fibroblast foci that are observed in lung fibrosis, suggesting the relevance of these well-defined polymer-peptide hydrogels for investigating fibrosis and decoupling biochemical and biophysical cues.
Collapse
Affiliation(s)
- Megan E Smithmyer
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Joseph B Spohn
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - April M Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, United States
| |
Collapse
|
13
|
Fibronectin promotes elastin deposition, elasticity and mechanical strength in cellularised collagen-based scaffolds. Biomaterials 2018; 180:130-142. [PMID: 30036726 DOI: 10.1016/j.biomaterials.2018.07.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 12/21/2022]
Abstract
One of the tightest bottlenecks in vascular tissue engineering (vTE) is the lack of strength and elasticity of engineered vascular wall models caused by limited elastic fiber deposition. In this study, flat and tubular collagen gel-based scaffolds were cellularised with vascular smooth muscle cells (SMCs) and supplemented with human plasma fibronectin (FN), a known master organizer of several extracellular matrix (ECM) fiber systems. The consequences of FN on construct maturation was investigated in terms of geometrical contraction, viscoelastic mechanical properties and deposition of core elastic fiber proteins. FN was retained in the constructs and promoted deposition of elastin by SMCs as well as of several proteins required for elastogenesis such as fibrillin-1, lysyl oxidase, fibulin-4 and latent TGF-β binding protein-4. Notably, gel contraction, tensile equilibrium elastic modulus and elasticity were strongly improved in tubular engineered tissues, approaching the behaviour of native arteries. In conclusion, this study demonstrates that FN exerts pivotal roles in directing SMC-mediated remodeling of scaffolds toward the production of a physiological-like, elastin-containing ECM with excellent mechanical properties. The developed FN-supplemented systems are promising for tissue engineering applications where the generation of mature elastic tissue is desired and represent valuable advanced in vitro models to investigate elastogenesis.
Collapse
|
14
|
Boffito M, Di Meglio F, Mozetic P, Giannitelli SM, Carmagnola I, Castaldo C, Nurzynska D, Sacco AM, Miraglia R, Montagnani S, Vitale N, Brancaccio M, Tarone G, Basoli F, Rainer A, Trombetta M, Ciardelli G, Chiono V. Surface functionalization of polyurethane scaffolds mimicking the myocardial microenvironment to support cardiac primitive cells. PLoS One 2018; 13:e0199896. [PMID: 29979710 PMCID: PMC6034803 DOI: 10.1371/journal.pone.0199896] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 06/15/2018] [Indexed: 11/28/2022] Open
Abstract
Scaffolds populated with human cardiac progenitor cells (CPCs) represent a therapeutic opportunity for heart regeneration after myocardial infarction. In this work, square-grid scaffolds are prepared by melt-extrusion additive manufacturing from a polyurethane (PU), further subjected to plasma treatment for acrylic acid surface grafting/polymerization and finally grafted with laminin-1 (PU-LN1) or gelatin (PU-G) by carbodiimide chemistry. LN1 is a cardiac niche extracellular matrix component and plays a key role in heart formation during embryogenesis, while G is a low-cost cell-adhesion protein, here used as a control functionalizing molecule. X-ray photoelectron spectroscopy analysis shows nitrogen percentage increase after functionalization. O1s and C1s core-level spectra and static contact angle measurements show changes associated with successful functionalization. ELISA assay confirms LN1 surface grafting. PU-G and PU-LN1 scaffolds both improve CPC adhesion, but LN1 functionalization is superior in promoting proliferation, protection from apoptosis and expression of differentiation markers for cardiomyocytes, endothelial and smooth muscle cells. PU-LN1 and PU scaffolds are biodegraded into non-cytotoxic residues. Scaffolds subcutaneously implanted in mice evoke weak inflammation and integrate with the host tissue, evidencing a significant blood vessel density around the scaffolds. PU-LN1 scaffolds show their superiority in driving CPC behavior, evidencing their promising role in myocardial regenerative medicine.
