1
|
Hussein KH, Ahmadzada B, Correa JC, Sultan A, Wilken S, Amiot B, Nyberg SL. Liver tissue engineering using decellularized scaffolds: Current progress, challenges, and opportunities. Bioact Mater 2024; 40:280-305. [PMID: 38973992 PMCID: PMC11226731 DOI: 10.1016/j.bioactmat.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 07/09/2024] Open
Abstract
Liver transplantation represents the only definitive treatment for patients with end-stage liver disease. However, the shortage of liver donors provokes a dramatic gap between available grafts and patients on the waiting list. Whole liver bioengineering, an emerging field of tissue engineering, holds great potential to overcome this gap. This approach involves two main steps; the first is liver decellularization and the second is recellularization. Liver decellularization aims to remove cellular and nuclear materials from the organ, leaving behind extracellular matrices containing different structural proteins and growth factors while retaining both the vascular and biliary networks. Recellularization involves repopulating the decellularized liver with appropriate cells, theoretically from the recipient patient, to reconstruct the parenchyma, vascular tree, and biliary network. The aim of this review is to identify the major advances in decellularization and recellularization strategies and investigate obstacles for the clinical application of bioengineered liver, including immunogenicity of the designed liver extracellular matrices, the need for standardization of scaffold fabrication techniques, selection of suitable cell sources for parenchymal repopulation, vascular, and biliary tree reconstruction. In vivo transplantation models are also summarized for evaluating the functionality of bioengineered livers. Finally, the regulatory measures and future directions for confirming the safety and efficacy of bioengineered liver are also discussed. Addressing these challenges in whole liver bioengineering may offer new solutions to meet the demand for liver transplantation and improve patient outcomes.
Collapse
Affiliation(s)
- Kamal H. Hussein
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
- Department of Surgery, Anesthesiology, and Radiology, College of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Boyukkhanim Ahmadzada
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Julio Cisneros Correa
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Ahmer Sultan
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Silvana Wilken
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Bruce Amiot
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Scott L. Nyberg
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
2
|
Rezvanian P, Álvarez-López A, Tabraue-Rubio R, Daza R, Colchero L, Elices M, Guinea GV, González-Nieto D, Pérez-Rigueiro J. Modulation of Cell Response through the Covalent Binding of Fibronectin to Titanium Substrates. J Funct Biomater 2023; 14:342. [PMID: 37504837 PMCID: PMC10381834 DOI: 10.3390/jfb14070342] [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: 05/24/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023] Open
Abstract
Titanium (Ti-6Al-4V) substrates were functionalized through the covalent binding of fibronectin, and the effect of the existence of this extracellular matrix protein on the surface of the material was assessed by employing mesenchymal stem cell (MSC) cultures. The functionalization process comprised the usage of the activation vapor silanization (AVS) technique to deposit a thin film with a high surface density of amine groups on the material, followed by the covalent binding of fibronectin to the amine groups using the N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride/N-hydroxysuccinimide (EDC/NHS) crosslinking chemistry. The biological effect of the fibronectin on murine MSCs was assessed in vitro. It was found that functionalized samples not only showed enhanced initial cell adhesion compared with bare titanium, but also a three-fold increase in the cell area, reaching values comparable to those found on the polystyrene controls. These results provide compelling evidence of the potential to modulate the response of the organism to an implant through the covalent binding of extracellular matrix proteins on the prosthesis.
Collapse
Affiliation(s)
- Parsa Rezvanian
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan 8159358686, Iran
| | - Aroa Álvarez-López
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Raquel Tabraue-Rubio
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Bioactive Surfaces S.L. C/Puerto de Navacerrada 18, Galapagar, 28260 Madrid, Spain
| | - Rafael Daza
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Luis Colchero
- Bioactive Surfaces S.L. C/Puerto de Navacerrada 18, Galapagar, 28260 Madrid, Spain
| | - Manuel Elices
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Gustavo V Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, 28040 Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Bioactive Surfaces S.L. C/Puerto de Navacerrada 18, Galapagar, 28260 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, 28040 Madrid, Spain
| |
Collapse
|
3
|
Yong J, Mellick AS, Whitelock J, Wang J, Liang K. A Biomolecular Toolbox for Precision Nanomotors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2205746. [PMID: 36055646 DOI: 10.1002/adma.202205746] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/31/2022] [Indexed: 06/15/2023]
Abstract
The application of nanomotors for cancer diagnosis and therapy is a new and exciting area of research, which when combined with precision nanomedicine, promises to solve many of the issues encountered by previous development of passive nanoparticles. The goal of this article is to introduce nanomotor and nanomedicine researchers to the deep pool of knowledge available regarding cancer cell biology and biochemistry, as well as provide a greater appreciation of the complexity of cell membrane compositions, extracellular surfaces, and their functional consequences. A short description of the nanomotor state-of-art for cancer therapy and diagnosis is first provided, as well as recommendations for future directions of the field. Then, a biomolecular targeting toolbox has been collated for researchers looking to apply their nanomaterial of choice to a biological setting, as well as providing a glimpse into currently available clinical therapies and technologies. This toolbox contains an overview of different classes of targeting molecules available for high affinity and specific targeting and cell surface targets to aid researchers in the selection of a clinical disease model and targeting methodology. It is hoped that this review will provide biological context, inspiration, and direction to future nanomotor and nanomedicine research.
Collapse
Affiliation(s)
- Joel Yong
- School of Chemical Engineering and Australian Centre for NanoMedicine, The University of New South Wales, Kensington, New South Wales, 2052, Australia
| | - Albert S Mellick
- Graduate School of Biomedical Engineering, The University of New South Wales, Kensington, New South Wales, 2052, Australia
- Ingham Institute for Applied Medical Research, Liverpool, New South Wales, 2170, Australia
| | - John Whitelock
- Graduate School of Biomedical Engineering, The University of New South Wales, Kensington, New South Wales, 2052, Australia
| | - Joseph Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Kang Liang
- School of Chemical Engineering and Australian Centre for NanoMedicine, The University of New South Wales, Kensington, New South Wales, 2052, Australia
- Graduate School of Biomedical Engineering, The University of New South Wales, Kensington, New South Wales, 2052, Australia
| |
Collapse
|
4
|
Nakashima Y, Iguchi H, Takakura K, Nakamura Y, Izumi K, Koba N, Haneda S, Tsukahara M. Adhesion Characteristics of Human Pancreatic Islets, Duct Epithelial Cells, and Acinar Cells to a Polymer Scaffold. Cell Transplant 2022; 31:9636897221120500. [PMID: 36062469 PMCID: PMC9449504 DOI: 10.1177/09636897221120500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
We reported in 2018 that among several extracellular matrices, fibronectin, type I collagen, type IV collagen, laminin I, fibrinogen, and bovine serum albumin, fibronectin is particularly useful for adhesion of porcine pancreatic tissue. Subsequently, we developed a technology that enables the chemical coating of the constituent motifs of fibronectin onto cell culture dishes. In this experiment, we used islets (purity ≥ 90%), duct epithelial cells (purity ≥ 60%), and acinar cells (purity ≥ 99%) isolated from human pancreas according to the Edmonton protocol published in 2000 and achieved adhesion to the constituent motifs of fibronectin. A solution including cGMP Prodo Islet Media was used as the assay solution. In islets, adhesion was enhanced with the constitutive motifs of fibronectin compared with uncoated islets. In the functional evaluation of islets, insulin mRNA expression and insulin secretion were enhanced by the constitutive motif of fibronectin compared with non-coated islets. The stimulation index was comparable between non-coated islets and fibronectin motifs. In duct epithelial cells, adhesion was mildly promoted by the fibronectin component compared with non-coated component, while in acinar cells, adhesion was inhibited by the fibronectin component compared with the non-coated component. These data suggest that the constitutive motifs of fibronectin are useful for the adhesion of islets and duct epithelial cells.
Collapse
Affiliation(s)
- Yoshiki Nakashima
- Center for iPS Cell Research and Application Foundation, Facility for iPS Cell Therapy, Kyoto University, Kyoto, Japan
| | - Hiroki Iguchi
- R&D Center Corporate Advanced Technology Institute Life Science Development Center, Sekisui Chemical Co., Ltd., Osaka, Japan
| | - Kenta Takakura
- R&D Center Corporate Advanced Technology Institute Life Science Development Center, Sekisui Chemical Co., Ltd., Osaka, Japan
| | - Yuta Nakamura
- R&D Center Corporate Advanced Technology Institute Life Science Development Center, Sekisui Chemical Co., Ltd., Osaka, Japan
| | | | | | - Satoshi Haneda
- R&D Center Corporate Advanced Technology Institute Life Science Development Center, Sekisui Chemical Co., Ltd., Osaka, Japan
| | - Masayoshi Tsukahara
- Center for iPS Cell Research and Application Foundation, Facility for iPS Cell Therapy, Kyoto University, Kyoto, Japan
| |
Collapse
|
5
|
Peptide-modified substrate enhances cell migration and migrasome formation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112495. [PMID: 34857281 DOI: 10.1016/j.msec.2021.112495] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/06/2021] [Accepted: 10/13/2021] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs) are cell-to-cell communication tools. Migrasomes are recently discovered microscale EVs formed at the rear ends of migrating cells, and thus are suggested to be involved in communicating with neighboring cells. In cell culture, peptide scaffolds on substrates have been used to demonstrate cellular function for regenerative medicine. In this study, we evaluated peptide scaffolds, including cell penetrating, virus fusion, and integrin-binding peptides, for their potential to enable the formation of migrasome-like vesicles. Through structural and functional analyses, we confirmed that the EVs formed on these peptide-modified substrates were migrasomes. We further noted that the peptide interface comprising cell-penetrating peptides (pVEC and R9) and virus fusion peptide (SIV) have superior properties for enabling cell migration and migrasome formation than fibronectin protein, integrin-binding peptide (RGD), or bare substrate. This is the first report of migrasome formation on peptide-modified substrates. Additionally, the combination of 95% RGD and 5% pVEC peptides provided a functional interface for effective migrasome formation and desorption of cells from the substrate via a simple ethylenediaminetetraacetic acid treatment. These results provide a functional substrate for the enhancement of migrasome formation and functional analysis.
