1
|
Castoldi G, Mauri M, D’Aliberti D, Spinelli S, Testa L, Gaverina F, Rubinacci A, Villa I, Bellelli G, Zerbini G, Piazza R, Zatti G. Transcriptomic Profile of Human Osteoblast-like Cells Grown on Trabecular Titanium. Int J Mol Sci 2025; 26:3598. [PMID: 40332083 PMCID: PMC12026869 DOI: 10.3390/ijms26083598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Trabecular titanium implants are widely used in orthopedic surgery and are known to promote osseointegration. In this study, we investigated whether primary human osteoblast-like cells grown inside a 3D trabecular titanium scaffold undergo changes in migration capacity, transcriptomic profile, and cellular phenotype as compared to the same osteoblasts not grown inside the scaffold. Scratch tests have shown that primary human osteoblast-like cells grown inside the 3D trabecular titanium scaffold promote the migration of cells from the external environment into the scaffold. Next generation sequencing analysis demonstrated that primary human osteoblast-like cells grown inside the 3D trabecular titanium scaffold modified the expression of genes involved in cell cycle and extracellular matrix remodeling, while maintaining a normal expression of the specific osteoblast markers, such as osteocalcin and osterix, as well as a comparable mineralization capacity. These data demonstrate that primary human osteoblast-like cells grown inside the titanium scaffold in a 3D environment acquire specific features favoring osseointegration.
Collapse
Affiliation(s)
- Giovanna Castoldi
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
| | - Mario Mauri
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
| | - Deborah D’Aliberti
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
| | - Silvia Spinelli
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
| | - Leonardo Testa
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
- Clinica Ortopedica, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Federico Gaverina
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
- Clinica Ortopedica, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | | | - Isabella Villa
- Laboratorio di Endocrinologia e Metabolismo Osseo, Istituto di Scienze Endocrine e Metaboliche, IRCCS Ospedale San Raffaele, 20132 Milano, Italy;
| | - Giuseppe Bellelli
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
- Acute Geriatric Unit, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Gianpaolo Zerbini
- Unità Complicanze del Diabete, Diabetes Research Institute, IRCCS Istituto Scientifico San Raffaele, 20132 Milano, Italy;
| | - Rocco Piazza
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
- Divisione di Ematologia, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Giovanni Zatti
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Milano-Bicocca, 20900 Monza, Italy; (M.M.); (D.D.); (S.S.); (L.T.); (F.G.); (G.B.); (R.P.); (G.Z.)
- Clinica Ortopedica, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| |
Collapse
|
2
|
Park YS, Choi Y, Lee JS. Focal adhesion dynamics-mediated cell migration and proliferation on silica bead arrays. Biomater Sci 2025; 13:1849-1857. [PMID: 40012335 DOI: 10.1039/d4bm01659a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Interactions between cells and the extracellular matrix (ECM) alter cellular behaviors, including adhesion, migration, proliferation, and differentiation via focal adhesions that link the ECM to the actin cytoskeleton as an intracellular signaling pathway. Although nanomaterials with various mechanical, geometrical, and topographical features have been used to provide a variety of cell-ECM interactions, it remains unclear how their nanostructured surfaces affect cellular behavior. In this study, we investigated focal adhesion dynamics during the migration and proliferation of HeLa cells on silica bead (SB) arrays with various nanotopographies. Cell adhesion was altered according to the surface curvature and pinhole size of the SB arrays, and cell morphology was determined by the ratio of the adhesive and non-adhesive areas of cells on the SB arrays. In turn, this triggered different focal adhesion dynamics in cells. In addition, we demonstrated the rapid migration and high proliferation characteristics of rounded cells with weak adhesion based on confocal microscopy analysis and migration trajectory on SB arrays, indicating focal adhesion dynamics-mediated cell migration and proliferation on nanostructured surfaces.
Collapse
Affiliation(s)
- Yi-Seul Park
- Department of Chemistry, Sookmyung Women's University, Seoul 04310, Korea.
| | - Yerin Choi
- Department of Chemistry and Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, South Korea.
| | - Jin Seok Lee
- Department of Chemistry and Research Institute for Convergence of Basic Sciences, Hanyang University, Seoul 04763, South Korea.
| |
Collapse
|
3
|
Latte Bovio C, Campione P, Wu H, Li Q, De La Fuente Durán A, Salleo A, Fabiano S, Messina GML, Santoro F. Evaluation of the Biocompatibility of Poly(benzimidazobenzophenanthroline)(BBL) Polymer Films with Living Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2404451. [PMID: 39711257 PMCID: PMC11798344 DOI: 10.1002/smll.202404451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 10/13/2024] [Indexed: 12/24/2024]
Abstract
The integration of organic electronic materials with biological systems to monitor, interface with, and regulate physiological processes is a key area in the field of bioelectronics. Central to this advancement is the development of cell-chip coupling, where materials engineering plays a critical role in enhancing biointerfacing capabilities. Conductive polymers have proven particularly useful in cell interfacing applications due to their favorable biophysical and chemical properties. However, n-type conductive polymers remain underexplored, primarily due to their limited long-term stability. In this study, it is demonstrated that the conductive polymer poly(benzimidazobenzophenanthroline) (BBL), commonly used in organic electronic devices, can effectively support neuronal cell viability and spreading, both as a bare cell culture material and when coated with exracellular matrix proteins. This work provides a preliminary validation of BBL's potential for future integration into bioelectronic devices and in biointerfacing.
Collapse
Affiliation(s)
- Claudia Latte Bovio
- Tissue ElectronicsIstituto Italiano di TecnologiaNaples80125Italy
- Dipartimento di ChimicaMateriali e Produzione IndustrialeUniversità di Napoli Federico IINaples80125Italy
| | - Paola Campione
- Laboratory for Molecular Surface and Nanotechnology (LAMSUN)Department of Chemical SciencesUniversity of Catania. and CSGIViale A. Doria, 6Catania95125Italy
| | - Han‐Yan Wu
- Laboratory of Organic Electronics Department of Science and TechnologyLinköping UniversityNorrköpingSE‐60174Sweden
| | - Qifan Li
- Laboratory of Organic Electronics Department of Science and TechnologyLinköping UniversityNorrköpingSE‐60174Sweden
| | | | - Alberto Salleo
- Department of Materials Science and EngineeringStanford UniversityStanfordCA94305USA
| | - Simone Fabiano
- Laboratory of Organic Electronics Department of Science and TechnologyLinköping UniversityNorrköpingSE‐60174Sweden
| | - Grazia Maria Lucia Messina
- Laboratory for Molecular Surface and Nanotechnology (LAMSUN)Department of Chemical SciencesUniversity of Catania. and CSGIViale A. Doria, 6Catania95125Italy
| | - Francesca Santoro
- Tissue ElectronicsIstituto Italiano di TecnologiaNaples80125Italy
- Faculty of Electrical Engineering and ITRWTH52074AachenGermany
- Institute for Biological Information Processing‐BioelectronicsForschungszentrum Juelich52428JulichGermany
| |
Collapse
|
4
|
Zeng L, Armstrong S, Zhu Y, Gregory SD, Huang A, Dyson JM. 3D-printed surfaces of titanium implant: the fibroblasts response. BIOMATERIALS ADVANCES 2025; 166:214006. [PMID: 39265449 DOI: 10.1016/j.bioadv.2024.214006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/14/2024]
Abstract
Ti-6Al-4V (wt%) is the most widely used titanium alloy and its additive manufactured (or 3D printed) parts with near net-shape have provided great advantages for biomedical applications. While the impact of surface roughness on the biocompatibility of 3D-printed Ti-6Al-4V part is recognized, further exploration is needed to fully understand this complex relationship. Hence, this study presents a comprehensive evaluation of as-printed Ti-6Al-4V structures, both with and without surface texturing, with particular focus on the fibroblast response. Alongside a flat surface, or as-printed surface, two different types of surface textures: diamond texture and diamond crystal texture, were meticulously designed and printed through laser powder bed fusion (LPBF). The viability, cell adhesion, and morphology of human and murine fibroblasts seeded on the surface patterns was investigated, as well as the distribution of extracellular matrix (ECM) proteins (collagen I, fibronectin). The results demonstrated that the as-fabricated surface morphologies did not impact fibroblast viability, however, a reduced density of human fibroblasts was observed on the diamond texture surface, likely owing to the upright strut structure preventing cell adhesion. Interestingly, spreading of the human, but not murine, fibroblasts was limited by the remaining partially-sintered powders. The relative intensity of ECM protein signals was unaffected, however, ECM protein distribution across the surfaces was also altered. Thus, the as-printed substrates, particularly with diamond crystal struts, present a promising avenue for the cost-effective and efficient fabrication of Ti-6Al-4V components for medical applications in the future.
Collapse
Affiliation(s)
- Lingxiao Zeng
- Monash Centre for Additive Manufacturing, Monash University, Notting Hill, VIC 3168, Australia; Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Sophie Armstrong
- CardioRespiratory Engineering and Technology Laboratory (CREATElab), Department of Mechanical and Aerospace Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Yuman Zhu
- Monash Centre for Additive Manufacturing, Monash University, Notting Hill, VIC 3168, Australia; Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia.
| | - Shaun D Gregory
- CardioRespiratory Engineering and Technology Laboratory (CREATElab), Department of Mechanical and Aerospace Engineering, Monash University, Clayton, VIC 3800, Australia; Centre for Biomedical Technologies and School of Mechanical, Medical, and Process Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Aijun Huang
- Monash Centre for Additive Manufacturing, Monash University, Notting Hill, VIC 3168, Australia; Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| | - Jennifer M Dyson
- Department of Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia; Cancer Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia.
| |
Collapse
|
5
|
Pulat G, Gökmen O, Özcan Ş, Karaman O. Collagen binding and mimetic peptide-functionalized self-assembled peptide hydrogel enhance chondrogenic differentiation of human mesenchymal stem cells. J Biomed Mater Res A 2025; 113:e37786. [PMID: 39237470 DOI: 10.1002/jbm.a.37786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/07/2024]
Abstract
The avascular structure and low cell migration to the damaged area due to the low number of cells do not allow spontaneous repair of the articular cartilage tissue. Therefore, functional scaffolds obtained from biomaterials are used for the regeneration of cartilage tissue. Here, we functionalized one of the self-assembling peptide (SAP) scaffolds KLD (KLDLKLDLKLDL) with short bioactive motifs, which are the α1 chain of type II collagen binding peptide WYRGRL (C1) and the triple helical collagen mimetic peptide GFOGER (C2) by direct coupling. Our goal was to develop injectable functional SAP hydrogels with proper mechanical characteristics that would improve chondrogenesis. Scanning electron microscopy (SEM) was used to observe the integration of peptide scaffold structure at the molecular level. To assure the stability of SAPs, the rheological characteristics and degradation profile of SAP hydrogels were assessed. The biochemical study of the DNA, glycosaminoglycan (GAG), and collagen content revealed that the developed bioactive SAP hydrogels greatly increased hMSCs proliferation compared with KLD scaffolds. Moreover, the addition of bioactive peptides to KLD dramatically increased the expression levels of important chondrogenic markers such as aggrecan, SOX-9, and collagen Type II as evaluated by real-time polymerase chain reaction (PCR). We showed that hMSC proliferation and chondrogenic differentiation were encouraged by the developed SAP scaffolds. Although the chondrogenic potentials of WYRGRL and GFOGER were previously investigated, no study compares the effect of the two peptides integrated into 3-D SAP hydrogels in chondrogenic differentiation. Our findings imply that these specifically created bioactive peptide scaffolds might help enhance cartilage tissue regeneration.