Collapse
Affiliation(s)
- Monica Boffito
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Franca Di Meglio
- Department of Public Health, University of Naples ‘Federico II’, Naples, Italy
| | - Pamela Mozetic
- Department of Engineering, Tissue Engineering Unit, Università Campus Bio-Medico di Roma, Rome, Italy
- Center for Translational Medicine, International Clinical Research Center, St.Anne’s University Hospital, Brno, Czechia
| | - Sara Maria Giannitelli
- Department of Engineering, Tissue Engineering Unit, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Irene Carmagnola
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Clotilde Castaldo
- Department of Public Health, University of Naples ‘Federico II’, Naples, Italy
| | - Daria Nurzynska
- Department of Public Health, University of Naples ‘Federico II’, Naples, Italy
| | - Anna Maria Sacco
- Department of Public Health, University of Naples ‘Federico II’, Naples, Italy
| | - Rita Miraglia
- Department of Public Health, University of Naples ‘Federico II’, Naples, Italy
| | - Stefania Montagnani
- Department of Public Health, University of Naples ‘Federico II’, Naples, Italy
| | - Nicoletta Vitale
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Mara Brancaccio
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Guido Tarone
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin, Italy
| | - Francesco Basoli
- Department of Engineering, Tissue Engineering Unit, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Alberto Rainer
- Department of Engineering, Tissue Engineering Unit, Università Campus Bio-Medico di Roma, Rome, Italy
- Institute for Photonics and Nanotechnology, National Research Council, Rome, Italy
| | - Marcella Trombetta
- Department of Engineering, Tissue Engineering Unit, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Gianluca Ciardelli
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| |
Collapse
|
15
|
Islami M, Mortazavi Y, Soleimani M, Nadri S. In vitro expansion of CD 133+ cells derived from umbilical cord blood in poly-L-lactic acid (PLLA) scaffold coated with fibronectin and collagen. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2017; 46:1025-1033. [PMID: 28782391 DOI: 10.1080/21691401.2017.1358733] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
CONTEXT Due to their renewal and potency, umbilical cord blood (UCB) stem cells have the ability to proliferate and serve as an attractive alternative source for bone marrow transplantation. However, insufficient number of haematopoietic stem cells (HSCs) in UCB is still a major constraint in clinical applications. OBJECTIVE In vitro expansion of stem cells on fibronectin (Fn)-coated poly-L-lactic acid (PLLA) scaffold can be a proper way to overcome this limitation. MATERIALS AND METHOD UCB CD133 + cells were isolated by magnetic cell sorting (MACS), and then the flowcytometry method was used for analysing CD133 + cells. Confirmed cells were seeded on the Fn-coated PLLA scaffold; also, collagen-coated PLLA scaffold, PLLA scaffold and two-dimensional (2D) culture system were expanded for 7 days. During this time, we used the flowcytometric method for analysing CD133 + cells and real-time PCR for the expression level of CXCR4 gene. The number of total cells and CD133 + cells, as well as MTT assay and colony-forming unit (CFU) assay were evaluated. RESULTS Flowcytometry data indicated that the purity of CD133 + before expansion was 93%. After 7 days, there was higher number of CD133 + cells on the Fn-coated PLLA scaffold compared to other groups. Moreover, results of MTT and colony assays showed higher viability and proliferation of CD133 + on the Fn-coated PLLA scaffold. Also, the quantity of CXCR4 gene expression increased compared to other groups. DISCUSSION The Fn-coated scaffold was the most effective scaffold for proliferation and improved the adhesiveness to the scaffold. CONCLUSION The Fn-coated PLLA scaffold could be a suitable in vitro mimicry niche over a 2D system.