Collapse
|
6
|
Nagase K. Thermoresponsive interfaces obtained using poly(N-isopropylacrylamide)-based copolymer for bioseparation and tissue engineering applications. Adv Colloid Interface Sci 2021; 295:102487. [PMID: 34314989 DOI: 10.1016/j.cis.2021.102487] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/09/2021] [Accepted: 07/10/2021] [Indexed: 12/11/2022]
Abstract
Poly(N-isopropylacrylamide) (PNIPAAm) is the most well-known and widely used stimuli-responsive polymer in the biomedical field owing to its ability to undergo temperature-dependent hydration and dehydration with temperature variations, causing hydrophilic and hydrophobic alterations. This temperature-dependent property of PNIPAAm provides functionality to interfaces containing PNIPAAm. Notably, the hydrophilic and hydrophobic alterations caused by the change in the temperature-responsive property of PNIPAAm-modified interfaces induce temperature-modulated interactions with biomolecules, proteins, and cells. This intrinsic property of PNIPAAm can be effectively used in various biomedical applications, particularly in bioseparation and tissue engineering applications, owing to the functionality of PNIPAAm-modified interfaces based on the temperature modulation of the interaction between PNIPAAm-modified interfaces and biomolecules and cells. This review focuses on PNIPAAm-modified interfaces in terms of preparation method, properties, and their applications. Advances in PNIPAAm-modified interfaces for existing and developing applications are also summarized.
Collapse
Affiliation(s)
- Kenichi Nagase
- Faculty of Pharmacy, Keio University, 1-5-30 Shibakoen, Minato, Tokyo 105-8512, Japan.
| |
Collapse
|
7
|
Dal Sasso E, Zamuner A, Filippi A, Romanato F, Palmosi T, Vedovelli L, Gregori D, Gómez Ribelles JL, Russo T, Gloria A, Iop L, Gerosa G, Dettin M. Covalent functionalization of decellularized tissues accelerates endothelialization. Bioact Mater 2021; 6:3851-3864. [PMID: 33937589 PMCID: PMC8065253 DOI: 10.1016/j.bioactmat.2021.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/17/2022] Open
Abstract
In the field of tissue regeneration, the lack of a stable endothelial lining may affect the hemocompatibility of both synthetic and biological replacements. These drawbacks might be prevented by specific biomaterial functionalization to induce selective endothelial cell (EC) adhesion. Decellularized bovine pericardia and porcine aortas were selectively functionalized with a REDV tetrapeptide at 10−5 M and 10−6 M working concentrations. The scaffold-bound peptide was quantified and REDV potential EC adhesion enhancement was evaluated in vitro by static seeding of human umbilical vein ECs. The viable cells and MTS production were statistically higher in functionalized tissues than in control. Scaffold histoarchitecture, geometrical features, and mechanical properties were unaffected by peptide anchoring. The selective immobilization of REDV was effective in accelerating ECs adhesion while promoting proliferation in functionalized decellularized tissues intended for blood-contacting applications. Covalent functionalization of the decellularized tissues with REDV peptide accelerates endothelialization. New covalent grafting method not inducing collagen cross-linking. Measurements through two photon miscroscopy allow the quantification of biological matrix bound peptide. The decellularized tissues can be changed by chemical procedures to promote specific cellular behaviour with ECM preservation.
Collapse
Affiliation(s)
- Eleonora Dal Sasso
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - Annj Zamuner
- Department of Industrial Engineering, University of Padua, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| | - Andrea Filippi
- LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy.,Department of Physics and Astronomy "G. Galilei", University of Padua, Padua, Italy.,Fondazione Bruno Kessler, Trento, Italy.,Institute of Pediatric Research Città della Speranza, Padua, Italy
| | - Filippo Romanato
- LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy.,Department of Physics and Astronomy "G. Galilei", University of Padua, Padua, Italy.,Institute of Pediatric Research Città della Speranza, Padua, Italy
| | - Tiziana Palmosi
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - Luca Vedovelli
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - Dario Gregori
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - José Luís Gómez Ribelles
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, València, Spain.,Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| | - Teresa Russo
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Antonio Gloria
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Laura Iop
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| | - Gino Gerosa
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| | - Monica Dettin
- Department of Industrial Engineering, University of Padua, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| |
Collapse
|
8
|
Ayo A, Laakkonen P. Peptide-Based Strategies for Targeted Tumor Treatment and Imaging. Pharmaceutics 2021; 13:pharmaceutics13040481. [PMID: 33918106 PMCID: PMC8065807 DOI: 10.3390/pharmaceutics13040481] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/03/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. The development of cancer-specific diagnostic agents and anticancer toxins would improve patient survival. The current and standard types of medical care for cancer patients, including surgery, radiotherapy, and chemotherapy, are not able to treat all cancers. A new treatment strategy utilizing tumor targeting peptides to selectively deliver drugs or applicable active agents to solid tumors is becoming a promising approach. In this review, we discuss the different tumor-homing peptides discovered through combinatorial library screening, as well as native active peptides. The different structure–function relationship data that have been used to improve the peptide’s activity and conjugation strategies are highlighted.
Collapse
Affiliation(s)
- Abiodun Ayo
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Laboratory Animal Center, HiLIFE—Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-50-4489100
| |
Collapse
|
9
|
Nagase K, Shimura M, Shimane R, Hanaya K, Yamada S, Akimoto AM, Sugai T, Kanazawa H. Selective capture and non-invasive release of cells using a thermoresponsive polymer brush with affinity peptides. Biomater Sci 2021; 9:663-674. [DOI: 10.1039/d0bm01453b] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Thermoresponsive block copolymer brush with cell affinity peptides was prepared via two steps of ATRP and subsequent click reaction. The prepared polymer brush can purify cells with high selectivity by simply changing temperature.
Collapse
Affiliation(s)
| | | | | | | | - Sota Yamada
- Faculty of Pharmacy
- Keio University
- Minato
- Japan
| | - Aya Mizutani Akimoto
- Department of Materials Engineering
- School of Engineering
- The University of Tokyo
- Bunkyo
- Japan
| | | | | |
Collapse
|
10
|
Biointerface Materials for Cellular Adhesion: Recent Progress and Future Prospects. ACTUATORS 2020. [DOI: 10.3390/act9040137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
While many natural instances of adhesion between cells and biological macromolecules have been elucidated, understanding how to mimic these adhesion events remains to be a challenge. Discovering new biointerface materials that can provide an appropriate environment, and in some cases, also providing function similar to the body’s own extracellular matrix, would be highly beneficial to multiple existing applications in biomedical and biological engineering, and provide the necessary insight for the advancement of new technology. Such examples of current applications that would benefit include biosensors, high-throughput screening and tissue engineering. From a mechanical perspective, these biointerfaces would function as bioactuators that apply focal adhesion points onto cells, allowing them to move and migrate along a surface, making biointerfaces a very relevant application in the field of actuators. While it is evident that great strides in progress have been made in the area of synthetic biointerfaces, we must also acknowledge their current limitations as described in the literature, leading to an inability to completely function and dynamically respond like natural biointerfaces. In this review, we discuss the methods, materials and, possible applications of biointerface materials used in the current literature, and the trends for future research in this area.
Collapse
|
11
|
Chen YF, Goodheart C, Rua D. The Body's Cellular and Molecular Response to Protein-Coated Medical Device Implants: A Review Focused on Fibronectin and BMP Proteins. Int J Mol Sci 2020; 21:ijms21228853. [PMID: 33238458 PMCID: PMC7700595 DOI: 10.3390/ijms21228853] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/24/2022] Open
Abstract
Recent years have seen a marked rise in implantation into the body of a great variety of devices: hip, knee, and shoulder replacements, pacemakers, meshes, glucose sensors, and many others. Cochlear and retinal implants are being developed to restore hearing and sight. After surgery to implant a device, adjacent cells interact with the implant and release molecular signals that result in attraction, infiltration of the tissue, and attachment to the implant of various cell types including monocytes, macrophages, and platelets. These cells release additional signaling molecules (chemokines and cytokines) that recruit tissue repair cells to the device site. Some implants fail and require additional revision surgery that is traumatic for the patient and expensive for the payer. This review examines the literature for evidence to support the possibility that fibronectins and BMPs could be coated on the implants as part of the manufacturing process so that the proteins could be released into the tissue surrounding the implant and improve the rate of successful implantation.