Collapse
Affiliation(s)
- Günnur Pulat
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey
| | - Oğuzhan Gökmen
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey
| | - Şerife Özcan
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey
| | - Ozan Karaman
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey
- Bonegraft Biomaterials Co., Ege University Technopolis, İzmir, Turkey
| |
Collapse
|
6
|
Xu L, Ren W, Long Y, Yang B, Chen L, Chen W, Chen S, Cao Y, Wu D, Qu J, Li H, Yu Y, Zhang A, Wang S, Wang H, Chen T, Fan G, Li Q, Chen Z. Antisenescence Expansion of Mesenchymal Stem Cells Using Piezoelectric β-Poly(vinylidene fluoride) Film-Based Culture. ACS APPLIED MATERIALS & INTERFACES 2024; 16:63207-63224. [PMID: 39503875 DOI: 10.1021/acsami.4c12725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Regenerative therapies based on mesenchymal stem cells (MSCs) show promise in treating a wide range of disorders. However, the replicative senescence of MSCs during in vitro expansion poses a challenge to obtaining a substantial quantity of high-quality MSCs. In this investigation, a piezoelectric β-poly(vinylidene fluoride) film-based culture plate (β-CP) was developed with an antisenescence effect on cultured human umbilical cord-derived MSCs. Compared to traditional tissue culture plates (TCPs) and α-poly(vinylidene fluoride) film-based culture plates, the senescence markers of p21, p53, interleukin-6 and insulin-like growth factor-binding protein-7, stemness markers of OCT4 and NANOG, and telomere length of MSCs cultured on β-CPs were significantly improved. Additionally, MSCs at passage 18 cultured on β-CPs showed significantly better multipotency and pro-angiogenic capacities in vitro, and higher wound healing abilities in a mouse model. Mechanistically, β-CPs rejuvenated senescent MSCs by improving mitochondrial functions and mitigating oxidative and glycoxidative stresses. Overall, this study presents β-CPs as a promising approach for efficient and straightforward antisenescence expansion of MSCs while preserving their stemness, thereby holding great potential for large-scale production of MSCs for clinical application in cell therapies.
Collapse
Affiliation(s)
- Liuyue Xu
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenxiang Ren
- Center for Hematology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yaoying Long
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bianlei Yang
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Chen
- Department of Hematology, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Wenlan Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Siyi Chen
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yulin Cao
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Di Wu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiao Qu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - He Li
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yali Yu
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anyuan Zhang
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shan Wang
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongxiang Wang
- Department of Hematology, Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Ting Chen
- Hubei Engineering Research Center for Application of Extracellular Vesicles, Hubei University of Science and Technology, Xianning 437100, China
| | - Guifen Fan
- School of Optical and Electronic Information, Key Lab of Functional Materials for Electronic Information(B), MOE, Huazhong University of Science and Technology, Wuhan 430074, China
- Wenzhou Advanced Manufacturing Institute, Huazhong University of Science and Technology, Wenzhou 325035, China
| | - Qiubai Li
- Department of Rheumatology and Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Engineering Research Center for Application of Extracellular Vesicles, Hubei University of Science and Technology, Xianning 437100, China
| | - Zhichao Chen
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
7
|
Saporito S, Panzetta V, Netti PA. Time and space modulation of substrate curvature to regulate cell mechanical identity. Acta Biomater 2024; 186:300-315. [PMID: 39127326 DOI: 10.1016/j.actbio.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/08/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Recently, a variety of microenvironmental biophysical stimuli have been proved to play a crucial role in regulating cell functions. Among them, morpho-physical cues, like curvature, are emerging as key regulators of cellular behavior. Changes in substrate curvature have been shown to impact the arrangement of Focal Adhesions (FAs), influencing the direction and intensity of cytoskeleton generated forces and resulting in an overall alteration of cell mechanical identity. In their native environment, cells encounter varying degrees of substrate curvature, and in specific organs, they are exposed to dynamic changes of curvature due to periodic tissue deformation. However, the mechanism by which cells perceive substrate curvature remains poorly understood. To this aim, a micro-pneumatic device was designed and implemented. This device enables the controlled application of substrate curvature, both statically and dynamically. Employing a combined experimental and simulative approach, human adipose-derived stem cells were exposed to controlled curvature intensity and frequency. During this exposure, measurements were taken on FAs extension and orientation, cytoskeleton organization and cellular/nuclear alignment. The data clearly indicated a significant influence of the substrate curvature on cell adhesion processes. These findings contribute to a better understanding of the mechanisms through which cells perceive and respond to substrate curvature signals. STATEMENT OF SIGNIFICANCE: This work is our contribution to the comprehension of substrate curvature's function as a crucial regulator of cell adhesion at the scale of focal adhesions and cell mechanical identity. In recent years, a large body of knowledge is continuously growing providing comprehension of the role of various microenvironmental biophysical stimuli in regulating cell functions. Nevertheless, little is known about the role of substrate curvature, in particular, when cells are exposed to this stimulus in a dynamic manner. To address the role of substrate curvature on cellular behavior, a micro-pneumatic device was designed and implemented. This device enables the controlled application of substrate curvature, both statically and dynamically. The experiment data made it abundantly evident that the substrate curvature had a major impact on the mechanisms involved in cell adhesion.
Collapse
Affiliation(s)
- Stefania Saporito
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy
| | - Valeria Panzetta
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy; Interdisciplinary research Center on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy
| | - Paolo Antonio Netti
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Italy; Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano Di Tecnologia, Italy; Interdisciplinary research Center on Biomaterials (CRIB), University of Naples Federico II, Naples, Italy.
| |
Collapse
|
8
|
Kapat K, Gondane P, Kumbhakarn S, Takle S, Sable R. Challenges and Opportunities in Developing Tracheal Substitutes for the Recovery of Long-Segment Defects. Macromol Biosci 2024; 24:e2400054. [PMID: 39008817 DOI: 10.1002/mabi.202400054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/21/2024] [Indexed: 07/17/2024]
Abstract
Tracheal resection and reconstruction procedures are necessary when stenosis, tracheomalacia, tumors, vascular lesions, or tracheal injury cause a tracheal blockage. Replacement with a tracheal substitute is often recommended when the trauma exceeds 50% of the total length of the trachea in adults and 30% in children. Recently, tissue engineering and other advanced techniques have shown promise in fabricating biocompatible tracheal substitutes with physical, morphological, biomechanical, and biological characteristics similar to native trachea. Different polymers and biometals are explored. Even with limited success with tissue-engineered grafts in clinical settings, complete healing of tracheal defects remains a substantial challenge due to low mechanical strength and durability of the graft materials, inadequate re-epithelialization and vascularization, and restenosis. This review has covered a range of reconstructive and regenerative techniques, design criteria, the use of bioprostheses and synthetic grafts for the recovery of tracheal defects, as well as the traditional and cutting-edge methods of their fabrication, surface modification for increased immuno- or biocompatibility, and associated challenges.
Collapse
Affiliation(s)
- Kausik Kapat
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata, West Bengal, 700054, India
| | - Prashil Gondane
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata, West Bengal, 700054, India
| | - Sakshi Kumbhakarn
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata, West Bengal, 700054, India
| | - Shruti Takle
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata, West Bengal, 700054, India
| | - Rahul Sable
- Department of Medical Devices, National Institute of Pharmaceutical Education and Research Kolkata, 168, Maniktala Main Road, Kankurgachi, Kolkata, West Bengal, 700054, India
| |
Collapse
|
9
|
Abbadessa A, Ronca A, Salerno A. Integrating bioprinting, cell therapies and drug delivery towards in vivo regeneration of cartilage, bone and osteochondral tissue. Drug Deliv Transl Res 2024; 14:858-894. [PMID: 37882983 DOI: 10.1007/s13346-023-01437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/27/2023]
Abstract
The biological and biomechanical functions of cartilage, bone and osteochondral tissue are naturally orchestrated by a complex crosstalk between zonally dependent cells and extracellular matrix components. In fact, this crosstalk involves biomechanical signals and the release of biochemical cues that direct cell fate and regulate tissue morphogenesis and remodelling in vivo. Three-dimensional bioprinting introduced a paradigm shift in tissue engineering and regenerative medicine, since it allows to mimic native tissue anisotropy introducing compositional and architectural gradients. Moreover, the growing synergy between bioprinting and drug delivery may enable to replicate cell/extracellular matrix reciprocity and dynamics by the careful control of the spatial and temporal patterning of bioactive cues. Although significant advances have been made in this direction, unmet challenges and open research questions persist. These include, among others, the optimization of scaffold zonality and architectural features; the preservation of the bioactivity of loaded active molecules, as well as their spatio-temporal release; the in vitro scaffold maturation prior to implantation; the pros and cons of each animal model and the graft-defect mismatch; and the in vivo non-invasive monitoring of new tissue formation. This work critically reviews these aspects and reveals the state of the art of using three-dimensional bioprinting, and its synergy with drug delivery technologies, to pattern the distribution of cells and/or active molecules in cartilage, bone and osteochondral engineered tissues. Most notably, this work focuses on approaches, technologies and biomaterials that are currently under in vivo investigations, as these give important insights on scaffold performance at the implantation site and its interaction/integration with surrounding tissues.
Collapse
Affiliation(s)
- Anna Abbadessa
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), IDIS Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Campus Vida, Santiago de Compostela, Spain.
| | - Alfredo Ronca
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125, Naples, Italy.
| | - Aurelio Salerno
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, 80125, Naples, Italy.
| |
Collapse
|
10
|
Alaoui Selsouli Y, Rho HS, Eischen-Loges M, Galván-Chacón VP, Stähli C, Viecelli Y, Döbelin N, Bohner M, Tahmasebi Birgani Z, Habibović P. Optimization of a tunable process for rapid production of calcium phosphate microparticles using a droplet-based microfluidic platform. Front Bioeng Biotechnol 2024; 12:1352184. [PMID: 38600949 PMCID: PMC11004461 DOI: 10.3389/fbioe.2024.1352184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/08/2024] [Indexed: 04/12/2024] Open
Abstract
Calcium phosphate (CaP) biomaterials are amongst the most widely used synthetic bone graft substitutes, owing to their chemical similarities to the mineral part of bone matrix and off-the-shelf availability. However, their ability to regenerate bone in critical-sized bone defects has remained inferior to the gold standard autologous bone. Hence, there is a need for methods that can be employed to efficiently produce CaPs with different properties, enabling the screening and consequent fine-tuning of the properties of CaPs towards effective bone regeneration. To this end, we propose the use of droplet microfluidics for rapid production of a variety of CaP microparticles. Particularly, this study aims to optimize the steps of a droplet microfluidic-based production process, including droplet generation, in-droplet CaP synthesis, purification and sintering, in order to obtain a library of CaP microparticles with fine-tuned properties. The results showed that size-controlled, monodisperse water-in-oil microdroplets containing calcium- and phosphate-rich solutions can be produced using a flow-focusing droplet-generator microfluidic chip. We optimized synthesis protocols based on in-droplet mineralization to obtain a range of CaP microparticles without and with inorganic additives. This was achieved by adjusting synthesis parameters, such as precursor concentration, pH value, and aging time, and applying heat treatment. In addition, our results indicated that the synthesis and fabrication parameters of CaPs in this method can alter the microstructure and the degradation behavior of CaPs. Overall, the results highlight the potential of the droplet microfluidic platform for engineering CaP microparticle biomaterials with fine-tuned properties.