Collapse
Affiliation(s)
- Maryam Islami
- a Medical Biotechnology and Nanotechnology Department , Zanjan University of Medical Sciences , Zanjan , Iran
| | - Yousef Mortazavi
- a Medical Biotechnology and Nanotechnology Department , Zanjan University of Medical Sciences , Zanjan , Iran.,b Department of Pathology, Cancer Gene Therapy Research Center , Zanjan University of Medical Sciences , Zanjan , Iran
| | - Masoud Soleimani
- c Hematology Department , Tarbiat Modares University , Tehran , Iran
| | - Samad Nadri
- a Medical Biotechnology and Nanotechnology Department , Zanjan University of Medical Sciences , Zanjan , Iran
| |
Collapse
|
16
|
Bone Formation from Porcine Dental Germ Stem Cells on Surface Modified Polybutylene Succinate Scaffolds. Stem Cells Int 2016; 2016:8792191. [PMID: 27413380 PMCID: PMC4927991 DOI: 10.1155/2016/8792191] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/05/2016] [Accepted: 05/16/2016] [Indexed: 01/09/2023] Open
Abstract
Designing and providing a scaffold are very important for the cells in tissue engineering. Polybutylene succinate (PBS) has high potential as a scaffold for bone regeneration due to its capacity in cell proliferation and differentiation. Also, stem cells from 3rd molar tooth germs were favoured in this study due to their developmentally and replicatively immature nature. In this study, porcine dental germ stem cells (pDGSCs) seeded PBS scaffolds were used to investigate the effects of surface modification with fibronectin or laminin on these scaffolds to improve cell attachment, proliferation, and osteogenic differentiation for tissue engineering applications. The osteogenic potentials of pDGSCs on these modified and unmodified foams were examined to heal bone defects and the effects of fibronectin or laminin modified PBS scaffolds on pDGSC differentiation into bone were compared for the first time. For this study, MTS assay was used to assess the cytotoxic effects of modified and unmodified surfaces. For the characterization of pDGSCs, flow cytometry analysis was carried out. Besides, alkaline phosphatase (ALP) assay, von Kossa staining, real-time PCR, CM-Dil, and immunostaining were applied to analyze osteogenic potentials of pDGSCs. The results of these studies demonstrated that pDGSCs were differentiated into osteogenic cells on fibronectin modified PBS foams better than those on unmodified and laminin modified PBS foams.
Collapse
|
17
|
Pashneh-Tala S, MacNeil S, Claeyssens F. The Tissue-Engineered Vascular Graft-Past, Present, and Future. TISSUE ENGINEERING PART B-REVIEWS 2015; 22:68-100. [PMID: 26447530 PMCID: PMC4753638 DOI: 10.1089/ten.teb.2015.0100] [Citation(s) in RCA: 451] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cardiovascular disease is the leading cause of death worldwide, with this trend predicted to continue for the foreseeable future. Common disorders are associated with the stenosis or occlusion of blood vessels. The preferred treatment for the long-term revascularization of occluded vessels is surgery utilizing vascular grafts, such as coronary artery bypass grafting and peripheral artery bypass grafting. Currently, autologous vessels such as the saphenous vein and internal thoracic artery represent the gold standard grafts for small-diameter vessels (<6 mm), outperforming synthetic alternatives. However, these vessels are of limited availability, require invasive harvest, and are often unsuitable for use. To address this, the development of a tissue-engineered vascular graft (TEVG) has been rigorously pursued. This article reviews the current state of the art of TEVGs. The various approaches being explored to generate TEVGs are described, including scaffold-based methods (using synthetic and natural polymers), the use of decellularized natural matrices, and tissue self-assembly processes, with the results of various in vivo studies, including clinical trials, highlighted. A discussion of the key areas for further investigation, including graft cell source, mechanical properties, hemodynamics, integration, and assessment in animal models, is then presented.
Collapse
Affiliation(s)
- Samand Pashneh-Tala
- Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield , Broad Lane, Sheffield, United Kingdom
| | - Sheila MacNeil
- Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield , Broad Lane, Sheffield, United Kingdom
| | - Frederik Claeyssens
- Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield , Broad Lane, Sheffield, United Kingdom
| |
Collapse
|
18
|
Drug delivery in aortic valve tissue engineering. J Control Release 2014; 196:307-23. [DOI: 10.1016/j.jconrel.2014.10.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/07/2014] [Accepted: 10/09/2014] [Indexed: 01/08/2023]
|
19
|
Knight DK, Gillies ER, Mequanint K. Biomimetic L-aspartic acid-derived functional poly(ester amide)s for vascular tissue engineering. Acta Biomater 2014; 10:3484-96. [PMID: 24769110 DOI: 10.1016/j.actbio.2014.04.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Revised: 04/04/2014] [Accepted: 04/15/2014] [Indexed: 12/17/2022]
Abstract
Functionalization of polymeric biomaterials permits the conjugation of cell signaling molecules capable of directing cell function. In this study, l-phenylalanine and l-aspartic acid were used to synthesize poly(ester amide)s (PEAs) with pendant carboxylic acid groups through an interfacial polycondensation approach. Human coronary artery smooth muscle cell (HCASMC) attachment, spreading and proliferation was observed on all PEA films. Vinculin expression at the cell periphery suggested that HCASMCs formed focal adhesions on the functional PEAs, while the absence of smooth muscle α-actin (SMαA) expression implied the cells adopted a proliferative phenotype. The PEAs were also electrospun to yield nanoscale three-dimensional (3-D) scaffolds with average fiber diameters ranging from 130 to 294nm. Immunoblotting studies suggested a potential increase in SMαA and calponin expression from HCASMCs cultured on 3-D fibrous scaffolds when compared to 2-D films. X-ray photoelectron spectroscopy and immunofluorescence demonstrated the conjugation of transforming growth factor-β1 to the surface of the functional PEA through the pendant carboxylic acid groups. Taken together, this study demonstrates that PEAs containing aspartic acid are viable biomaterials for further investigation in vascular tissue engineering.