Collapse
Affiliation(s)
- Yi-Fan Chen
- Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, MD 20993, USA;
| | | | - Diego Rua
- Center for Devices and Radiological Health, Food and Drug Administration, Silver Spring, MD 20993, USA;
- Correspondence:
| |
Collapse
|
12
|
Chu W, Prodromou R, Day KN, Schneible JD, Bacon KB, Bowen JD, Kilgore RE, Catella CM, Moore BD, Mabe MD, Alashoor K, Xu Y, Xiao Y, Menegatti S. Peptides and pseudopeptide ligands: a powerful toolbox for the affinity purification of current and next-generation biotherapeutics. J Chromatogr A 2020; 1635:461632. [PMID: 33333349 DOI: 10.1016/j.chroma.2020.461632] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 02/08/2023]
Abstract
Following the consolidation of therapeutic proteins in the fight against cancer, autoimmune, and neurodegenerative diseases, recent advancements in biochemistry and biotechnology have introduced a host of next-generation biotherapeutics, such as CRISPR-Cas nucleases, stem and car-T cells, and viral vectors for gene therapy. With these drugs entering the clinical pipeline, a new challenge lies ahead: how to manufacture large quantities of high-purity biotherapeutics that meet the growing demand by clinics and biotech companies worldwide. The protein ligands employed by the industry are inadequate to confront this challenge: while featuring high binding affinity and selectivity, these ligands require laborious engineering and expensive manufacturing, are prone to biochemical degradation, and pose safety concerns related to their bacterial origin. Peptides and pseudopeptides make excellent candidates to form a new cohort of ligands for the purification of next-generation biotherapeutics. Peptide-based ligands feature excellent target biorecognition, low or no toxicity and immunogenicity, and can be manufactured affordably at large scale. This work presents a comprehensive and systematic review of the literature on peptide-based ligands and their use in the affinity purification of established and upcoming biological drugs. A comparative analysis is first presented on peptide engineering principles, the development of ligands targeting different biomolecular targets, and the promises and challenges connected to the industrial implementation of peptide ligands. The reviewed literature is organized in (i) conventional (α-)peptides targeting antibodies and other therapeutic proteins, gene therapy products, and therapeutic cells; (ii) cyclic peptides and pseudo-peptides for protein purification and capture of viral and bacterial pathogens; and (iii) the forefront of peptide mimetics, such as β-/γ-peptides, peptoids, foldamers, and stimuli-responsive peptides for advanced processing of biologics.
Collapse
Affiliation(s)
- Wenning Chu
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Raphael Prodromou
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kevin N Day
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - John D Schneible
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kaitlyn B Bacon
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - John D Bowen
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Ryan E Kilgore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Carly M Catella
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Brandyn D Moore
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Matthew D Mabe
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606
| | - Kawthar Alashoor
- Department of Biochemistry and Biophysics, University of Rochester, Rochester, NY 14642
| | - Yiman Xu
- College of Material Science and Engineering, Donghua University, 201620 Shanghai, People's Republic of China
| | - Yuanxin Xiao
- College of Textile, Donghua University, Songjiang District, Shanghai, 201620, People's Republic of China
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, North Carolina State University, 911 Partners Way room 2-009, Raleigh, NC 27606.
| |
Collapse
|
13
|
Hussein KH, Park KM, Yu L, Song SH, Woo HM, Kwak HH. Vascular reconstruction: A major challenge in developing a functional whole solid organ graft from decellularized organs. Acta Biomater 2020; 103:68-80. [PMID: 31887454 DOI: 10.1016/j.actbio.2019.12.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/18/2019] [Accepted: 12/19/2019] [Indexed: 02/07/2023]
Abstract
Bioengineering a functional organ holds great potential to overcome the current gap between the organ need and shortage of available organs. Whole organ decellularization allows the removal of cells from large-scale organs, leaving behind extracellular matrices containing different growth factors, structural proteins, and a vascular network with a bare surface. Successful application of decellularized tissues as transplantable organs is hampered by the inability to completely reline the vasculature by endothelial cells (ECs), leading to blood coagulation, loss of vascular patency, and subsequent death of reseeded cells. Therefore, an intact, continuous layer of endothelium is essential to maintain proper functioning of the vascular system, which includes the transfer of nutrients to surrounding tissues and protecting other types of cells from shear stress. Here, we aimed to summarize the available cell sources that can be used for reendothelialization in addition to different trials performed by researchers to reconstruct vascularization of decellularized solid organs. Additionally, different techniques for enhancing reendothelialization and the methods used for evaluating reendothelialization efficiency along with the future prospective applications of this field are discussed. STATEMENT OF SIGNIFICANCE: Despite the great progress in whole organ decellularization, reconstruction of vasculature within the engineered constructs is still a major roadblock. Reconstructed endothelium acts as a multifunctional barrier of vessels, which can reduce thrombosis and help delivering of oxygen and nutrients throughout the whole organ. Successful reendothelialization can be achieved through reseeding of appropriate cell types on the naked vasculature with or without modification of its surface. Here, we present the current research milestones that so far established to reconstruct the vascular network in addition to the methods used for evaluating the efficiency of reendotheilization. Thus, this review is quite significant and will aid the researchers to know where we stand toward biofabricating a transplantable organ from decellularizd extracellular matrix.
Collapse
|
14
|
Xie C. Bio‐inspired nanofunctionalisation of biomaterial surfaces: a review. BIOSURFACE AND BIOTRIBOLOGY 2019. [DOI: 10.1049/bsbt.2019.0009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
- Chaoming Xie
- Key Laboratory of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031People's Republic of China
- School of Optoelectronic Science and EngineeringUniversity of Electronic Science and Technology of ChinaChengduSichuan610031People's Republic of China
| |
Collapse
|
15
|
Fitoussi R, Beauchef G, Guéré C, André N, Vié K. Localization, fate and interactions of Emilin-1 in human skin. Int J Cosmet Sci 2019; 41:183-193. [PMID: 30843221 DOI: 10.1111/ics.12524] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/04/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Emilin-1 is a versatile protein abundant in tissues where resilience and elastic recoil are prominent and interacting with components of the extracellular matrix. Still, little is known about Emilin-1 in the skin. Therefore, we investigated Emilin-1 in the skin, its localization, its fate upon ageing, its interactions with other proteins and the effect of its knockdown. METHODS Skin explants from young or old Caucasian women, immunofluorescently labelled by anti-Emilin-1, anti-Fibrillin-1 and anti-Elastin antibodies, were analysed using confocal microscopy. Skin explants subjected to UV-induced skin ageing were also analysed. Colocalization of Emilin-1 with Collagen IV, Fibrillin-1 and Elastin was studied by multiphoton microscopy and co-immunoprecipitation. Finally, the effect of Emilin-1 extinction was studied by producing small interfering RNA (siRNA) knockdown fibroblasts and by analysing the outcome on selected genes. RESULTS In skin sections from young donors, Emilin-1 localizes similarly to Elastin and Fibrillin-1. In the papillary dermis, it shows clear and ramified structures, perpendicular to the dermo-epidermal junction that are reminiscent of the oxytalan fibres. In the reticular dermis, Emilin-1 signal appears identical to that of the elastic fibres network. Upon intrinsic or UV-induced ageing, the signal associated with Emilin-1 is drastically reduced and disorganized. Multiphoton microscopy study shows that, as expected, Emilin-1 colocalizes with Elastin. It also colocalizes with Collagen IV in the basement membrane and within dermal fibroblasts. Interaction of Emilin-1 with Elastin and Collagen IV was also found by co-immunoprecipitation. It also reveals interaction with Laminin-5. Finally, siRNA-mediated knockdown of EMILIN-1 show little effect on the expression level of the 61 genes we studied. The most striking change is a downregulation of fibroblast growth factor receptor 2 that show a decrease similar to that of EMILIN-1 itself and after 8 days a downregulation of COL6A1. CONCLUSION In skin, Emilin-1 locates in the dermis, up to the basement membrane, interacting with components of the extracellular matrix but also with the anchoring complex. These interactions are important for cell adhesion, migration, proliferation and would suggest that Emilin-1 might be important for maintaining the 3D structure of the extracellular matrix.