Collapse
Affiliation(s)
- Y. Alaoui Selsouli
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - H. S. Rho
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - M. Eischen-Loges
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - V. P. Galván-Chacón
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - C. Stähli
- RMS Foundation, Bettlach, Switzerland
| | | | | | - M. Bohner
- RMS Foundation, Bettlach, Switzerland
| | - Z. Tahmasebi Birgani
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - P. Habibović
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
11
|
Aliakbarshirazi S, Ghobeira R, Asadian M, Narimisa M, Nikiforov A, De Baere I, Van Paepegem W, De Geyter N, Declercq H, Morent R. Advanced Hollow Cathode Discharge Plasma Treatment of Unique Bilayered Fibrous Nerve Guidance Conduits for Enhanced/Oriented Neurite Outgrowth. Biomacromolecules 2024; 25:1448-1467. [PMID: 38412382 DOI: 10.1021/acs.biomac.3c00976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
Despite all recent progresses in nerve tissue engineering, critical-sized nerve defects are still extremely challenging to repair. Therefore, this study targets the bridging of critical nerve defects and promoting an oriented neuronal outgrowth by engineering innovative nerve guidance conduits (NGCs) synergistically possessing exclusive topographical, chemical, and mechanical cues. To do so, a mechanically adequate mixture of polycaprolactone (PCL) and polylactic-co-glycolic acid (PLGA) was first carefully selected as base material to electrospin nanofibrous NGCs simulating the extracellular matrix. The electrospinning process was performed using a newly designed 2-pole air gap collector that leads to a one-step deposition of seamless NGCs having a bilayered architecture with an inner wall composed of highly aligned fibers and an outer wall consisting of randomly oriented fibers. This architecture is envisaged to afford guidance cues for the extension of long neurites on the underlying inner fiber alignment and to concurrently provide a sufficient nutrient supply through the pores of the outer random fibers. The surface chemistry of the NGCs was then modified making use of a hollow cathode discharge (HCD) plasma reactor purposely designed to allow an effective penetration of the reactive species into the NGCs to eventually treat their inner wall. X-ray photoelectron spectroscopy (XPS) results have indeed revealed a successful O2 plasma modification of the inner wall that exhibited a significantly increased oxygen content (24 → 28%), which led to an enhanced surface wettability. The treatment increased the surface nanoroughness of the fibers forming the NGCs as a result of an etching effect. This effect reduced the ultimate tensile strength of the NGCs while preserving their high flexibility. Finally, pheochromocytoma (PC12) cells were cultured on the NGCs to monitor their ability to extend neurites which is the base of a good nerve regeneration. In addition to remarkably improved cell adhesion and proliferation on the plasma-treated NGCs, an outstanding neural differentiation occurred. In fact, PC12 cells seeded on the treated samples extended numerous long neurites eventually establishing a neural network-like morphology with an overall neurite direction following the alignment of the underlying fibers. Overall, PCL/PLGA NGCs electrospun using the 2-pole air gap collector and O2 plasma-treated using an HCD reactor are promising candidates toward a full repair of critical nerve damage.
Collapse
Affiliation(s)
- Sheida Aliakbarshirazi
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Faculty of Engineering and Architecture, Ghent University, Sint-Pietersnieuwstraat 41 B4, 9000 Ghent, Belgium
| | - Rouba Ghobeira
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Faculty of Engineering and Architecture, Ghent University, Sint-Pietersnieuwstraat 41 B4, 9000 Ghent, Belgium
| | - Mahtab Asadian
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Faculty of Engineering and Architecture, Ghent University, Sint-Pietersnieuwstraat 41 B4, 9000 Ghent, Belgium
| | - Mehrnoush Narimisa
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Faculty of Engineering and Architecture, Ghent University, Sint-Pietersnieuwstraat 41 B4, 9000 Ghent, Belgium
| | - Anton Nikiforov
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Faculty of Engineering and Architecture, Ghent University, Sint-Pietersnieuwstraat 41 B4, 9000 Ghent, Belgium
| | - Ives De Baere
- Mechanics of Materials and Structures (MMS), Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Technologiepark-Zwijnaarde 903, 9052 Zwijnaarde, Belgium
| | - Wim Van Paepegem
- Mechanics of Materials and Structures (MMS), Department of Materials, Textiles and Chemical Engineering, Faculty of Engineering and Architecture, Ghent University, Technologiepark-Zwijnaarde 903, 9052 Zwijnaarde, Belgium
| | - Nathalie De Geyter
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Faculty of Engineering and Architecture, Ghent University, Sint-Pietersnieuwstraat 41 B4, 9000 Ghent, Belgium
| | - Heidi Declercq
- Tissue Engineering Lab, Department of Development and Regeneration, Faculty of Medicine, KU Leuven Campus Kulak, Etienne Sabbelaan 53, 8500 Kortrijk, Belgium
| | - Rino Morent
- Research Unit Plasma Technology (RUPT), Department of Applied Physics, Faculty of Engineering and Architecture, Ghent University, Sint-Pietersnieuwstraat 41 B4, 9000 Ghent, Belgium
| |
Collapse
|
12
|
Aqel S, Al-Thani N, Haider MZ, Abdelhady S, Al Thani AA, Kobeissy F, Shaito AA. Biomaterials in Traumatic Brain Injury: Perspectives and Challenges. BIOLOGY 2023; 13:21. [PMID: 38248452 PMCID: PMC10813103 DOI: 10.3390/biology13010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 01/23/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and long-term impairment globally. TBI has a dynamic pathology, encompassing a variety of metabolic and molecular events that occur in two phases: primary and secondary. A forceful external blow to the brain initiates the primary phase, followed by a secondary phase that involves the release of calcium ions (Ca2+) and the initiation of a cascade of inflammatory processes, including mitochondrial dysfunction, a rise in oxidative stress, activation of glial cells, and damage to the blood-brain barrier (BBB), resulting in paracellular leakage. Currently, there are no FDA-approved drugs for TBI, but existing approaches rely on delivering micro- and macromolecular treatments, which are constrained by the BBB, poor retention, off-target toxicity, and the complex pathology of TBI. Therefore, there is a demand for innovative and alternative therapeutics with effective delivery tactics for the diagnosis and treatment of TBI. Tissue engineering, which includes the use of biomaterials, is one such alternative approach. Biomaterials, such as hydrogels, including self-assembling peptides and electrospun nanofibers, can be used alone or in combination with neuronal stem cells to induce neurite outgrowth, the differentiation of human neural stem cells, and nerve gap bridging in TBI. This review examines the inclusion of biomaterials as potential treatments for TBI, including their types, synthesis, and mechanisms of action. This review also discusses the challenges faced by the use of biomaterials in TBI, including the development of biodegradable, biocompatible, and mechanically flexible biomaterials and, if combined with stem cells, the survival rate of the transplanted stem cells. A better understanding of the mechanisms and drawbacks of these novel therapeutic approaches will help to guide the design of future TBI therapies.
Collapse
Affiliation(s)
- Sarah Aqel
- Medical Research Center, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
| | - Najlaa Al-Thani
- Research and Development Department, Barzan Holdings, Doha P.O. Box 7178, Qatar
| | - Mohammad Z. Haider
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria 21544, Egypt;
| | - Asmaa A. Al Thani
- Biomedical Research Center and Department of Biomedical Sciences, College of Health Science, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310, USA
| | - Abdullah A. Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
13
|
Hussain Z, Ullah I, Liu X, Mehmood S, Wang L, Ma F, Ullah S, Lu Z, Wang Z, Pei R. GelMA-catechol coated FeHAp nanorods functionalized nanofibrous reinforced bio-instructive and mechanically robust composite hydrogel scaffold for bone tissue engineering. BIOMATERIALS ADVANCES 2023; 155:213696. [PMID: 37952462 DOI: 10.1016/j.bioadv.2023.213696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/24/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
Critical bone defects complicate tissue graft-based surgeries, raising healthcare expenditures and underscoring scaffold-based tissue-engineering strategies to support bone reconstruction. Our study highlighted that the phase-compatible combination of inorganic nanorods, nanofibers, and hydrogels is promising for developing biomimetic and cell-instructive scaffolds since the bone matrix is a porous organic/inorganic composite. In brief, methacrylated gelatin (GelMA) was reacted with dopamine to form catechol-modified GeLMA (GelMA-C). The GelMA-C was nanocoated onto an iron-doped hydroxyapatite (FeHAp) nanorod via metal-catechol network coordination. The modified nanorod (FeHAp@GelMA-C) was loaded onto GelMA-based nanofibers. The nanorods loaded pre-fibers were electrospun onto GelMA solution and photochemically crosslinked to fabricate a fiber-reinforced hydrogel. The structural, mechanical, physicochemical, biocompatibility, swelling properties, osteogenic potential, and bone remodelling potential (using rat femoral defect model) of modified nanorods, simple hydrogel, and nanorod-loaded fiber-reinforced hydrogel were studied. The results supported that the interface interaction between GelMA-C/nanorods, nanorods/nanofibers, nanorods/hydrogels, and nanofiber/hydrogels significantly improved the microstructural and mechanical properties of the scaffold. Compared to pristine hydrogel, the nanorod-loaded fiber-reinforced scaffold better supported cellular responses, osteogenic differentiation, matrix mineralization, and accelerated bone regeneration. The nanorod-loaded fiber-reinforced hydrogel proved more biomimetic and cell-instructive for guided bone reconstruction.
Collapse
Affiliation(s)
- Zahid Hussain
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China; CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Sciences, Suzhou 215123, PR China
| | - Ismat Ullah
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Sciences, Suzhou 215123, PR China
| | - Xingzhu Liu
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Sciences, Suzhou 215123, PR China
| | - Shah Mehmood
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China; CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Sciences, Suzhou 215123, PR China
| | - Li Wang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China; CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Sciences, Suzhou 215123, PR China
| | - Fanshu Ma
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Sciences, Suzhou 215123, PR China
| | - Salim Ullah
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China; CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Sciences, Suzhou 215123, PR China
| | - Zhongzhong Lu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China; CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Sciences, Suzhou 215123, PR China
| | - Zixun Wang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China; CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Sciences, Suzhou 215123, PR China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China (USTC), Hefei 230026, PR China; CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics (SINANO), Chinese Academy of Sciences, Suzhou 215123, PR China.
| |
Collapse
|
14
|
Adhikari B, Stager MA, Krebs MD. Cell-instructive biomaterials in tissue engineering and regenerative medicine. J Biomed Mater Res A 2023; 111:660-681. [PMID: 36779265 DOI: 10.1002/jbm.a.37510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/16/2023] [Accepted: 01/29/2023] [Indexed: 02/14/2023]
Abstract
The field of biomaterials aims to improve regenerative outcomes or scientific understanding for a wide range of tissue types and ailments. Biomaterials can be fabricated from natural or synthetic sources and display a plethora of mechanical, electrical, and geometrical properties dependent on their desired application. To date, most biomaterial systems designed for eventual translation to the clinic rely on soluble signaling moieties, such as growth factors, to elicit a specific cellular response. However, these soluble factors are often limited by high cost, convoluted synthesis, low stability, and difficulty in regulation, making the translation of these biomaterials systems to clinical or commercial applications a long and arduous process. In response to this, significant effort has been dedicated to researching cell-directive biomaterials which can signal for specific cell behavior in the absence of soluble factors. Cells of all tissue types have been shown to be innately in tune with their microenvironment, which is a biological phenomenon that can be exploited by researchers to design materials that direct cell behavior based on their intrinsic characteristics. This review will focus on recent developments in biomaterials that direct cell behavior using biomaterial properties such as charge, peptide presentation, and micro- or nano-geometry. These next generation biomaterials could offer significant strides in the development of clinically relevant medical devices which improve our understanding of the cellular microenvironment and enhance patient care in a variety of ailments.