Collapse
Affiliation(s)
- Darryl K Knight
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada
| | - Elizabeth R Gillies
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada; The Graduate Program of Biomedical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada; Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5B7, Canada.
| | - Kibret Mequanint
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada; The Graduate Program of Biomedical Engineering, The University of Western Ontario, London, Ontario N6A 5B9, Canada.
| |
Collapse
|
20
|
Shen Z, Mellati A, Bi J, Zhang H, Dai S. A thermally responsive cationic nanogel-based platform for three-dimensional cell culture and recovery. RSC Adv 2014. [DOI: 10.1039/c4ra02852j] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
21
|
Bhattacharyya A, Lin S, Sandig M, Mequanint K. Regulation of vascular smooth muscle cell phenotype in three-dimensional coculture system by Jagged1-selective Notch3 signaling. Tissue Eng Part A 2014; 20:1175-87. [PMID: 24138322 PMCID: PMC3993058 DOI: 10.1089/ten.tea.2013.0268] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 10/16/2013] [Indexed: 12/21/2022] Open
Abstract
The modulation of vascular smooth muscle cell (VSMC) phenotype is an essential element to fabricate engineered conduits of clinical relevance. In vivo, owing to their close proximity, endothelial cells (ECs) play a role in VSMC phenotype switching. Although considerable progress has been made in vascular tissue engineering, significant knowledge gaps exist on how the contractile VSMC phenotype is induced at the conclusion of the tissue fabrication process. The objectives of this study were as follows: (1) to establish ligand presentation modes on transcriptional activation of VSMC-specific genes, (2) to develop a three-dimensional (3D) coculture model using human coronary artery smooth muscle cells (HCASMCs) and human coronary artery endothelial cells (HCAECs) on porous synthetic scaffolds and, (3) to investigate EC-mediated Notch signaling in 3D cultures and the induction of the HCASMC contractile phenotype. Whereas transcriptional activation of VSMC-specific genes was not induced by presenting soluble Jagged1 and Jagged1 bound to protein G beads, a direct link between HCAEC-bound Jagged1 and HCASMC differentiation genes was observed. Our 3D culture results showed that HCASMCs seeded to scaffolds and cultured for up to 16 days readily attached, infiltrated the scaffold, proliferated, and formed dense confluent layers. HCAECs, seeded on top of an HCASMC layer, formed a distinct, separate monolayer with cell-type partitioning, suggesting that HCAEC growth was contact inhibited. While we observed EC monolayer formation with 200,000 HCAECs/scaffold, seeding 400,000 HCAECs/scaffold revealed the formation of cord-like structures akin to angiogenesis. Western blot analyses showed that 3D coculture induced an upregulation of Notch3 receptor in HCASMCs and its ligand Jagged1 in HCAECs. This was accompanied by a corresponding induction of the contractile HCASMC phenotype as demonstrated by increased expression of smooth muscle-α-actin (SM-α-actin) and calponin. Knockdown of Jagged1 with siRNA showed a reduction in SM-α-actin and calponin in cocultures, identifying a link between Jagged1 and the expression of contractile proteins in 3D cocultures. We therefore conclude that the Notch3 signaling pathway is an important regulator of VSMC phenotype and could be targeted when fabricating engineered vascular tissues.