Collapse
Affiliation(s)
- R Fitoussi
- Laboratoires Clarins, 5 rue Ampère, 95300, Pontoise, France
| | - G Beauchef
- Laboratoires Clarins, 5 rue Ampère, 95300, Pontoise, France
| | - C Guéré
- Laboratoires Clarins, 5 rue Ampère, 95300, Pontoise, France
| | - N André
- Laboratoires Clarins, 5 rue Ampère, 95300, Pontoise, France
| | - K Vié
- Laboratoires Clarins, 5 rue Ampère, 95300, Pontoise, France
| |
Collapse
|
16
|
Munisso MC, Yamaoka T. Peptide with endothelial cell affinity and antiplatelet adhesion property to improve hemocompatibility of blood‐contacting biomaterials. Pept Sci (Hoboken) 2019. [DOI: 10.1002/pep2.24114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Maria Chiara Munisso
- Department of Biomedical EngineeringNational Cerebral and Cardiovascular Center Research Institute Suita Osaka Japan
| | - Tetsuji Yamaoka
- Department of Biomedical EngineeringNational Cerebral and Cardiovascular Center Research Institute Suita Osaka Japan
| |
Collapse
|
17
|
Bai L, Zhao J, Li Q, Guo J, Ren X, Xia S, Zhang W, Feng Y. Biofunctionalized Electrospun PCL‐PIBMD/SF Vascular Grafts with PEG and Cell‐Adhesive Peptides for Endothelialization. Macromol Biosci 2018; 19:e1800386. [DOI: 10.1002/mabi.201800386] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/08/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Lingchuang Bai
- School of Chemical Engineering and TechnologyTianjin University Yaguan Road 135 Tianjin 300350 China
| | - Jing Zhao
- School of Chemical Engineering and TechnologyTianjin University Yaguan Road 135 Tianjin 300350 China
| | - Qian Li
- School of Chemical Engineering and TechnologyTianjin University Yaguan Road 135 Tianjin 300350 China
| | - Jintang Guo
- School of Chemical Engineering and TechnologyTianjin University Yaguan Road 135 Tianjin 300350 China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin) Tianjin 300350 China
| | - Xiangkui Ren
- School of Chemical Engineering and TechnologyTianjin University Yaguan Road 135 Tianjin 300350 China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin) Tianjin 300350 China
- Key Laboratory of Systems Bioengineering (Ministry of Education)Tianjin University Tianjin 300072 China
| | - Shihai Xia
- Department of Hepatopancreatobiliary and Splenic MedicineAffiliated HospitalLogistics University of People's Armed Police Force 220 Chenglin Road Tianjin 300162 China
| | - Wencheng Zhang
- Department of Physiology and PathophysiologyLogistics University of Chinese People's Armed Police Force Tianjin 300309 China
| | - Yakai Feng
- School of Chemical Engineering and TechnologyTianjin University Yaguan Road 135 Tianjin 300350 China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin) Tianjin 300350 China
- Key Laboratory of Systems Bioengineering (Ministry of Education)Tianjin University Tianjin 300072 China
| |
Collapse
|
18
|
Improving functional re-endothelialization of acellular liver scaffold using REDV cell-binding domain. Acta Biomater 2018; 78:151-164. [PMID: 30071351 DOI: 10.1016/j.actbio.2018.07.046] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 07/02/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022]
Abstract
Engineering of functional vascularized liver tissues holds great promise in addressing donor organ shortage for transplantation. Whole organ decellularization is a cell removal method that retains the native vascular structures of the organ such that it can be anastomosed with the recipient circulation after recellularization with healthy cells. However, a main hurdle to successful implantation of bioengineered organ is the inability to efficiently re-endothelialize the vasculature with a functional endothelium, resulting in blood clotting which is the primary cause of failure in early transplant studies. Here, we present an efficient approach for enhancing re-endothelialization of decellularized rat liver scaffolds by conjugating the REDV cell-binding domain to improve attachment of endothelial cells (EC) on vascular wall surfaces. In order to facilitate expression and purification of the peptide, REDV was fused with elastin-like peptide (ELP) that confers thermally triggered aggregation behavior to the fusion protein. After validating the adhesive properties of the REDV-ELP peptide, we covalently coupled REDV-ELP to the blood vasculature of decellularized rat livers and seeded EC using perfusion of the portal vein. We showed that REDV-ELP increased cell attachment, spreading and proliferation of EC within the construct resulting in uniform endothelial lining of the scaffold vasculature. We further observed that REDV-ELP conjugation dramatically reduced platelet adhesion and activation. Altogether, our results demonstrate that this method allowed functional re-endothelialization of liver scaffold and show great potential toward the generation of functional bioengineered liver for long-term transplantation. STATEMENT OF SIGNIFICANCE There is a critical need for novel organ replacement therapies as the grafts for transplantation fall short of demand. Recent advances in tissue engineering, through the use of decellularized scaffolds, have opened the possibility that engineered grafts could be used as substitutes for donor livers. However, successful implantation has been challenged by the inability to create a functional vasculature. Our research study reports a new strategy to increase efficiency of endothelialization by increasing the affinity of the vascular matrix for endothelial cells. We functionalized decellularized liver scaffold using elastin-like peptides grafted with REDV cell binding domain. We showed that REDV-ELP conjugation improve endothelial cell attachment and proliferation within the scaffold, demonstrating the feasibility of re-endothelializing a whole liver vasculature using our technique.
Collapse
|
19
|
Flora T, de Torre IG, Quintanilla L, Alonso M, Rodríguez-Cabello JC. Spatial control and cell adhesion selectivity on model gold surfaces grafted with elastin-like recombinamers. Eur Polym J 2018. [DOI: 10.1016/j.eurpolymj.2018.06.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
|
21
|
Morita S, Takasu A. Adhesion control of human umbilical vein endothelial cells using clickable poly(2-oxazoline)-grafted biosynthesized extracellular matrix protein. POLYMER 2018. [DOI: 10.1016/j.polymer.2017.12.059] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
22
|
Tang J, Saito T. Human plasma fibronectin promotes proliferation and differentiation of odontoblast. J Appl Oral Sci 2017; 25:299-309. [PMID: 28678949 PMCID: PMC5482253 DOI: 10.1590/1678-7757-2016-0442] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/18/2016] [Indexed: 11/21/2022] Open
Abstract
Objective To assess the effect of fibronectin (Fn) and porcine type I collagen (PCOL) on odontoblast-like cells in vitro. Material and Methods Rat odontoblast-like cells (MDPC-23 cells) were inoculated and cultured on Fn-coated or type I collagen-coated substrates. Proliferation assay, alkaline phosphatase activity (ALP activity), mRNA expression of hard tissue-forming markers, and Alizarin red staining were investigated over a period of 10 days. Results Cells maintained a high proliferation activity on Fn and PCOL even at a low seeding concentration (0.5×104/mL) as demonstrated by CCK-8 assay. The proliferation activity of cells on Fn increases in a concentration-dependent manner while it reached a plateau after 10 µg/mL. Cells adopted long, thin and spindle shape on Fn(10-50) and PCOL. Parallel actin filaments were observed in MDPC-23 cells cultured on Fn and PCOL. ALP activity was markedly up-regulated on Fn and PCOL-coated surfaces. Importantly, gene expression of BSP (Fn10: 2.44±0.32; Fn20: 3.05±0.01; Fn30: 2.90±0.21; Fn40: 2.74±0.30; Fn50: 2.64±0.12; PCOL: 2.20±0.03) and OCN (Fn10: 2.52±0.23; Fn20: 2.28±0.24; Fn30: 2.34±0.21; Fn40: 2.34±0.25; Fn50: 2.20±0.22; PCOL: 1.56±0.16) was significantly enhanced on Fn and PCOL substrates as compared with control; moreover, expression of integrin beta 1 (ITGB1), an ubiquitous cell surface receptor was augmented in Fn(10-50) and PCOL groups simultaneously. In accordance with the ALP activity and gene expression data, calcific deposition in cells grown on Fn(10-50) and PCOL was observed as well. Conclusion Despite the limitation of this study, the findings indicate that a surface coating of Fn enhances the proliferation, differentiation and mineralization of odontoblast-like cells by activation of integrin beta 1 (ITG B1). The promoting effects of Fn on MDPC-23 cells were achieved at a comparatively lower coating concentration than type I collagen (300 µg/mL). Specifically, it is suggested that the optimum coating concentration of Fn to be 10 µg/mL.
Collapse
Affiliation(s)
- Jia Tang
- Health Sciences University of Hokkaido, School of Dentistry, Department of Oral Rehabilitation, Division of Clinical Cariology and Endodontology, Hokkaido, Japan
| | - Takashi Saito
- Health Sciences University of Hokkaido, School of Dentistry, Department of Oral Rehabilitation, Division of Clinical Cariology and Endodontology, Hokkaido, Japan
| |
Collapse
|
23
|
Silva CCF, Menezes MC, Palomino M, Oliveira AK, Iwai LK, Faria M, Portaro FV. Peptides derived from plasma proteins released by bothropasin, a metalloprotease present in the Bothrops jararaca venom. Toxicon 2017; 137:65-72. [PMID: 28690013 DOI: 10.1016/j.toxicon.2017.07.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 07/04/2017] [Accepted: 07/06/2017] [Indexed: 12/31/2022]
Abstract
Viperid snake venoms contain proteases that affect hemostasis by degrading important proteins such as those that participate in the coagulation cascade. The Bothrops jararaca venom presents as its main components metallo and serine proteases, which comprise around 65% of the venom composition. Bothropasin is a hemorrhagic metalloprotease from the B. jararaca venom which causes disruption of the basement membrane of the vascular endothelium, resulting in bleeding. Although the bothropasin ability to degrade plasmatic and extracellular matrix proteins in vitro has been described, the primary sequence of the released peptides is unknown. This research study presents the peptide identification from both fibrinogen and fibronectin, generated by bothropasin proteolytic activity. Among the fibrinogen derived peptides identified by mass spectrometry, analogous of endogenous products like the fibrinopeptides A and B were found, as well as other sequences described in the literature with vasoactive or antiangiogenic properties. A series of peptides derived from fibronectin by the action of bothropasin were described, and for most of them no biological activity has been described. However, exceptionally a peptide that is known as a bond site for B cells was found. This study indicates that, beyond to the degradation of human proteins, bothropasin can generate bioactive peptides, which may participate in the envenoming process by Bothrops snakes. Also important, the knowledge of the formed peptides, based on the cleavage sites of the hydrolyzed proteins, provided the opportunity to study the primary specificity of bothropasin.