Collapse
Affiliation(s)
- Bikram Adhikari
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Michael A Stager
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Melissa D Krebs
- Quantitative Biosciences and Engineering, Colorado School of Mines, Golden, Colorado, USA
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| |
Collapse
|
15
|
Adhikari J, Roy A, Chanda A, D A G, Thomas S, Ghosh M, Kim J, Saha P. Effects of surface patterning and topography on the cellular functions of tissue engineered scaffolds with special reference to 3D bioprinting. Biomater Sci 2023; 11:1236-1269. [PMID: 36644788 DOI: 10.1039/d2bm01499h] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The extracellular matrix (ECM) of the tissue organ exhibits a topography from the nano to micrometer range, and the design of scaffolds has been inspired by the host environment. Modern bioprinting aims to replicate the host tissue environment to mimic the native physiological functions. A detailed discussion on the topographical features controlling cell attachment, proliferation, migration, differentiation, and the effect of geometrical design on the wettability and mechanical properties of the scaffold are presented in this review. Moreover, geometrical pattern-mediated stiffness and pore arrangement variations for guiding cell functions have also been discussed. This review also covers the application of designed patterns, gradients, or topographic modulation on 3D bioprinted structures in fabricating the anisotropic features. Finally, this review accounts for the tissue-specific requirements that can be adopted for topography-motivated enhancement of cellular functions during the fabrication process with a special thrust on bioprinting.
Collapse
Affiliation(s)
- Jaideep Adhikari
- School of Advanced Materials, Green Energy and Sensor Systems, Indian Institute of Engineering Science and Technology, Shibpur, Howrah 711103, India
| | - Avinava Roy
- Department of Metallurgy and Materials Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah 711103, India
| | - Amit Chanda
- Department of Mechanical Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Gouripriya D A
- Centre for Interdisciplinary Sciences, JIS Institute of Advanced Studies and Research (JISIASR) Kolkata, JIS University, GP Block, Salt Lake, Sector-5, West Bengal 700091, India.
| | - Sabu Thomas
- School of Chemical Sciences, MG University, Kottayam 686560, Kerala, India
| | - Manojit Ghosh
- Department of Metallurgy and Materials Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah 711103, India
| | - Jinku Kim
- Department of Bio and Chemical Engineering, Hongik University, Sejong, 30016, South Korea.
| | - Prosenjit Saha
- Centre for Interdisciplinary Sciences, JIS Institute of Advanced Studies and Research (JISIASR) Kolkata, JIS University, GP Block, Salt Lake, Sector-5, West Bengal 700091, India.
| |
Collapse
|
16
|
Belousov A, Patlay A, Silant’ev V, Kovalev VV, Kumeiko V. Preparation of Hydrogels Based on Modified Pectins by Tuning Their Properties for Anti-Glioma Therapy. Int J Mol Sci 2022; 24:ijms24010630. [PMID: 36614073 PMCID: PMC9820215 DOI: 10.3390/ijms24010630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
The extracellular matrix (ECM) of the central nervous system (CNS), characterized by low stiffness and predominance of carbohydrates on protein components, mediates limited cell proliferation and migration. Pectins are polysaccharides derived from plants and could be very promising for a tunable hydrogel design that mimics the neural ECM. Aiming to regulate gel structure and viscoelastic properties, we elaborated 10 variants of pectin-based hydrogels via tuning the concentration of the polymer and the number of free carboxyl groups expressed in the degree of esterification (DE). Viscoelastic properties of hydrogels varied in the range of 3 to 900 Pa for G' and were chosen as the first criteria for the selection of variants suitable for CNS remodeling. For extended reciprocal characterization, two pairs of hydrogels were taken to test pectins with opposite DEs close to 0% and 50%, respectively, but with a similar rheology exceeding 100 Pa (G'), which was achieved by adjusting the concentration of pectin. Hydrogel swelling properties and in vitro stability, together with structure characterization using SEM and FTIR spectroscopy, displayed some differences that may sense for biomedical application. Bioassays on C6 and U87MG glioblastoma cultures testified the potential prospects of the anti-glioma activity of hydrogels developed by decreasing cell proliferation and modulating migration but supporting the high viability of neural cells.
Collapse
Affiliation(s)
- Andrei Belousov
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Aleksandra Patlay
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Vladimir Silant’ev
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok 690922, Russia
- Laboratory of Electrochemical Processes, Institute of Chemistry, FEB RAS, Vladivostok 690022, Russia
| | - Valeri V. Kovalev
- A.V. Zhirmunsky National Scientific Center of Marine Biology, FEB RAS, Vladivostok 690041, Russia
| | - Vadim Kumeiko
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, Vladivostok 690922, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, FEB RAS, Vladivostok 690041, Russia
- Correspondence:
| |
Collapse
|
17
|
Modaresifar K, Ganjian M, Díaz-Payno PJ, Klimopoulou M, Koedam M, van der Eerden BC, Fratila-Apachitei LE, Zadpoor AA. Mechanotransduction in high aspect ratio nanostructured meta-biomaterials: The role of cell adhesion, contractility, and transcriptional factors. Mater Today Bio 2022; 16:100448. [PMID: 36238966 PMCID: PMC9552121 DOI: 10.1016/j.mtbio.2022.100448] [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: 01/23/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022] Open
Abstract
Black Ti (bTi) surfaces comprising high aspect ratio nanopillars exhibit a rare combination of bactericidal and osteogenic properties, framing them as cell-instructive meta-biomaterials. Despite the existing data indicating that bTi surfaces induce osteogenic differentiation in cells, the mechanisms by which this response is regulated are not fully understood. Here, we hypothesized that high aspect ratio bTi nanopillars regulate cell adhesion, contractility, and nuclear translocation of transcriptional factors, thereby inducing an osteogenic response in the cells. Upon the observation of significant changes in the morphological characteristics, nuclear localization of Yes-associated protein (YAP), and Runt-related transcription factor 2 (Runx2) expression in the human bone marrow-derived mesenchymal stem cells (hMSCs), we inhibited focal adhesion kinase (FAK), Rho-associated protein kinase (ROCK), and YAP in separate experiments to elucidate their effects on the subsequent expression of Runx2. Our findings indicated that the increased expression of Runx2 in the cells residing on the bTi nanopillars compared to the flat Ti is highly dependent on the activity of FAK and ROCK. A mechanotransduction pathway is then postulated in which the FAK-dependent adhesion of cells to the extreme topography of the surface is in close relation with ROCK to increase the endogenous forces within the cells, eventually determining the cell shape and area. The nuclear translocation of YAP may also enhance in response to the changes in cell shape and area, resulting in the translation of mechanical stimuli to biochemical factors such as Runx2.
Collapse
Affiliation(s)
- Khashayar Modaresifar
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628CD, Delft, the Netherlands
| | - Mahya Ganjian
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628CD, Delft, the Netherlands
| | - Pedro J. Díaz-Payno
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628CD, Delft, the Netherlands
- Department of Orthopedics and Sports Medicine, Erasmus MC University Medical Center, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Maria Klimopoulou
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628CD, Delft, the Netherlands
| | - Marijke Koedam
- Department of Internal Medicine, Erasmus MC University Medical Center, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Bram C.J. van der Eerden
- Department of Internal Medicine, Erasmus MC University Medical Center, Doctor Molewaterplein 40, 3015GD, Rotterdam, the Netherlands
| | - Lidy E. Fratila-Apachitei
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628CD, Delft, the Netherlands
| | - Amir A. Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Mekelweg 2, 2628CD, Delft, the Netherlands
| |
Collapse
|
18
|
Zhang Y, Jiang N, Gan Z. Poly(ε-Caprolactone) Substrates with Micro/Nanohierarchical Patterned Structures for Cell Culture. Macromol Biosci 2022; 22:e2200300. [PMID: 36086924 DOI: 10.1002/mabi.202200300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/23/2022] [Indexed: 01/15/2023]
Abstract
A simple, efficient and controllable one-step template method is proposed to fabricate poly(ε-caprolactone) substrates with micro/nanohierarchical patterned structures. Two kinds of geometric patterns with and without nanowires, i.e., hexagonal and strip with controllable island size and spacing are designed and fabricated. Furthermore, the influence of geometric patterns, island size, island spacing, and patterned nanowires (pNW) on the growth behavior of MG-63 cells is studied in terms of cell density, distribution, proliferation, morphogenesis, and cellular alignment. It is found that MG-63 cells prefer to adhere and grow on the substrate with smaller island size or spacing. Moreover, unlike the hexagonal structure, the strip structure can guide cellular alignment on its surface. In addition, the microisland structures and the pNW play different roles in promoting cell proliferation, distribution, and morphogenesis. It is concluded that the growth behavior of MG-63 cells can be well controlled by precisely adjusting the micro/nanostructure of the substrate surface. A simple and effective method is provided here for the regulation of cell growth behavior.
Collapse
Affiliation(s)
- Yuan Zhang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ni Jiang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhihua Gan
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
19
|
Anbumani S, da Silva AM, Alaferdov A, Puydinger dos Santos MV, Carvalho IGB, de Souza e Silva M, Moshkalev S, Carvalho HF, de Souza AA, Cotta MA. Physiochemically Distinct Surface Properties of SU-8 Polymer Modulate Bacterial Cell-Surface Holdfast and Colonization. ACS APPLIED BIO MATERIALS 2022; 5:4903-4912. [PMID: 36162102 PMCID: PMC9580523 DOI: 10.1021/acsabm.2c00632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/13/2022] [Indexed: 11/28/2022]
Abstract
SU-8 polymer is an excellent platform for diverse applications due to its high aspect ratio of micro/nanostructure fabrication and exceptional physicochemical and biocompatible properties. Although SU-8 polymer has often been investigated for various biological applications, how its surface properties influence the interaction of bacterial cells with the substrate and its colonization is poorly understood. In this work, we tailor SU-8 nanoscale surface properties to investigate single-cell motility, adhesion, and successive colonization of phytopathogenic bacteria, Xylella fastidiosa. Different surface properties of SU-8 thin films have been prepared using photolithography processing and oxygen plasma treatment. A more significant density of carboxyl groups in hydrophilic plasma-treated SU-8 surfaces promotes faster cell motility in the earlier growth stage. The hydrophobic nature of pristine SU-8 surfaces shows no trackable bacterial motility and 5-10 times more single cells adhered to the surface than its plasma-treated counterpart. In addition, plasma-treated SU-8 samples suppressed bacterial adhesion, with surfaces showing less than 5% coverage. These results not only showcase that SU-8 surface properties can impact the spatiotemporal bacterial behavior but also provide insights into pathogens' prominent ability to evolve and adapt to different surface properties.