Collapse
Affiliation(s)
- Aparna Bhattacharyya
- Graduate Program of Biomedical Engineering, The University of Western Ontario, London, Canada
| | - Shigang Lin
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Canada
| | - Martin Sandig
- Graduate Program of Biomedical Engineering, The University of Western Ontario, London, Canada
- Department of Anatomy and Cell Biology, The University of Western Ontario, London, Canada
| | - Kibret Mequanint
- Graduate Program of Biomedical Engineering, The University of Western Ontario, London, Canada
- Department of Chemical and Biochemical Engineering, The University of Western Ontario, London, Canada
| |
Collapse
|
22
|
Sartori S, Chiono V, Tonda-Turo C, Mattu C, Gianluca C. Biomimetic polyurethanes in nano and regenerative medicine. J Mater Chem B 2014; 2:5128-5144. [DOI: 10.1039/c4tb00525b] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Nature's inspiration is a promising tool to design new biomaterials especially for frontier technological areas such as tissue engineering and nanomedicine.
Collapse
Affiliation(s)
- Susanna Sartori
- Politecnico di Torino
- Dep. of Mechanical and Aerospace Engineering
- Turin, Italy
| | - Valeria Chiono
- Politecnico di Torino
- Dep. of Mechanical and Aerospace Engineering
- Turin, Italy
| | - Chiara Tonda-Turo
- Politecnico di Torino
- Dep. of Mechanical and Aerospace Engineering
- Turin, Italy
| | - Clara Mattu
- Politecnico di Torino
- Dep. of Mechanical and Aerospace Engineering
- Turin, Italy
| | - Ciardelli Gianluca
- Politecnico di Torino
- Dep. of Mechanical and Aerospace Engineering
- Turin, Italy
| |
Collapse
|
23
|
Nandakumar A, Truckenmüller R, Ahmed M, Damanik F, Santos DR, Auffermann N, de Boer J, Habibovic P, van Blitterswijk C, Moroni L. A fast process for imprinting micro and nano patterns on electrospun fiber meshes at physiological temperatures. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:3405-9. [PMID: 23447336 DOI: 10.1002/smll.201300220] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 01/31/2013] [Indexed: 05/24/2023]
Abstract
Electrospun fiber meshes are patterned at length scales comparable to or lower than their fiber diameter. Simple nano- and microgrooves and closed geometric shapes are imprinted in different tones using a fast imprint process at physiological temperatures. Human mesenchymal stromal cells cultured on patterned scaffolds show differences in cellular morphology and cytoskeleton organization. Microgrooved electrospun fibers support upregulation of alkaline phosphatase and bone morphogenetic protein-2 gene expression when cells are cultured in osteogenic medium.
Collapse
Affiliation(s)
- Anandkumar Nandakumar
- Department of Tissue Regeneration, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, PO Box 217, 7500AE Enschede, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Mottaghitalab F, Farokhi M, Zaminy A, Kokabi M, Soleimani M, Mirahmadi F, Shokrgozar MA, Sadeghizadeh M. A biosynthetic nerve guide conduit based on silk/SWNT/fibronectin nanocomposite for peripheral nerve regeneration. PLoS One 2013; 8:e74417. [PMID: 24098649 PMCID: PMC3787046 DOI: 10.1371/journal.pone.0074417] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2013] [Accepted: 07/31/2013] [Indexed: 01/12/2023] Open
Abstract
As a contribution to the functionality of nerve guide conduits (NGCs) in nerve tissue engineering, here we report a conduit processing technique through introduction and evaluation of topographical, physical and chemical cues. Porous structure of NGCs based on freeze-dried silk/single walled carbon nanotubes (SF/SWNTs) has shown a uniform chemical and physical structure with suitable electrical conductivity. Moreover, fibronectin (FN) containing nanofibers within the structure of SF/SWNT conduits produced through electrospinning process have shown aligned fashion with appropriate porosity and diameter. Moreover, fibronectin remained its bioactivity and influenced the adhesion and growth of U373 cell lines. The conduits were then implanted to 10 mm left sciatic nerve defects in rats. The histological assessment has shown that nerve regeneration has taken places in proximal region of implanted nerve after 5 weeks following surgery. Furthermore, nerve conduction velocities (NCV) and more myelinated axons were observed in SF/SWNT and SF/SWNT/FN groups after 5 weeks post implantation, indicating a functional recovery for the injured nerves. With immunohistochemistry, the higher S-100 expression of Schwann cells in SF/SWNT/FN conduits in comparison to other groups was confirmed. In conclusion, an oriented conduit of biocompatible SF/SWNT/FN has been fabricated with acceptable structure that is particularly applicable in nerve grafts.