Collapse
Affiliation(s)
| | - Milene Cristina Menezes
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| | - Miryam Palomino
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| | - Ana Karina Oliveira
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| | - Leo Kei Iwai
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| | - Marcella Faria
- Special Laboratory of Applied Toxinology, Center of Toxins, Immune-Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| | | |
Collapse
|
24
|
Dennis JE, Haynesworth SE, Young RG, Caplan AI. Osteogenesis in Marrow-Derived Mesenchymal Cell Porous Ceramic Composites Transplanted Subcutaneously: Effect of Fibronectin and Laminin on Cell Retention and Rate of Osteogenic Expression. Cell Transplant 2017; 1:23-32. [PMID: 1344289 DOI: 10.1177/096368979200100106] [Citation(s) in RCA: 228] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Cultured-expanded rat marrow-derived mesenchymal cells differentiate into osteoblasts when combined with a porous calcium phosphate delivery vehicle and subsequently implanted in vivo. In this study, the effects of ceramic pretreatment with the cell-binding proteins fibronectin and laminin on the osteogenic expression of marrow-derived mesenchymal cells were assessed by scanning electron microscopy, [3H]-thymidine-labeled cell quantitation, and histological evaluation of bone formation. Scanning electron microscopic observations showed that marrow-derived mesenchymal cells rapidly spread and attach to both fibronectin- or laminin-adsorbed ceramic surfaces but retain a rounded morphology on untreated ceramic surfaces. Quantitation of [3H]-thymidine labeled cells demonstrated that laminin and fibronectin preadsorbed ceramics retain approximately double the number of marrow-derived mesenchymal cells than do untreated ceramics harvested 1 wk postimplantation. Histological observations indicate that the amount of time required to first detect osteogenesis was shortened significantly by pretreatment of the ceramic with either fibronectin or laminin. Fibronectin- and laminin-coated ceramic composite samples were observed to contain bone within 2 wk postimplantation, while in untreated ceramic the earliest observation of bone was at 4 wk postimplantation. A comparison was made of the initial cell-loading, in vivo cell retention characteristics, and rate of osteogenesis initiation of marrow-derived mesenchymal cells on two types of ceramic with different pore structure and chemical composition, with and without preadsorption with fibronectin or laminin. “Biphasic” ceramics contain randomly distributed pores 200-400 μm in diameter, and “coral-based” ceramics have continuous pores of approximately 200 μm in diameter. Laminin or fibronectin preadsorption significantly increases the number of cells retained in all ceramic test groups by day 7 postimplantation. In addition, by day 7 postimplantation, the biphasic ceramics retain a significantly greater number of cells for all test groups than do coral-based ceramics. The biphasic ceramics consistently have more specimens positive for bone with the identical cell-loading conditions used throughout this study. These results indicate that the retention of cells within the ceramic is an important factor for optimization of marrow mesenchymal cell initiated bone formation. The retention of cells within ceramics is augmented by the adsorption of the cell-binding proteins laminin and fibronectin, but this effect varies depending on ceramic pore structure and/or chemical composition.
Collapse
Affiliation(s)
- J E Dennis
- Department of Biology, Case Western Reserve University, Cleveland, OH 44106
| | | | | | | |
Collapse
|
25
|
Initial cell adhesion of three cell types in the presence and absence of serum proteins. Histochem Cell Biol 2017; 148:273-288. [DOI: 10.1007/s00418-017-1571-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2017] [Indexed: 10/19/2022]
|
26
|
Tanner MR, Pennington MW, Laragione T, Gulko PS, Beeton C. KCa1.1 channels regulate β 1-integrin function and cell adhesion in rheumatoid arthritis fibroblast-like synoviocytes. FASEB J 2017; 31:3309-3320. [PMID: 28428266 DOI: 10.1096/fj.201601097r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 04/05/2017] [Indexed: 01/01/2023]
Abstract
Large-conductance calcium-activated potassium channel (KCa1.1; BK, Slo1, MaxiK, KCNMA1) is the predominant potassium channel expressed at the plasma membrane of rheumatoid arthritis fibroblast-like synoviocytes (RA-FLSs) isolated from the synovium of patients with RA. It is a critical regulator of RA-FLS migration and invasion and therefore represents an attractive target for the therapy of RA. However, the molecular mechanisms by which KCa1.1 regulates RA-FLS invasiveness have remained largely unknown. Here, we demonstrate that KCa1.1 regulates RA-FLS adhesion through controlling the plasma membrane expression and activation of β1 integrins, but not α4, α5, or α6 integrins. Blocking KCa1.1 disturbs calcium homeostasis, leading to the sustained phosphorylation of Akt and the recruitment of talin to β1 integrins. Interestingly, the pore-forming α subunit of KCa1.1 coimmunoprecipitates with β1 integrins, suggesting that this physical association underlies the functional interaction between these molecules. Together, these data outline a new signaling mechanism by which KCa1.1 regulates β1-integrin function and therefore invasiveness of RA-FLSs.-Tanner, M. R., Pennington, M. W., Laragione, T., Gulko, P. S., Beeton, C. KCa1.1 channels regulate β1-integrin function and cell adhesion in rheumatoid arthritis fibroblast-like synoviocytes.
Collapse
Affiliation(s)
- Mark R Tanner
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA.,Graduate School of Biomedical Sciences, Baylor College of Medicine, Houston, Texas, USA
| | | | - Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Pércio S Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA; .,Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas, USA.,Center for Drug Discovery, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
27
|
Investigation of cellular response to covalent immobilization of peptide and hydrophobic attachment of peptide amphiphiles on substrates. Biochem Eng J 2017. [DOI: 10.1016/j.bej.2016.10.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
28
|
Munisso MC, Yamaoka T. Novel peptides for small-caliber graft functionalization selected by a phage display of endothelial-positive/platelet-negative combined selection. J Mater Chem B 2017; 5:9354-9364. [DOI: 10.1039/c7tb02652h] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
A new protocol to identify peptides with EPCs high affinity and at the same time the ability to suppress the interaction with platelets was presented.
Collapse
Affiliation(s)
- Maria Chiara Munisso
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center Research Institute
- Suita
- Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center Research Institute
- Suita
- Japan
| |
Collapse
|
29
|
Kambe Y, Murakoshi A, Urakawa H, Kimura Y, Yamaoka T. Vascular induction and cell infiltration into peptide-modified bioactive silk fibroin hydrogels. J Mater Chem B 2017; 5:7557-7571. [DOI: 10.1039/c7tb02109g] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A vascular-inducing peptide was produced recombinantly to stimulate the replacement of silk fibroin hydrogels by vascularized tissuesin vivo.
Collapse
Affiliation(s)
- Yusuke Kambe
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center (NCVC) Research Institute
- Suita
- Japan
| | - Akie Murakoshi
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center (NCVC) Research Institute
- Suita
- Japan
- Department of Biobased Materials Science
| | - Hiroshi Urakawa
- Department of Biobased Materials Science
- Kyoto Institute of Technology
- Kyoto 606-8585
- Japan
| | - Yoshiharu Kimura
- Center for Fiber and Textile Sience
- Kyoto Institute of Technology
- Kyoto 606-8585
- Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering
- National Cerebral and Cardiovascular Center (NCVC) Research Institute
- Suita
- Japan
| |
Collapse
|
30
|
Challenges for Cartilage Regeneration. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2017. [DOI: 10.1007/978-3-662-53574-5_14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
Zhao J, Li Q, Hao X, Ren X, Guo J, Feng Y, Shi C. Multi-targeting peptides for gene carriers with high transfection efficiency. J Mater Chem B 2017; 5:8035-8051. [DOI: 10.1039/c7tb02012k] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Non-viral gene carriers for gene therapy have been developed for many years.
Collapse
Affiliation(s)
- Jing Zhao
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Qian Li
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Xuefang Hao
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Xiangkui Ren
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- China
- Tianjin University-Helmholtz-Zentrum Geesthacht
| | - Jintang Guo
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- China
- Tianjin University-Helmholtz-Zentrum Geesthacht
| | - Yakai Feng
- School of Chemical Engineering and Technology
- Tianjin University
- Tianjin 300350
- China
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin)
| | - Changcan Shi
- Institute of Biomaterials and Engineering
- Wenzhou Medical University
- Wenzhou
- China
- Wenzhou Institute of Biomaterials and Engineering
| |
Collapse
|
32
|
Kesireddy V, Kasper FK. Approaches for building bioactive elements into synthetic scaffolds for bone tissue engineering. J Mater Chem B 2016; 4:6773-6786. [PMID: 28133536 PMCID: PMC5267491 DOI: 10.1039/c6tb00783j] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Bone tissue engineering (BTE) is emerging as a possible solution for regeneration of bone in a number of applications. For effective utilization, BTE scaffolds often need modifications to impart biological cues that drive diverse cellular functions such as adhesion, migration, survival, proliferation, differentiation, and biomineralization. This review provides an outline of various approaches for building bioactive elements into synthetic scaffolds for BTE and classifies them broadly under two distinct schemes; namely, the top-down approach and the bottom-up approach. Synthetic and natural routes for top-down approaches to production of bioactive constructs for BTE, such as generation of scaffold-extracellular matrix (ECM) hybrid constructs or decellularized and demineralized scaffolds, are provided. Similarly, traditional scaffold-based bottom-up approaches, including growth factor immobilization or peptide-tethered scaffolds, are provided. Finally, a brief overview of emerging bottom-up approaches for generating biologically active constructs for BTE is given. A discussion of the key areas for further investigation, challenges, and opportunities is also presented.