Collapse
Affiliation(s)
- Silambarasan Anbumani
- Institute
of Physics “Gleb Wataghin”, University of Campinas, Campinas, SP 13083-859, Brazil
| | - Aldeliane M. da Silva
- Institute
of Physics “Gleb Wataghin”, University of Campinas, Campinas, SP 13083-859, Brazil
| | - Andrei Alaferdov
- Center
for Semiconductor Components and Nanotechnologies, University of Campinas, Campinas, SP 13083-870, Brazil
| | | | - Isis G. B. Carvalho
- Citrus
Center APTA “Sylvio Moreira” Agronomic Institute of
Campinas, Cordeirópolis, SP 13490-970, Brazil
| | - Mariana de Souza e Silva
- Citrus
Center APTA “Sylvio Moreira” Agronomic Institute of
Campinas, Cordeirópolis, SP 13490-970, Brazil
| | - Stanislav Moshkalev
- Center
for Semiconductor Components and Nanotechnologies, University of Campinas, Campinas, SP 13083-870, Brazil
| | - Hernandes F. Carvalho
- Department
of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Alessandra A. de Souza
- Citrus
Center APTA “Sylvio Moreira” Agronomic Institute of
Campinas, Cordeirópolis, SP 13490-970, Brazil
| | - Monica A. Cotta
- Institute
of Physics “Gleb Wataghin”, University of Campinas, Campinas, SP 13083-859, Brazil
| |
Collapse
|
20
|
Modeling Self-Rollable Elastomeric Films for Building Bioinspired Hierarchical 3D Structures. Int J Mol Sci 2022; 23:ijms23158467. [PMID: 35955601 PMCID: PMC9369037 DOI: 10.3390/ijms23158467] [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: 05/19/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 01/09/2023] Open
Abstract
In this work, an innovative model is proposed as a design tool to predict both the inner and outer radii in rolled structures based on polydimethylsiloxane bilayers. The model represents an improvement of Timoshenko's formula taking into account the friction arising from contacts between layers arising from rolling by more than one turn, hence broadening its application field towards materials based on elastomeric bilayers capable of large deformations. The fabricated structures were also provided with surface topographical features that would make them potentially usable in different application scenarios, including cell/tissue engineering ones. The bilayer design parameters were varied, such as the initial strain (from 20 to 60%) and the bilayer thickness (from 373 to 93 µm). The model matched experimental data on the inner and outer radii nicely, especially when a high friction condition was implemented in the model, particularly reducing the error below 2% for the outer diameter while varying the strain. The model outperformed the current literature, where self-penetration is not excluded, and a single value of the radius of spontaneous rolling is used to describe multiple rolls. A complex 3D bioinspired hierarchical elastomeric microstructure made of seven spirals arranged like a hexagon inscribed in a circumference, similar to typical biological architectures (e.g., myofibrils within a sarcolemma), was also developed. In this case also, the model effectively predicted the spirals' features (error smaller than 18%), opening interesting application scenarios in the modeling and fabrication of bioinspired materials.
Collapse
|
21
|
Cimmino C, Netti PA, Ventre M. A switchable light-responsive azopolymer conjugating protein micropatterns with topography for mechanobiological studies. Front Bioeng Biotechnol 2022; 10:933410. [PMID: 35935479 PMCID: PMC9355574 DOI: 10.3389/fbioe.2022.933410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
Stem cell shape and mechanical properties in vitro can be directed by geometrically defined micropatterned adhesion substrates. However, conventional methods are limited by the fixed micropattern design, which cannot recapitulate the dynamic changes of the natural cell microenvironment. Current methods to fabricate dynamic platforms usually rely on complex chemical strategies or require specialized apparatuses. Also, with these methods, the integration of dynamic signals acting on different length scales is not straightforward, whereas, in some applications, it might be beneficial to act on both a microscale level, that is, cell shape, and a nanoscale level, that is, cell adhesions. Here, we exploited a confocal laser-based technique on a light-responsive azopolymer displaying micropatterns of adhesive islands. The laser light promotes a directed mass migration and the formation of submicrometric topographic relieves. Also, by changing the surface chemistry, the surfacing topography affects cell spreading and shape. This method enabled us to monitor in a non-invasive manner the dynamic changes in focal adhesions, cytoskeleton structures, and nucleus conformation that followed the changes in the adhesive characteristic of the substrate. Focal adhesions reconfigured after the surfacing of the topography, and the actin filaments reoriented to coalign with the newly formed adhesive island. Changes in cell morphology also affected nucleus shape, chromatin conformation, and cell mechanics with different timescales. The reported strategy can be used to investigate mechanotransduction-related events dynamically by controlling cell adhesion at cell shape and focal adhesion levels. The integrated technique enables achieving a submicrometric resolution in a facile and cost-effective manner.
Collapse
Affiliation(s)
- Chiara Cimmino
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Fondazione Istituto Italiano di Tecnologia, Naples, Italy
| | - Paolo A. Netti
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Fondazione Istituto Italiano di Tecnologia, Naples, Italy
- Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy
| | - Maurizio Ventre
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for Healthcare@CRIB, Fondazione Istituto Italiano di Tecnologia, Naples, Italy
- Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy
- *Correspondence: Maurizio Ventre,
| |
Collapse
|
22
|
Ganjian M, Modaresifar K, Rompolas D, Fratila-Apachitei LE, Zadpoor AA. Nanoimprinting for high-throughput replication of geometrically precise pillars in fused silica to regulate cell behavior. Acta Biomater 2022; 140:717-729. [PMID: 34875357 DOI: 10.1016/j.actbio.2021.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/04/2021] [Accepted: 12/01/2021] [Indexed: 12/31/2022]
Abstract
Developing high-throughput nanopatterning techniques that also allow for precise control over the dimensions of the fabricated features is essential for the study of cell-nanopattern interactions. Here, we developed a process that fulfills both of these criteria. Firstly, we used electron-beam lithography (EBL) to fabricate precisely controlled arrays of submicron pillars with varying values of interspacing on a large area of fused silica. Two types of etching procedures with two different systems were developed to etch the fused silica and create the final desired height. We then studied the interactions of preosteoblasts (MC3T3-E1) with these pillars. Varying interspacing was observed to significantly affect the morphological characteristics of the cell, the organization of actin fibers, and the formation of focal adhesions. The expression of osteopontin (OPN) significantly increased on the patterns, indicating the potential of the pillars for inducing osteogenic differentiation. The EBL pillars were thereafter used as master molds in two subsequent processing steps, namely soft lithography and thermal nanoimprint lithography for high-fidelity replication of the pillars on the substrates of interest. The molding parameters were optimized to maximize the fidelity of the generated patterns and minimize the wear and tear of the master mold. Comparing the replicated feature with those present on the original mold confirmed that the geometry and dimensions of the replicated pillars closely resemble those of the original ones. The method proposed in this study, therefore, enables the precise fabrication of submicron- and nanopatterns on a wide variety of materials that are relevant for systematic cell studies. STATEMENT OF SIGNIFICANCE: Submicron pillars with specific dimensions on the bone implants have been proven to be effective in controlling cell behaviors. Nowadays, numerous methods have been proposed to produce bio-instructive submicron-topographies. However, most of these techniques are suffering from being low-throughput, low-precision, and expensive. Here, we developed a high-throughput nanopatterning technique that allows for control over the dimensions of the features for the study of cell-nanotopography interactions. Assessing the adaptation of preosteoblast cells showed the potential of the pillars for inducing osteogenic differentiation. Afterward, the pillars were used for high-fidelity replication of the bio-instructive features on the substrates of interest. The results show the advantages of nanoimprint lithography as a unique technique for the patterning of large areas of bio-instructive surfaces.
Collapse
|
23
|
Moses JC, Dey S, Bandyopadhyay A, Agarwala M, Mandal BB. Silk-Based Bioengineered Diaphyseal Cortical Bone Unit Enclosing an Implantable Bone Marrow toward Atrophic Nonunion Grafting. Adv Healthc Mater 2022; 11:e2102031. [PMID: 34881525 DOI: 10.1002/adhm.202102031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/02/2021] [Indexed: 12/11/2022]
Abstract
Postnatal fracture healing of atrophic long bone diaphyseal nonunions remains a challenge for orthopedic surgeons. Paucity of autologous spongiosa has potentiated the use of tissue engineered bone grafts to improve success rates of bone marrow engraftment used in plate reosteosynthesis. Herein, the development and in vitro validation of a "sandwich-type" biofabricated diaphyseal cross-sectional unit, with an outer mechanically robust bioprinted cortical bone shell, encompassing an engineered bone marrow, are reported. Channelized silk fibroin blend sponges derived from Bombyx mori and Antheraea assama help in developing compartmentalized endosteum, exhibiting specialized osteoblasts (endosteal niche) and discontinuous endothelium (vascular niche). The cellular cross-talk between these two niches triggered via integrin-mediated cell adhesion, enables in preserving quiescence state of CD34+ /CD38- hematopoietic stem cells and their recycling in the engineered marrow. The outer cortical bone strut is developed through multimaterial microextrusion bioprinting strategy. Osteogenically primed mesenchymal stem cells-laden silk fibroin-nano-hydroxyapatite bioink is bioprinted alongside paramagnetic Fe-doped bioactive glass-polycaprolactone blend thermoplastic ink, reinforcing it for mechanical stability. Pulsed magnetic field actuation positively influences the osteogenic commitment and maturation of the bioprinted constructs via mechanotransductory route. Therefore, the assembled engineered marrow and bioprinted cortical shell hold promise as potential orthobiologic substitutes toward atrophic nonunion repairs.
Collapse
Affiliation(s)
- Joseph Christakiran Moses
- Biomaterials and Tissue Engineering Laboratory Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Souradeep Dey
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Ashutosh Bandyopadhyay
- Biomaterials and Tissue Engineering Laboratory Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| | - Manoj Agarwala
- GNRC Institute of Medical Sciences (formerly known as Guwahati Neurological Research Centre) Guwahati Assam 781039 India
| | - Biman B. Mandal
- Biomaterials and Tissue Engineering Laboratory Department of Biosciences and Bioengineering Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- Centre for Nanotechnology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
- School of Health Science and Technology Indian Institute of Technology Guwahati Guwahati Assam 781039 India
| |
Collapse
|
24
|
Carthew J, Taylor JBJ, Garcia-Cruz MR, Kiaie N, Voelcker NH, Cadarso VJ, Frith JE. The Bumpy Road to Stem Cell Therapies: Rational Design of Surface Topographies to Dictate Stem Cell Mechanotransduction and Fate. ACS APPLIED MATERIALS & INTERFACES 2022; 14:23066-23101. [PMID: 35192344 DOI: 10.1021/acsami.1c22109] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cells sense and respond to a variety of physical cues from their surrounding microenvironment, and these are interpreted through mechanotransductive processes to inform their behavior. These mechanisms have particular relevance to stem cells, where control of stem cell proliferation, potency, and differentiation is key to their successful application in regenerative medicine. It is increasingly recognized that surface micro- and nanotopographies influence stem cell behavior and may represent a powerful tool with which to direct the morphology and fate of stem cells. Current progress toward this goal has been driven by combined advances in fabrication technologies and cell biology. Here, the capacity to generate precisely defined micro- and nanoscale topographies has facilitated the studies that provide knowledge of the mechanotransducive processes that govern the cellular response as well as knowledge of the specific features that can drive cells toward a defined differentiation outcome. However, the path forward is not fully defined, and the "bumpy road" that lays ahead must be crossed before the full potential of these approaches can be fully exploited. This review focuses on the challenges and opportunities in applying micro- and nanotopographies to dictate stem cell fate for regenerative medicine. Here, key techniques used to produce topographic features are reviewed, such as photolithography, block copolymer lithography, electron beam lithography, nanoimprint lithography, soft lithography, scanning probe lithography, colloidal lithography, electrospinning, and surface roughening, alongside their advantages and disadvantages. The biological impacts of surface topographies are then discussed, including the current understanding of the mechanotransductive mechanisms by which these cues are interpreted by the cells, as well as the specific effects of surface topographies on cell differentiation and fate. Finally, considerations in translating these technologies and their future prospects are evaluated.