Collapse
Affiliation(s)
- Fatemeh Mottaghitalab
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Farokhi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Zaminy
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran
| | - Mehrdad Kokabi
- Department of Polymer Engineering, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | | | - Majid Sadeghizadeh
- Department of Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
- * E-mail: (MS); (MAS)
| |
Collapse
|
25
|
3D scaffolds in tissue engineering and regenerative medicine: beyond structural templates? ACTA ACUST UNITED AC 2013. [DOI: 10.4155/pbp.13.21] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
26
|
McConnell KI, Gomez EJ, Suh J. The identity of the cell adhesive protein substrate affects the efficiency of adeno-associated virus reverse transduction. Acta Biomater 2012; 8:4073-9. [PMID: 22771459 DOI: 10.1016/j.actbio.2012.06.038] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 06/25/2012] [Accepted: 06/27/2012] [Indexed: 11/25/2022]
Abstract
Delivering genes from surfaces, called substrate-mediated gene delivery or reverse transduction, is a useful method to achieve spatial localization of gene delivery. We tested the compatibility of adeno-associated virus (AAV) vectors with various cell adhesive proteins to mediate gene delivery from surfaces. Our studies demonstrate that AAV vectors can be successfully adsorbed on collagen I, elastin, and laminin substrates leading to robust gene delivery to overlying cells. Notably, AAV immobilization on laminin yields the highest efficiency of gene expression. This increased gene expression cannot be explained by increases in the levels of virus deposition, transcriptional activity of cells, or virus vector uptake into cells. Further refinement of our knowledge of AAV interactions with extracellular matrix proteins may have important implications in a variety of applications ranging from tissue engineering to in vivo gene therapy.
Collapse
|
27
|
Biomimicking Polysaccharide Nanofibers Promote Vascular Phenotypes: A Potential Application for Vascular Tissue Engineering. Macromol Biosci 2011; 12:395-401. [DOI: 10.1002/mabi.201100336] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 10/26/2011] [Indexed: 11/07/2022]
|
28
|
Seifu DG, Isimjan TT, Mequanint K. Tissue engineering scaffolds containing embedded fluorinated-zeolite oxygen vectors. Acta Biomater 2011; 7:3670-8. [PMID: 21704199 DOI: 10.1016/j.actbio.2011.06.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Revised: 05/15/2011] [Accepted: 06/07/2011] [Indexed: 11/16/2022]
Abstract
Efficient oxygen supply is a continuing challenge for the fabrication of successful tissue engineered constructs with clinical relevance. In an effort to enhance oxygen delivery we report the feasibility of using fluorinated zeolite particles embedded in three-dimensional (3-D) polyurethane scaffolds as novel oxygen vectors. First, 1H,1H,2H,2H-perfluorodecyltriethoxysilane was successfully coupled to zeolite framework particles to examine the dose-dependent dissolved oxygen concentration. Following this, the fluorinated-zeolite (FZ) particles were embedded in 3-D tissue engineering polyurethane scaffolds. Our data demonstrates an even distribution of FZ particles in the 3-D scaffolds without affecting the scaffold porosity or pore size. Human coronary artery smooth muscle cell (HCASMC) proliferation on FZ-containing polyurethane (PCU-FZ) scaffolds was significantly greater than on control scaffolds (P=0.05). Remarkably, cell infiltration depths on the PCU-FZ scaffolds was double that on PCU control scaffolds. Taken together, our data suggest the potential of PCU-FZ scaffolds for tissue engineering with enhanced oxygen delivery to cells.
Collapse
Affiliation(s)
- Dawit G Seifu
- Department of Chemical and Biochemical Engineering, University of Western Ontario, London, Ontario, Canada N5A 5B9
| | | | | |
Collapse
|