Collapse
Affiliation(s)
- Venu Kesireddy
- Department of Orthodontics, The University of Texas Health Science Center at Houston, School of Dentistry
| | - F. Kurtis Kasper
- Department of Orthodontics, The University of Texas Health Science Center at Houston, School of Dentistry
| |
Collapse
|
33
|
Wronska MA, O'Connor IB, Tilbury MA, Srivastava A, Wall JG. Adding Functions to Biomaterial Surfaces through Protein Incorporation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5485-5508. [PMID: 27164952 DOI: 10.1002/adma.201504310] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 03/16/2016] [Indexed: 06/05/2023]
Abstract
The concept of biomaterials has evolved from one of inert mechanical supports with a long-term, biologically inactive role in the body into complex matrices that exhibit selective cell binding, promote proliferation and matrix production, and may ultimately become replaced by newly generated tissues in vivo. Functionalization of material surfaces with biomolecules is critical to their ability to evade immunorecognition, interact productively with surrounding tissues and extracellular matrix, and avoid bacterial colonization. Antibody molecules and their derived fragments are commonly immobilized on materials to mediate coating with specific cell types in fields such as stent endothelialization and drug delivery. The incorporation of growth factors into biomaterials has found application in promoting and accelerating bone formation in osteogenerative and related applications. Peptides and extracellular matrix proteins can impart biomolecule- and cell-specificities to materials while antimicrobial peptides have found roles in preventing biofilm formation on devices and implants. In this progress report, we detail developments in the use of diverse proteins and peptides to modify the surfaces of hard biomaterials in vivo and in vitro. Chemical approaches to immobilizing active biomolecules are presented, as well as platform technologies for isolation or generation of natural or synthetic molecules suitable for biomaterial functionalization.
Collapse
Affiliation(s)
- Małgorzata A Wronska
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Iain B O'Connor
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Maura A Tilbury
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - Akshay Srivastava
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| | - J Gerard Wall
- Microbiology and Center for Research in Medical Devices (CÚRAM), National University of Ireland, Galway, Ireland
| |
Collapse
|
34
|
Butruk-Raszeja BA, Dresler MS, Kuźmińska A, Ciach T. Endothelialization of polyurethanes: Surface silanization and immobilization of REDV peptide. Colloids Surf B Biointerfaces 2016; 144:335-343. [PMID: 27110909 DOI: 10.1016/j.colsurfb.2016.04.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 04/05/2016] [Accepted: 04/07/2016] [Indexed: 01/19/2023]
Abstract
The paper presents method for chemical immobilization of arginine-glutamic acid-aspartic acid-valine (REDV) peptide on polyurethane surface. The peptide has been covalently bonded using silanes as a spacer molecules. The aim of this work was to investigate the proposed modification process and assess its biological effectiveness, especially in contact with blood and endothelial cells. Physicochemical properties were examined in terms of wettability, atomic composition and density of introduced functional groups and peptide molecules. Experiments with blood showed that material coating reduced number of surface-adhered platelets and fibrinogen molecules. In contrast to polyurethane (PU), there were no blood components deposited on REDV-modified materials (PU-REDV); fibrinogen adsorption on PU-REDV surface has been strongly reduced compared to PU. Analysis of cell adhesion after 1, 2, 3, 4, and 5 days of culture showed a significant increase of the cell-coated area on PU-REDV compared to PU. However, an intense cell growth appeared also on the control surface modified without the addition of REDV. Thus, the positive effect of REDV peptide on the adhesion of HUVEC could not be unequivocally confirmed.
Collapse
Affiliation(s)
- Beata A Butruk-Raszeja
- Laboratory of Biomedical Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warynskiego 1, 00-645 Warsaw, Poland.
| | - Magdalena S Dresler
- Laboratory of Biomedical Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warynskiego 1, 00-645 Warsaw, Poland
| | - Aleksandra Kuźmińska
- Laboratory of Biomedical Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warynskiego 1, 00-645 Warsaw, Poland
| | - Tomasz Ciach
- Laboratory of Biomedical Engineering, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warynskiego 1, 00-645 Warsaw, Poland; CEZAMAT PW, Polna 50, 00-644 Warsaw, Poland
| |
Collapse
|
35
|
Hastings CL, Roche ET, Ruiz-Hernandez E, Schenke-Layland K, Walsh CJ, Duffy GP. Drug and cell delivery for cardiac regeneration. Adv Drug Deliv Rev 2015; 84:85-106. [PMID: 25172834 DOI: 10.1016/j.addr.2014.08.006] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Revised: 07/24/2014] [Accepted: 08/15/2014] [Indexed: 12/12/2022]
Abstract
The spectrum of ischaemic cardiomyopathy, encompassing acute myocardial infarction to congestive heart failure is a significant clinical issue in the modern era. This group of diseases is an enormous source of morbidity and mortality and underlies significant healthcare costs worldwide. Cardiac regenerative therapy, whereby pro-regenerative cells, drugs or growth factors are administered to damaged and ischaemic myocardium has demonstrated significant potential, especially preclinically. While some of these strategies have demonstrated a measure of success in clinical trials, tangible clinical translation has been slow. To date, the majority of clinical studies and a significant number of preclinical studies have utilised relatively simple delivery methods for regenerative therapeutics, such as simple systemic administration or local injection in saline carrier vehicles. Here, we review cardiac regenerative strategies with a particular focus on advanced delivery concepts as a potential means to enhance treatment efficacy and tolerability and ultimately, clinical translation. These include (i) delivery of therapeutic agents in biomaterial carriers, (ii) nanoparticulate encapsulation, (iii) multimodal therapeutic strategies and (iv) localised, minimally invasive delivery via percutaneous transcatheter systems.
Collapse
|
36
|
Yamada T, Takasu A. Click Grafting of Alkyne-containing Vinyl Polymers onto Biosynthesized Extracellular Matrix Protein Containing Azide Functionality and Adhesion Control of Human Umbilical Vein Endothelial Cells. RSC Adv 2015; 5:41445-41456. [PMID: 26294960 PMCID: PMC4539266 DOI: 10.1039/c5ra04643b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In vivo incorporation of a phenylalanine (Phe) analogue, p-azidophenylalanine (p-N3Phe) into an artificial extracellular matrix protein (aECM-CS5-ELF) was accomplished using a bacterial expression host that harbors the mutant phenylalanyl-tRNA synthetase (PheRS) with an enlarged binding pocket, in which the Ala294Gly/Thr251Gly mutant PheRS (PheRS**) was expressed under the control of T7 promoters. In this study, biosynthesized aECM-CS5-ELF containing p-N3Phe (aECM-CS5-ELF-N3) was coupled with alkyne-containing vinyl polymers prepared via controlled radical polymerization of three vinyl monomers, (styrene, acrylamide, and N-isopropylacrylamide) using a trithiocarbonate as the RAFT agent. Grafting of the vinyl polymers onto the aECM was accomplished via a copper-catalyzed alkyne-azide click reaction. The lower critical transition temperature (LCST) was evaluated, as well as the solubility in aqueous and organic media, which are dependent on the incorporation ratio of p-N3Phe and species of graft chains, in which the LCST behavior was altered remarkably when poly(N-isopropylacrylamide) moieties were attached as side chains. Circular dichroism measurements indicate conformational change was not induced by the grafting. Specific adhesion of human umbilical vein endothelial cells (HUVECs) onto the aECM-CS5-ELF-N3-graft-poly(N-isopropylacrylamide) composite surface and subsequent temperature-sensitive detachment were also demonstrated.
Collapse
Affiliation(s)
- Tomoki Yamada
- Department of Frontier Materials, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan, Telephone: +81-52-735-7159; Fax: +81-52-735-5342
| | - Akinori Takasu
- Department of Frontier Materials, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan, Telephone: +81-52-735-7159; Fax: +81-52-735-5342
| |
Collapse
|
37
|
Liu R, Chen X, Gellman SH, Masters KS. Nylon-3 polymers that enable selective culture of endothelial cells. J Am Chem Soc 2014; 135:16296-9. [PMID: 24156536 DOI: 10.1021/ja408634a] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Substrates that selectively encourage the growth of specific cell types are valuable for the engineering of complex tissues. Some cell-selective peptides have been identified from extracellular matrix proteins; these peptides have proven useful for biomaterials-based approaches to tissue repair or regeneration. However, there are very few examples of synthetic materials that display selectivity in supporting cell growth. We describe nylon-3 polymers that support in vitro culture of endothelial cells but do not support the culture of smooth muscle cells or fibroblasts. These materials may be promising for vascular biomaterials applications.