Collapse
Affiliation(s)
- James Carthew
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Jason B J Taylor
- Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Maria R Garcia-Cruz
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Nasim Kiaie
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| | - Nicolas H Voelcker
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria 3168, Australia
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
- ARC Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, Victoria 3800, Australia
- CSIRO Manufacturing, Bayview Avenue, Clayton, VIC 3168, Australia
| | - Victor J Cadarso
- Mechanical and Aerospace Engineering, Monash University, Clayton, Victoria 3800, Australia
- Centre to Impact Antimicrobial Resistance, Monash University, Clayton, Victoria 3800, Australia
| | - Jessica E Frith
- Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
- ARC Centre for Cell and Tissue Engineering Technologies, Monash University, Clayton, Victoria 3800, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
25
|
Lin J, Song X, Yin H, Song N, Wang Y, Li Z, Luo F, Tan H, He X, Li J. Citicoline–liposome/polyurethane composite scaffolds regulate the inflammatory response of microglia to promote nerve regeneration. JOURNAL OF MATERIALS SCIENCE 2022; 57:2073-2088. [DOI: 10.1007/s10853-021-06628-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 10/12/2021] [Indexed: 01/03/2025]
|
26
|
Ivanov AA, Kuznetsova AV, Popova OP, Danilova TI, Yanushevich OO. Modern Approaches to Acellular Therapy in Bone and Dental Regeneration. Int J Mol Sci 2021; 22:13454. [PMID: 34948251 PMCID: PMC8708083 DOI: 10.3390/ijms222413454] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 12/09/2021] [Accepted: 12/13/2021] [Indexed: 02/06/2023] Open
Abstract
An approach called cell-free therapy has rapidly developed in regenerative medicine over the past decade. Understanding the molecular mechanisms and signaling pathways involved in the internal potential of tissue repair inspires the development of new strategies aimed at controlling and enhancing these processes during regeneration. The use of stem cell mobilization, or homing for regeneration based on endogenous healing mechanisms, prompted a new concept in regenerative medicine: endogenous regenerative medicine. The application of cell-free therapeutic agents leading to the recruitment/homing of endogenous stem cells has advantages in overcoming the limitations and risks associated with cell therapy. In this review, we discuss the potential of cell-free products such as the decellularized extracellular matrix, growth factors, extracellular vesicles and miRNAs in endogenous bone and dental regeneration.
Collapse
Affiliation(s)
- Alexey A. Ivanov
- Laboratory of Molecular and Cellular Pathology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (A.V.K.); (O.P.P.); (T.I.D.)
| | - Alla V. Kuznetsova
- Laboratory of Molecular and Cellular Pathology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (A.V.K.); (O.P.P.); (T.I.D.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Str., 119334 Moscow, Russia
| | - Olga P. Popova
- Laboratory of Molecular and Cellular Pathology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (A.V.K.); (O.P.P.); (T.I.D.)
| | - Tamara I. Danilova
- Laboratory of Molecular and Cellular Pathology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (A.V.K.); (O.P.P.); (T.I.D.)
| | - Oleg O. Yanushevich
- Department of Paradontology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia;
| |
Collapse
|
27
|
Wagner G, Eggers B, Duddeck D, Kramer FJ, Bourauel C, Jepsen S, Deschner J, Nokhbehsaim M. Influence of cold atmospheric plasma on dental implant materials - an in vitro analysis. Clin Oral Investig 2021; 26:2949-2963. [PMID: 34907458 PMCID: PMC8898257 DOI: 10.1007/s00784-021-04277-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/01/2021] [Indexed: 12/25/2022]
Abstract
Background and objectives Alterations in the microenvironment of implant surfaces could influence the cellular crosstalk and adhesion patterns of dental implant materials. Cold plasma has been described to have an influence on cells, tissues, and biomaterials. Hence, the mechanisms of osseointegration may be altered by non-thermal plasma treatment depending on different chemical compositions and surface coatings of the biomaterial. The aim of the present study is to investigate the influence of cold atmospheric plasma (CAP) treatment on implant surfaces and its biological and physicochemical side effects. Materials and methods Dental implant discs from titanium and zirconia with different surface modifications were treated with CAP at various durations. Cell behavior and adhesion patterns of human gingival fibroblast (HGF-1) and osteoblast-like cells (MG-63) were examined using scanning electron microscopy and fluorescence microscopy. Surface chemical characterization was analyzed using energy-dispersive X-ray spectroscopy (EDS). Quantitative analysis of cell adhesion, proliferation, and extracellular matrix formation was conducted including real-time PCR. Results CAP did not affect the elemental composition of different dental implant materials. Additionally, markers for cell proliferation, extracellular matrix formation, and cell adhesion were differently regulated depending on the application time of CAP treatment in MG-63 cells and gingival fibroblasts. Conclusions CAP application is beneficial for dental implant materials to allow for faster proliferation and adhesion of cells from the surrounding tissue on both titanium and zirconia implant surfaces with different surface properties. Clinical relevance The healing capacity provided through CAP treatment could enhance osseointegration of dental implants and has the potential to serve as an effective treatment option in periimplantitis therapy.
Collapse
Affiliation(s)
- Gunar Wagner
- Department of Periodontology, Operative and Preventive Dentistry, Center of Dento-Maxilo-Facial Medicine, University of Bonn, Welschnonnenstr. 17, 53111, Bonn, Germany.
| | - Benedikt Eggers
- Department of Oral Surgery, Center of Dento-Maxillo-Facial Medicine, University of Bonn, 53111, Bonn, Germany
| | - Dirk Duddeck
- Department of Prosthodontics, Geriatric Dentistry and Craniomandibular Disorders, University Charité Berlin, 14197, Berlin, Germany.,Research Department, CleanImplant Foundation, 10117, Berlin, Germany
| | - Franz-Josef Kramer
- Department of Oral Surgery, Center of Dento-Maxillo-Facial Medicine, University of Bonn, 53111, Bonn, Germany.,Department of Cranio-Maxillofacial Surgery, Center of Dento-Maxillo-Facial Medicine, University of Bonn, 53111, Bonn, Germany
| | - Christoph Bourauel
- Department of Oral Technology, School of Dentistry, University of Bonn, 53111, Bonn, Germany
| | - Søren Jepsen
- Department of Periodontology, Operative and Preventive Dentistry, Center of Dento-Maxilo-Facial Medicine, University of Bonn, Welschnonnenstr. 17, 53111, Bonn, Germany
| | - James Deschner
- Department of Periodontology and Operative Dentistry, University of Mainz, 55131, Mainz, Germany
| | - Marjan Nokhbehsaim
- Section of Experimental Dento-Maxillo-Facial Medicine, Center of Dento-Maxillo-Facial Medicine, University of Bonn, 53111, Bonn, Germany
| |
Collapse
|
28
|
Williams DF. Biocompatibility pathways and mechanisms for bioactive materials: The bioactivity zone. Bioact Mater 2021; 10:306-322. [PMID: 34901548 PMCID: PMC8636667 DOI: 10.1016/j.bioactmat.2021.08.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 08/07/2021] [Indexed: 12/14/2022] Open
Abstract
This essay analyzes the scientific evidence that forms the basis of bioactive materials, covering the fundamental understanding of bioactivity phenomena and correlation with the mechanisms of biocompatibility of biomaterials. This is a detailed assessment of performance in areas such as bone-induction, cell adhesion, immunomodulation, thrombogenicity and antimicrobial behavior. Bioactivity is the modulation of biological activity by characteristics of the interfacial region that incorporates the material surface and the immediate local host tissue. Although the term ‘bioactive material’ is widely used and has a well understood general meaning, it would be useful now to concentrate on this interfacial region, considered as ‘the bioactivity zone’. Bioactivity phenomena are either due to topographical/micromechanical characteristics, or to biologically active species that are presented in the bioactivity zone. Examples of topographical/micromechanical effects are the modulation of the osteoblast – osteoclast balance, nanotopographical regulation of cell adhesion, and bactericidal nanostructures. Regulation of bioactivity by biologically active species include their influence, especially of metal ions, on signaling pathways in bone formation, the role of cell adhesion molecules and bioactive peptides in cell attachment, macrophage polarization by immunoregulatory molecules and antimicrobial peptides. While much experimental data exists to demonstrate the potential of such phenomena, there are considerable barriers to their effective clinical translation. This essay shows that there is solid scientific evidence of the existence of bioactivity mechanisms that are associated with some types of biomaterials, especially when the material is modified in a manner designed to specifically induce that activity.
Collapse
Affiliation(s)
- David F Williams
- Wake Forest Institute of Regenerative Medicine, 391 Technology Way. Winston-Salem, North Carolina, 27101, USA
| |
Collapse
|
29
|
Ruland A, Schenker S, Schirmer L, Friedrichs J, Meinhardt A, Schwartz VB, Kaiser N, Konradi R, MacDonald W, Helmecke T, Sikosana MKLN, Valtin J, Hahn D, Renner LD, Werner C, Freudenberg U. Amphiphilic Copolymers for Versatile, Facile, and In Situ Tunable Surface Biofunctionalization. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2102489. [PMID: 34431569 PMCID: PMC11468472 DOI: 10.1002/adma.202102489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/13/2021] [Indexed: 06/13/2023]
Abstract
Precision surface engineering is key to advanced biomaterials. A new platform of PEGylated styrene-maleic acid copolymers for adsorptive surface biofunctionalization is reported. Balanced amphiphilicity renders the copolymers water-soluble but strongly affine for surfaces. Fine-tuning of their molecular architecture provides control over adsorptive anchorage onto specific materials-which is why they are referred to as "anchor polymers" (APs)-and over structural characteristics of the adsorbed layers. Conjugatable with an array of bioactives-including cytokine-complexing glycosaminoglycans, cell-adhesion-mediating peptides and antimicrobials-APs can be applied to customize materials for demanding biotechnologies in uniquely versatile, simple, and robust ways. Moreover, homo- and heterodisplacement of adsorbed APs provide unprecedented means of in situ alteration and renewal of the functionalized surfaces. The related options are exemplified with proof-of-concept experiments of controlled bacterial adhesion, human umbilical vein endothelial cell, and induced pluripotent cell growth on AP-functionalized surfaces.