Collapse
|
38
|
Wang JT, Liu Y, Kan X, Liu M, Lu JG. Cilengitide, a small molecule antagonist, targeted to integrin αν inhibits proliferation and induces apoptosis of laryngeal cancer cells in vitro. Eur Arch Otorhinolaryngol 2014; 271:2233-40. [PMID: 24515920 DOI: 10.1007/s00405-014-2918-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 01/24/2014] [Indexed: 10/25/2022]
Abstract
Cilengitide is a chemical synthesis cyclopeptide containing RGD sequence, which can be used as a small molecule antagonist targeted to integrin αν (ITGAV). The aim of present study was to investigate the effect on proliferation and cell apoptosis of the cilengitide in laryngeal cancer cells. In the study, we have treatmented the cultured cells of laryngeal cancer (Hep-2) with cilengitide. After the medication, the proliferation of the Hep-2 cells was detected by MTT assay, the expression of ITGAV was detected by RT-PCR and the activity of caspase-3 protein was detected by a specialized kit. RGD linear peptides (GRGDSP), non-RGD linear peptide (GRGESP), and 5-fluorouracil (5-Fu) were used as controls. Results showed that the proliferation of Hep-2 cells was signally inhibited by the cilengitide with a time and dose compliance. Its inhibition effect was significantly higher than that of 5-Fu and GRGDSP, but the GRGESP showed no obvious inhibitory effect. After intervene of cilengitide, the activity of caspase-3 protein of Hep-2 cells was significantly increased, and the expression of ITGAV was significantly decreased. 5-Fu significantly inhibited the proliferation of Hep-2 cells, but no significant changes of ITGAV expression were observed. In conclusion, cilengitide can significantly down-regulate ITGAV expression and inhibit cell proliferation in laryngeal cancer cells, it will also to induce cell apoptosis through caspase-3 pathway. Therefore, it could be as a kind of effective chemotherapy drugs that will be used in clinical treatment of the laryngeal cancer.
Collapse
Affiliation(s)
- Jing Ting Wang
- Department of Otolaryngology/Head and Neck Surgery, Second Affiliated Hospital of Harbin Medical University, No. 246, Xue Fu Road, Harbin City, 150081, People's Republic of China
| | | | | | | | | |
Collapse
|
39
|
Abstract
Tissue engineering aims to create, repair and/or replace tissues and organs by using cells, scaffolds, biologically active molecules and physiologic signals. It is an interdisciplinary field that integrates aspects of engineering, chemistry, biology and medicine. One of the most challenging goals in the field of cardiovascular tissue engineering is the creation of a heart muscle patch. This review describes the principles, achievements and challenges of achieving this ambitious goal of creating contractile heart muscle. In addition, the new strategy of in situ and injectable tissue engineering for myocardial repair and regeneration is presented.
Collapse
Affiliation(s)
- Jonathan Leor
- Sheba-Medical Center, Neufeld Cardiac Research Institute, Tel-Aviv University, Tel-Hashomer 52621, Israel.
| | | | | |
Collapse
|
40
|
Zeltz C, Orgel J, Gullberg D. Molecular composition and function of integrin-based collagen glues-introducing COLINBRIs. Biochim Biophys Acta Gen Subj 2013; 1840:2533-48. [PMID: 24361615 DOI: 10.1016/j.bbagen.2013.12.022] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/13/2013] [Accepted: 12/14/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Despite detailed knowledge about the structure and signaling properties of individual collagen receptors, much remains to be learned about how these receptors participate in linking cells to fibrillar collagen matrices in tissues. In addition to collagen-binding integrins, a group of proteins with affinity both for fibrillar collagens and integrins link these two protein families together. We have introduced the name COLINBRI (COLlagen INtegrin BRIdging) for this set of molecules. Whereas collagens are the major building blocks in tissues and defects in these structural proteins have severe consequences for tissue integrity, the mild phenotypes of the integrin type of collagen receptors have raised questions about their importance in tissue biology and pathology. SCOPE OF REVIEW We will discuss the two types of cell linkages to fibrillar collagen (direct- versus indirect COLINBRI-mediated) and discuss how the parallel existence of direct and indirect linkages to collagens may ensure tissue integrity. MAJOR CONCLUSIONS The observed mild phenotypes of mice deficient in collagen-binding integrins and the relatively restricted availability of integrin-binding sequences in mature fibrillar collagen matrices support the existence of indirect collagen-binding mechanisms in parallel with direct collagen binding in vivo. GENERAL SIGNIFICANCE A continued focus on understanding the molecular details of cell adhesion mechanisms to collagens will be important and will benefit our understanding of diseases like tissue- and tumor fibrosis where collagen dynamics are disturbed. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway
| | - Joseph Orgel
- Departments of Biology, Physics and Biomedical Engineering, Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, 3440 S. Dearborn Ave, Chicago, IL 60616, USA
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway.
| |
Collapse
|
41
|
Humphries M. Martin Humphries: Attached to adhesion. Interview by Caitlin Sedwick. J Cell Biol 2013; 200:554-5. [PMID: 23460674 PMCID: PMC3587834 DOI: 10.1083/jcb.2005pi] [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] [Indexed: 11/23/2022] Open
Abstract
Humphries studies how interactions with the extracellular matrix influence cell phenotype.
Collapse
|
42
|
Rahmany MB, Van Dyke M. Biomimetic approaches to modulate cellular adhesion in biomaterials: A review. Acta Biomater 2013. [PMID: 23178862 DOI: 10.1016/j.actbio.2012.11.019] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Natural extracellular matrix (ECM) proteins possess critical biological characteristics that provide a platform for cellular adhesion and activation of highly regulated signaling pathways. However, ECM-based biomaterials can have several limitations, including poor mechanical properties and risk of immunogenicity. Synthetic biomaterials alleviate the risks associated with natural biomaterials but often lack the robust biological activity necessary to direct cell function beyond initial adhesion. A thorough understanding of receptor-mediated cellular adhesion to the ECM and subsequent signaling activation has facilitated development of techniques that functionalize inert biomaterials to provide a biologically active surface. Here we review a range of approaches used to modify biomaterial surfaces for optimal receptor-mediated cell interactions, as well as provide insights into specific mechanisms of downstream signaling activation. In addition to a brief overview of integrin receptor-mediated cell function, so-called "biomimetic" techniques reviewed here include (i) surface modification of biomaterials with bioadhesive ECM macromolecules or specific binding motifs, (ii) nanoscale patterning of the materials and (iii) the use of "natural-like" biomaterials.
Collapse
|
43
|
Goswami S. Importance of integrin receptors in the field of pharmaceutical & medical science. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/abc.2013.32028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
44
|
Rice JJ, Martino MM, De Laporte L, Tortelli F, Briquez PS, Hubbell JA. Engineering the regenerative microenvironment with biomaterials. Adv Healthc Mater 2013. [PMID: 23184739 DOI: 10.1002/adhm.201200197] [Citation(s) in RCA: 278] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Modern synthetic biomaterials are being designed to integrate bioactive ligands within hydrogel scaffolds for cells to respond and assimilate within the matrix. These advanced biomaterials are only beginning to be used to simulate the complex spatio-temporal control of the natural healing microenvironment. With increasing understanding of the role of growth factors and cytokines and their interactions with components of the extracellular matrix, novel biomaterials are being developed that more closely mimic the natural healing environments of tissues, resulting in increased efficacy in applications of tissue repair and regeneration. Herein, the important aspects of the healing microenvironment, and how these features can be incorporated within innovative hydrogel scaffolds, are presented.
Collapse
Affiliation(s)
- Jeffrey J Rice
- Institute for Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
45
|
Osteodifferentiation of human preadipocytes induced by strontium released from hydrogels. Int J Biomater 2012; 2012:865291. [PMID: 22927856 PMCID: PMC3423935 DOI: 10.1155/2012/865291] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 06/21/2012] [Indexed: 02/07/2023] Open
Abstract
In recent years, there has been an increasing interest in interactive application principles of biology and engineering for the development of valid biological systems for tissue regeneration, such as for the treatment of bone fractures or skeletal defects. The application of stem cells together with biomaterials releasing bioactive factors promotes the formation of bone tissue by inducing proliferation and/or cell differentiation. In this study, we used a clonal cell line from human adipose tissue-derived mesenchymal stem cells (hADSCs or preadipocytes), named PA2-E12, to evaluate the effects of strontium (Sr2+) released in the culture medium from an amidated carboxymethylcellulose (CMCA) hydrogel enriched with different Sr2+ concentrations on osteodifferentiation. The osteoinductive effect was evaluated through both the expression of alkaline phophatase (ALP) activity and the hydroxyapatite (HA) production during 42 days of induction. Present data have shown that Sr2+ released from CMCA promotes the osteodifferentiation induced by an osteogenic medium as shown by the increase of ALP activity at 7 and 14 days and of HA production at 14 days. In conclusion, the use of biomaterials able to release in situ osteoinductive agents, like Sr2+, could represent a new strategy for future applications in bone tissue engineering.