Collapse
Affiliation(s)
- André Ruland
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| | - Saskia Schenker
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| | - Lucas Schirmer
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| | - Jens Friedrichs
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| | - Andrea Meinhardt
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| | | | - Nadine Kaiser
- BASF SE, RAM/OB – B001Carl‐Bosch‐Strasse 3867056Ludwigshafen am RheinGermany
| | - Rupert Konradi
- BASF SE, RAM/OB – B001Carl‐Bosch‐Strasse 3867056Ludwigshafen am RheinGermany
| | - William MacDonald
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
- Brown UniversityProvidenceRI02912USA
| | - Tina Helmecke
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| | - Melissa K. L. N. Sikosana
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| | - Juliane Valtin
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| | - Dominik Hahn
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| | - Lars D. Renner
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| | - Carsten Werner
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
- Center for Regenerative Therapies Dresden (CRTD)Technische Universität DresdenFetscherstraße 10501307DresdenGermany
| | - Uwe Freudenberg
- Leibniz Institute of Polymer Research Dresden (IPF)Max Bergmann Center of Biomaterials Dresden (MBC)Hohe Str. 601069DresdenGermany
| |
Collapse
|
30
|
Mackay BS, Marshall K, Grant-Jacob JA, Kanczler J, Eason RW, Oreffo ROC, Mills B. The future of bone regeneration: integrating AI into tissue engineering. Biomed Phys Eng Express 2021; 7. [PMID: 34271556 DOI: 10.1088/2057-1976/ac154f] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/16/2021] [Indexed: 01/16/2023]
Abstract
Tissue engineering is a branch of regenerative medicine that harnesses biomaterial and stem cell research to utilise the body's natural healing responses to regenerate tissue and organs. There remain many unanswered questions in tissue engineering, with optimal biomaterial designs still to be developed and a lack of adequate stem cell knowledge limiting successful application. Advances in artificial intelligence (AI), and deep learning specifically, offer the potential to improve both scientific understanding and clinical outcomes in regenerative medicine. With enhanced perception of how to integrate artificial intelligence into current research and clinical practice, AI offers an invaluable tool to improve patient outcome.
Collapse
Affiliation(s)
- Benita S Mackay
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Karen Marshall
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6HW, United Kingdom
| | - James A Grant-Jacob
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Janos Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6HW, United Kingdom
| | - Robert W Eason
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom.,Institute of Developmental Sciences, Faculty of Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, SO16 6HW, United Kingdom.,Institute of Developmental Sciences, Faculty of Life Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| | - Ben Mills
- Optoelectronics Research Centre, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, United Kingdom
| |
Collapse
|
31
|
Eftekhari BS, Eskandari M, Janmey PA, Samadikuchaksaraei A, Gholipourmalekabadi M. Conductive chitosan/polyaniline hydrogel with cell-imprinted topography as a potential substrate for neural priming of adipose derived stem cells. RSC Adv 2021; 11:15795-15807. [PMID: 35481217 PMCID: PMC9029165 DOI: 10.1039/d1ra00413a] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022] Open
Abstract
Biophysical characteristics of engineered scaffolds such as topography and electroconductivity have shown potentially beneficial effects on stem cell morphology, proliferation, and differentiation toward neural cells. In this study, we fabricated a conductive hydrogel made from chitosan (CS) and polyaniline (PANI) with induced PC12 cell surface topography using a cell imprinting technique to provide both topographical properties and conductivity in a platform. The engineered hydrogel's potential for neural priming of rat adipose-derived stem cells (rADSCs) was determined in vitro. The biomechanical analysis revealed that the electrical conductivity, stiffness, and hydrophobicity of flat (F) and cell-imprinted (CI) substrates increased with increased PANI content in the CS/PANI scaffold. The conductive substrates exhibited a lower degradation rate compared to non-conductive substrates. According to data obtained from F-actin staining and AFM micrographs, both CI(CS) and CI(CS-PANI) substrates induced the morphology of rADSCs from their irregular shape (on flat substrates) into the elongated and bipolar shape of the neuronal-like PC12 cells. Immunostaining analysis revealed that both CI(CS) and CI (CS-PANI) significantly upregulated the expression of GFAP and MAP2, two neural precursor-specific genes, in rADSCs compared with flat substrates. Although the results reveal that both cell-imprinted topography and electrical conductivity affect the neural lineage differentiation, some data demonstrate that the topography effects of the cell-imprinted surface have a more critical role than electrical conductivity on neural priming of ADSCs. The current study provides new insight into the engineering of scaffolds for nerve tissue engineering.
Collapse
Affiliation(s)
- Behnaz Sadat Eftekhari
- Department of Biomedical Engineering, Amirkabir University of Technology 424 Hafez Ave Tehran 15875-4413 Iran +98 21 6454 23 62
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania 1010 Vagelos Research Laboratories, 3340 Smith Walk Philadelphia PA 19104-6383 USA +1 215 573 6815 +1 215 573 7380
| | - Mahnaz Eskandari
- Department of Biomedical Engineering, Amirkabir University of Technology 424 Hafez Ave Tehran 15875-4413 Iran +98 21 6454 23 62
| | - Paul A Janmey
- Department of Physiology, Institute for Medicine and Engineering, University of Pennsylvania 1010 Vagelos Research Laboratories, 3340 Smith Walk Philadelphia PA 19104-6383 USA +1 215 573 6815 +1 215 573 7380
| | | | - Mazaher Gholipourmalekabadi
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences Tehran Iran
- Cellular and Molecular Research Centre, Iran University of Medical Sciences Tehran Iran
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences Tehran Iran
| |
Collapse
|
32
|
Guo S, Huang H, Zeng W, Jiang Z, Wang X, Huang W, Wang X. Facile cell patterning induced by combined surface topography and chemistry on polydopamine-defined nanosubstrates. NANOTECHNOLOGY 2021; 32:145303. [PMID: 33361576 DOI: 10.1088/1361-6528/abd6d2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cell patterning holds significant implications for cell-based analysis and high-throughput screening. The challenge and key factor for formation of cell patterns is to precisely modulate the interaction between cells and substrate surfaces. Many nanosubstrates have been developed to control cell adhesion and patterning, however, requirements of complicated fabrication procedures, harsh reaction conditions, and delicate manipulation are not routinely feasible. Here, we developed a hierarchical polydimethylsiloxane nanosubstrate (HPNS) coated with mussel-inspired polydopamine (PDA) micropatterns for effective cell patterning, depending on both surface topography and chemistry. HPNSs obtained by facile template-assisted replication brought enhanced topographic interaction between cells and substrates, but they were innately hydrophobic and cell-repellent. The hydrophobic nanosubstrates were converted to be hydrophilic after PDA coatings formed via spontaneous self-polymerization, which greatly facilitated cell adhesion. As such, without resorting to any external forces or physical constraints, cells selectively adhered and spread on spatially defined PDA regions with high efficiency, and well-defined cell microarrays could be formed within 20 min. Therefore, this easy-to-fabricate nanosubstrate with no complex chemical modification will afford a facile yet effective platform for rapid cell patterning.
Collapse
Affiliation(s)
- Shan Guo
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Haiyan Huang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Weiwu Zeng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Zhuoran Jiang
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Xin Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Weihua Huang
- Key Laboratory of Analytical Chemistry for Biology and Medicine (Ministry of Education), College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Xinghuan Wang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| |
Collapse
|
33
|
Cui Y, Li B, Wang X, Tang R. Organism–Materials Integration: A Promising Strategy for Biomedical Applications. ADVANCED NANOBIOMED RESEARCH 2021. [DOI: 10.1002/anbr.202000044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Affiliation(s)
- Yihao Cui
- Center for Biomaterials and Biopathways Department of Chemistry Zhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
| | - Benke Li
- Center for Biomaterials and Biopathways Department of Chemistry Zhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
| | - Xiaoyu Wang
- Qiushi Academy for Advanced Studies Zhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
| | - Ruikang Tang
- Center for Biomaterials and Biopathways Department of Chemistry Zhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
- Qiushi Academy for Advanced Studies Zhejiang University No. 38 Zheda Road Hangzhou Zhejiang 310027 China
| |
Collapse
|
34
|
Biazar E, Kamalvand M, Avani F. Recent advances in surface modification of biopolymeric nanofibrous scaffolds. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2020.1857383] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Esmaeil Biazar
- Department of Biomaterials Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Mahshad Kamalvand
- Department of Biomaterials Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Farzaneh Avani
- Biomedical Engineering Faculty, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| |
Collapse
|
35
|
Pennacchio FA, Nastały P, Poli A, Maiuri P. Tailoring Cellular Function: The Contribution of the Nucleus in Mechanotransduction. Front Bioeng Biotechnol 2021; 8:596746. [PMID: 33490050 PMCID: PMC7820809 DOI: 10.3389/fbioe.2020.596746] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Cells sense a variety of different mechanochemical stimuli and promptly react to such signals by reshaping their morphology and adapting their structural organization and tensional state. Cell reactions to mechanical stimuli arising from the local microenvironment, mechanotransduction, play a crucial role in many cellular functions in both physiological and pathological conditions. To decipher this complex process, several studies have been undertaken to develop engineered materials and devices as tools to properly control cell mechanical state and evaluate cellular responses. Recent reports highlight how the nucleus serves as an important mechanosensor organelle and governs cell mechanoresponse. In this review, we will introduce the basic mechanisms linking cytoskeleton organization to the nucleus and how this reacts to mechanical properties of the cell microenvironment. We will also discuss how perturbations of nucleus-cytoskeleton connections, affecting mechanotransduction, influence health and disease. Moreover, we will present some of the main technological tools used to characterize and perturb the nuclear mechanical state.
Collapse
Affiliation(s)
- Fabrizio A. Pennacchio
- FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Paulina Nastały
- FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology (IFOM), Milan, Italy
- Laboratory of Translational Oncology, Institute of Medical Biotechnology and Experimental Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Alessandro Poli
- FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Paolo Maiuri
- FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology (IFOM), Milan, Italy
| |
Collapse
|
36
|
De Martino S, Netti PA. Dynamic azopolymeric interfaces for photoactive cell instruction. BIOPHYSICS REVIEWS 2020; 1:011302. [PMID: 38505629 PMCID: PMC10903377 DOI: 10.1063/5.0025175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/26/2020] [Indexed: 03/21/2024]
Abstract
The ability to affect a wide range of biophysical properties through the use of light has led to the development of dynamic cell instructive materials. Using photoresponsive materials such as azopolymers, smart systems that use external, minimally damaging, light irradiation can be used to trigger specific surface morpho-physical properties in the presence of living cells. The interaction of light with an azopolymer film induces a mass migration phenomenon, allowing a variety of topographic patterns to be embossed on the polymeric film. Photoisomerization induces conformational changes at the molecular and macroscopic scale, resulting in light-induced variations of substrate morphological, physical, and mechanical properties. In this review, we discuss the photoactuation of azopolymeric interfaces to provide guidelines for the engineering and design of azopolymer films. Laser micropatterning for the modulation of azopolymer surfaces is examined as a way to diversify the capabilities of these polymers in cellular systems. Mass migration effects induced by azopolymer switching provides a foundation for performing a broad range of cellular manipulation techniques. Applications of azopolymers are explored in the context of dynamic culture systems, gaining insight into the complex processes involved in dynamic cell-material interactions. The review highlights azopolymers as a candidate for various applications in cellular control, including cell alignment, migration, gene expression, and others. Recent advances have underlined the importance of these systems in applications regarding three-dimensional cell culture and stem cell morphology. Azopolymers can be used not only to manipulate cells but also to probe for mechanistic studies of cellular crosstalk in response to chemical and mechanical stimuli.