Collapse
|
46
|
Dong CL, Li SY, Wang Y, Dong Y, Tang JZ, Chen JC, Chen GQ. The cytocompatability of polyhydroxyalkanoates coated with a fusion protein of PHA repressor protein (PhaR) and Lys-Gln-Ala-Gly-Asp-Val (KQAGDV) polypeptide. Biomaterials 2012; 33:2593-9. [DOI: 10.1016/j.biomaterials.2011.12.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2011] [Accepted: 12/09/2011] [Indexed: 12/20/2022]
|
47
|
Colombatti A, Spessotto P, Doliana R, Mongiat M, Bressan GM, Esposito G. The EMILIN/Multimerin family. Front Immunol 2012; 2:93. [PMID: 22566882 PMCID: PMC3342094 DOI: 10.3389/fimmu.2011.00093] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 12/21/2011] [Indexed: 01/12/2023] Open
Abstract
Elastin microfibrillar interface proteins (EMILINs) and Multimerins (EMILIN1, EMILIN2, Multimerin1, and Multimerin2) constitute a four member family that in addition to the shared C-terminus gC1q domain typical of the gC1q/TNF superfamily members contain a N-terminus unique cysteine-rich EMI domain. These glycoproteins are homotrimeric and assemble into high molecular weight multimers. They are predominantly expressed in the extracellular matrix and contribute to several cellular functions in part associated with the gC1q domain and in part not yet assigned nor linked to other specific regions of the sequence. Among the latter is the control of arterial blood pressure, the inhibition of Bacillus anthracis cell cytotoxicity, the promotion of cell death, the proangiogenic function, and a role in platelet hemostasis. The focus of this review is to highlight the multiplicity of functions and domains of the EMILIN/Multimerin family with a particular emphasis on the regulatory role played by the ligand-receptor interactions of the gC1q domain. EMILIN1 is the most extensively studied member both from the structural and functional point of view. The structure of the gC1q of EMILIN1 solved by NMR highlights unique characteristics compared to other gC1q domains: it shows a marked decrease of the contact surface of the trimeric assembly and while conserving the jelly-roll topology with two β-sheets of antiparallel strands it presents a nine-stranded β-sandwich fold instead of the usual 10-stranded fold. This is likely due to the insertion of nine residues that disrupt the ordered strand organization and forma a highly dynamic protruding loop. In this loop the residue E933 is the site of interaction between gC1q and the α4β1 and α9β1 integrins, and contrary to integrin occupancy that usually upregulates cell growth, when gC1q is ligated by the integrin the cells reduce their proliferative activity.
Collapse
Affiliation(s)
- Alfonso Colombatti
- Experimental Oncology 2, Centro di Riferimento Oncologico, Istituto di Ricerca e Cura a Carattere Scientifico Aviano, Italy.
| | | | | | | | | | | |
Collapse
|
48
|
Takasu A, Kondo S, Ito A, Furukawa Y, Higuchi M, Kinoshita T, Kwon I. Artificial extracellular matrix proteins containing phenylalanine analogues biosynthesized in bacteria using T7 expression system and the PEGylation. Biomacromolecules 2011; 12:3444-52. [PMID: 21823658 DOI: 10.1021/bm200522d] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
In vivo incorporation of phenylalanine (Phe) analogues into an artificial extracellular matrix protein (aECM-CS5-ELF) was accomplished using a bacterial expression host that harbors the mutant phenylalanyl-tRNA synthetase (PheRS) with an enlarged binding pocket. Although the Ala294Gly/Thr251Gly mutant PheRS (PheRS**) under the control of T5 promoter allows incorporation of some Phe analogues into a protein, the T5 system is not suitable for material science studies because the amount of materials produced is not sufficient due to the moderate strength of the T5 promoter. This limitation can be overcome by using a pair of T7 promoter and T7 RNA polymerase instead. In the T7 expression system, it is difficult, however, to achieve a high incorporation level of Phe analogues, due to competition of Phe analogues for incorporation with the residual Phe that is required for synthesis of active T7 RNA polymerase. In this study, we prepared the PheRS** under T7 promoter and optimized culture condition to improve both the incorporation level of recombinant aECM protein and the incorporation level of Phe analogues. Incorporation and expression levels tend to increase in the case of p-azidophenylalanine, p-iodophenylalanine, and p-acetylphenylalanine. We evaluated the lower critical transition temperature, which is dependent on the incorporation ratio and the turbidity decreased when the incorporation level increased. Circular dichromism measurement indicated that this tendency is based on conformational change from random coil to β-turn structure. We demonstrated that polyethylene glycol (PEG) can be conjugated at reaction site of Phe analogues incorporated. We also demonstrated that the increased hydrophilicity of elastin-like sequences in the aECM-CS5-ELF made by PEG conjugation could suppress nonspecific adhesion of human umbilical vein endothelial cells (HUVEC).
Collapse
Affiliation(s)
- Akinori Takasu
- Department of Frontier Materials, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya 466-8555, Japan.
| | | | | | | | | | | | | |
Collapse
|
49
|
Synergistic enhancement of human bone marrow stromal cell proliferation and osteogenic differentiation on BMP-2-derived and RGD peptide concentration gradients. Acta Biomater 2011; 7:2091-100. [PMID: 21272672 DOI: 10.1016/j.actbio.2011.01.019] [Citation(s) in RCA: 102] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2010] [Revised: 01/05/2011] [Accepted: 01/12/2011] [Indexed: 01/10/2023]
Abstract
Rational design of bioactive tissue engineered scaffolds for directing bone regeneration in vivo requires a comprehensive understanding of cell interactions with the immobilized bioactive molecules. In the current study, substrates possessing gradient concentrations of immobilized peptides were used to measure the concentration-dependent proliferation and osteogenic differentiation of human bone marrow stromal cells. Two bioactive peptides, one derived from extracellular matrix protein (ECM), GRGDS, and one from bone morphogenic protein-2 (BMP-2), KIPKASSVPTELSAISTLYL, were found to synergistically enhance cell proliferation, up-regulate osteogenic mRNA markers bone sialoprotein (BSP) and Runt-related transcription factor 2, and produce mineralization at densities greater than 130 pmol cm(-2) (65 pmol cm(-2) for each peptide). In addition, COOH-terminated self-assembled monolayers alone led to up-regulated BSP mRNA levels at densities above 200 pmol cm(-2) and increased cell proliferation from day 3 to day 14. Taking further advantage of both the synergistic potentials and the concentration-dependent activities of ECM and growth-factor-derived peptides on proliferative activity and osteogenic differentiation, without the need for additional osteogenic supplements, will enable the successful incorporation of the bioactive species into biorelevant tissue engineering scaffolds.
Collapse
|
50
|
Schvartzman M, Palma M, Sable J, Abramson J, Hu X, Sheetz MP, Wind SJ. Nanolithographic control of the spatial organization of cellular adhesion receptors at the single-molecule level. NANO LETTERS 2011; 11:1306-12. [PMID: 21319842 PMCID: PMC3061283 DOI: 10.1021/nl104378f] [Citation(s) in RCA: 192] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
The ability to control the placement of individual molecules promises to enable a wide range of applications and is a key challenge in nanoscience and nanotechnology. Many biological interactions, in particular, are sensitive to the precise geometric arrangement of proteins. We have developed a technique which combines molecular-scale nanolithography with site-selective biochemistry to create biomimetic arrays of individual protein binding sites. The binding sites can be arranged in heterogeneous patterns of virtually any possible geometry with a nearly unlimited number of degrees of freedom. We have used these arrays to explore how the geometric organization of the extracellular matrix (ECM) binding ligand RGD (Arg-Gly-Asp) affects cell adhesion and spreading. Systematic variation of spacing, density, and cluster size of individual integrin binding sites was used to elicit different cell behavior. Cell spreading assays on arrays of different geometric arrangements revealed a dramatic increase in spreading efficiency when at least four liganded sites were spaced within 60 nm or less, with no dependence on global density. This points to the existence of a minimal matrix adhesion unit for fibronectin defined in space and stoichiometry. Developing an understanding of the ECM geometries that activate specific cellular functional complexes is a critical step toward controlling cell behavior. Potential practical applications range from new therapeutic treatments to the rational design of tissue scaffolds that can optimize healing without scarring. More broadly, spatial control at the single-molecule level can elucidate factors controlling individual molecular interactions and can enable synthesis of new systems based on molecular-scale architectures.
Collapse
Affiliation(s)
- Mark Schvartzman
- Department of Chemical Engineering, Columbia University, 500 West 120 St., New York, NY 10027
- Nanomedicine Center for Mechanobiology – Directing the Immune Response, Columbia University, New York, NY 10027
| | - Matteo Palma
- Department of Applied Physics and Applied Mathematics, Columbia University, 500 West 120 St., New York, NY 10027
- Department of Mechanical Engineering, Columbia University, 500 West 120 St., New York, NY 10027
- Nanomedicine Center for Mechanobiology – Directing the Immune Response, Columbia University, New York, NY 10027
| | - Julia Sable
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Ave., New York, NY 10027
- Nanomedicine Center for Mechanobiology – Directing the Immune Response, Columbia University, New York, NY 10027
| | - Justin Abramson
- Department of Mechanical Engineering, Columbia University, 500 West 120 St., New York, NY 10027
- Nanomedicine Center for Mechanobiology – Directing the Immune Response, Columbia University, New York, NY 10027
| | - Xian Hu
- Department of Biological Sciences National University of Singapore
| | - Michael P. Sheetz
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Ave., New York, NY 10027
- Nanomedicine Center for Mechanobiology – Directing the Immune Response, Columbia University, New York, NY 10027
| | - Shalom J. Wind
- Department of Applied Physics and Applied Mathematics, Columbia University, 500 West 120 St., New York, NY 10027
- Nanomedicine Center for Mechanobiology – Directing the Immune Response, Columbia University, New York, NY 10027
| |
Collapse
|