Collapse
|
37
|
Suter N, Stebel S, Rianna C, Radmacher M, Brüggemann D. Spatial patterning of nanofibrous collagen scaffolds modulates fibroblast morphology. Biofabrication 2020; 13:015007. [DOI: 10.1088/1758-5090/abb744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
38
|
Qi C, Jin Y, Chen Y, Li W, Li Y, Liang K, Li Y, Zhang Y, Du Y. TGase-mediated cell membrane modification and targeted cell delivery to inflammatory endothelium. Biomaterials 2020; 269:120276. [PMID: 32797997 DOI: 10.1016/j.biomaterials.2020.120276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/19/2020] [Accepted: 07/31/2020] [Indexed: 10/23/2022]
Abstract
Targeted cell delivery to lesion sites via minimally invasive approach remains an unmet need in regenerative medicine to endow controlled cell distribution and minimized side-effects. Current cell modification approaches to improve cell delivery tend to have adverse effects on cellular phenotype and functionality. Here, we rationally developed a facile and mild cell modification and targeted delivery strategy leveraging endogenous tissue transglutaminase (TGase) expressed on the surface of MSCs (Mesenchymal Stem Cells) and inflammatory endothelial cells (ECs). Cell modification by functional peptides was accomplished simply via TGase catalyzed cross-linking with naturally-expressed MSCs membrane proteins (e.g. Annexin II), without detectable disturbance of cellular viability and functionality. The modified functional peptides could facilitate adhesion of MSCs to inflammatory ECs (with up-regulated TGase expression compared with normal ECs) in vitro, as demonstrated by a one-fold increase of the MSC-EC adhesion force measured by atomic force microscopy (AFM) and by targeted delivery of modified MSC to inflammatory ECs in a flow chamber assay. When transplanted in vivo, modified MSCs demonstrated a dramatic increase in targeted efficiency to inflammatory endothelium compared with non-modified MSCs in both mice ear inflammation and acute/chronic liver injury models. The cell membrane modification strategy and targeted cell delivery mechanism described here can be readily extended for empowering cell engineering and cell therapy with multifaceted functionalities to combat refractory diseases.
Collapse
Affiliation(s)
- Chunxiao Qi
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China; Key Laboratory of Bioactive Materials for Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yuhong Jin
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuyang Chen
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenjing Li
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yaqian Li
- Central Laboratories, Department of Scientific Research, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Kai Liang
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Yan Li
- Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing, 100101, China
| | - Yonghui Zhang
- School of Pharmaceutical Sciences, MOE Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
39
|
Controlling osteoblast morphology and proliferation via surface micro-topographies of implant biomaterials. Sci Rep 2020; 10:12810. [PMID: 32732908 PMCID: PMC7393177 DOI: 10.1038/s41598-020-69685-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/15/2020] [Indexed: 12/20/2022] Open
Abstract
Current research on surface modifications has yielded advanced implant biomaterials. Various implant surface modifications have been shown to be promising in improving bone target cell response, but more comprehensive studies whether certain implant surface modifications can directly target cell behavioural features such as morphogenesis and proliferation are needed. Here, we studied the response of primary alveolar bone cells on various implant surface modifications in terms of osteoblast morphology and proliferation in vitro. Analyses of surface modifications led to surface-related test parameters including the topographical parameters micro-roughness, texture aspect and surface enlargement as well as the physicochemical parameter surface wettability. We compared osteoblast morphology and proliferation towards the above-mentioned parameters and found that texture aspect and surface enlargement but not surface roughness or wettability exhibited significant impact on osteoblast morphology and proliferation. Detailed analysis revealed osteoblast proliferation as a function of cell morphology, substantiated by an osteoblast size- and morphology-dependent increase in mitotic activity. These findings show that implant surface topography controls cell behavioural morphology and subsequently cell proliferation, thereby opening the road for cell instructive biomaterials.
Collapse
|
40
|
Onak G, Gökmen O, Yaralı ZB, Karaman O. Enhanced osteogenesis of human mesenchymal stem cells by self-assembled peptide hydrogel functionalized with glutamic acid templated peptides. J Tissue Eng Regen Med 2020; 14:1236-1249. [PMID: 32615018 DOI: 10.1002/term.3095] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/27/2020] [Accepted: 06/29/2020] [Indexed: 12/22/2022]
Abstract
Self-assembling peptide (SAP) hydrogel has been shown to be an excellent biological material for three-dimensional cell culture and stimulatie cell migration and differentiation into the scaffold, as well as for repairing bone tissue defects. Herein, we designed one of the SAP scaffolds KLD (KLDLKLDLKLDL) through direct coupling to short bioactive motif O1 (EEGGC) and O2 (EEEEE) of which bioactivity on osteogenic differentiation was previously demonstrated and self-assembled in different concentrations (0.5%, 1%, and 2%). Our aim was to enhance osteogenesis and biomineralization of injectable SAP hydrogels with controlled mechanical properties so that the peptide hydrogel also becomes capable of being injected to bone defects. The molecular integration of the nanofibrous peptide scaffolds was observed using atomic force microscopy (AFM) and scanning electron microscopy (SEM). The rheological properties and degradation profile of SAP hydrogels were evaluated to ensure stability of SAPs. Compared with pure KLD scaffold, we found that these designed bioactive peptide scaffolds significantly promoted hMSCs proliferation depicted by biochemical analysis of alkaline phosphatase (ALP) activity, total calcium deposition. Moreover, key osteogenic markers of ALP activity, collagen type I (COL-1), osteopontin (OP), and osteocalcin (OCN) expression levels determined by real-time polymerase chain reaction (PCR) and immunofluorescence analysis were also significantly increased with the addition of glutamic acid residues to KLD. We demonstrated that the designed SAP scaffolds promoted the proliferation and osteogenic differentiation of hMSCs. Our results suggest that these designed bioactive peptide scaffolds may be useful for promoting bone tissue regeneration.
Collapse
Affiliation(s)
- Günnur Onak
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey
| | - Oğuzhan Gökmen
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey
| | - Ziyşan Buse Yaralı
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey
| | - Ozan Karaman
- Tissue Engineering and Regenerative Medicine Laboratory, Department of Biomedical Engineering, İzmir Katip Çelebi University, İzmir, Turkey.,Bonegraft Biomaterials Co., Ege University Technopolis, İzmir, Turkey
| |
Collapse
|
41
|
De Martino S, Cavalli S, Netti PA. Photoactive Interfaces for Spatio-Temporal Guidance of Mesenchymal Stem Cell Fate. Adv Healthc Mater 2020; 9:e2000470. [PMID: 32431096 DOI: 10.1002/adhm.202000470] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/19/2020] [Indexed: 01/30/2023]
Abstract
Patterned surfaces have proved effective in guiding stem cells commitment to a specific lineage by presenting highly ordered biophysical/biochemical cues at the cellmaterial interface. Their potency in controlling cell fate can be significantly empowered by encoding logic of space and time control of signal presentation. Here, azopolymeric photoactive interfaces are proposed to present/withdraw morphophysical signals to living cells using a green light trigger in a non-invasive spatio-temporal controlled way. To assess the potency of these dynamic platforms in controlling cell decision and fate, topography changes are actuated by light at specific times to reverse the fate of otherwise committed human mesenchymal stem cells (hMSC) toward osteoblastic lineage. It is first proved by dynamic change from ordered parallel patterning to flat or grid surfaces, that it is possible to induce cyclic cellular and nuclear stretches. Furthermore, by culturing hMSCs on a specific pattern known to prime them toward osteoblast lineage, the possibility to reroute or reverse stem cell fate decision by dynamic modulation of morphophysical signal is proved. To conclude, dynamic topographies can control the spatial conformation of hMSCs, modulate lineage reversal even after several weeks of culture and redirect lineage specification in response to light-induced changes in the microenvironment.
Collapse
Affiliation(s)
- Selene De Martino
- Center for Advanced Biomaterials for Healthcare, IIT@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, 53, Napoli, 80125, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB) and Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale, DICMAPI, Università degli Studi di Napoli Federico II, Piazzale Tecchio, 80, Napoli, 80125, Italy
| | - Silvia Cavalli
- Center for Advanced Biomaterials for Healthcare, IIT@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, 53, Napoli, 80125, Italy
| | - Paolo Antonio Netti
- Center for Advanced Biomaterials for Healthcare, IIT@CRIB, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, 53, Napoli, 80125, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB) and Dipartimento di Ingegneria Chimica dei Materiali e della Produzione Industriale, DICMAPI, Università degli Studi di Napoli Federico II, Piazzale Tecchio, 80, Napoli, 80125, Italy
| |
Collapse
|
42
|
Nicolas J, Magli S, Rabbachin L, Sampaolesi S, Nicotra F, Russo L. 3D Extracellular Matrix Mimics: Fundamental Concepts and Role of Materials Chemistry to Influence Stem Cell Fate. Biomacromolecules 2020; 21:1968-1994. [PMID: 32227919 DOI: 10.1021/acs.biomac.0c00045] [Citation(s) in RCA: 312] [Impact Index Per Article: 62.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Synthetic 3D extracellular matrices (ECMs) find application in cell studies, regenerative medicine, and drug discovery. While cells cultured in a monolayer may exhibit unnatural behavior and develop very different phenotypes and genotypes than in vivo, great efforts in materials chemistry have been devoted to reproducing in vitro behavior in in vivo cell microenvironments. This requires fine-tuning the biochemical and structural actors in synthetic ECMs. This review will present the fundamentals of the ECM, cover the chemical and structural features of the scaffolds used to generate ECM mimics, discuss the nature of the signaling biomolecules required and exploited to generate bioresponsive cell microenvironments able to induce a specific cell fate, and highlight the synthetic strategies involved in creating functional 3D ECM mimics.
Collapse
Affiliation(s)
- Julien Nicolas
- Université Paris-Saclay, CNRS, Institut Galien Paris-Saclay, , 92296 Châtenay-Malabry, France
| | - Sofia Magli
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milan, Italy
| | - Linda Rabbachin
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milan, Italy
| | - Susanna Sampaolesi
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milan, Italy
| | - Francesco Nicotra
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milan, Italy
| | - Laura Russo
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Piazza della Scienza 2, 20126 Milan, Italy
| |
Collapse
|
43
|
McLuckie M, Robotti F, Sanchez-Macedo N, Enderlin D, Frese L, Cheng PF, Levesque MP, Egaña JT, Poulikakos D, Ferrari A, Lindenblatt N. Lipoconstruct surface topography grating size influences vascularization onset in the dorsal skinfold chamber model. Acta Biomater 2020; 106:136-144. [PMID: 32044460 DOI: 10.1016/j.actbio.2020.01.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/10/2020] [Accepted: 01/31/2020] [Indexed: 12/14/2022]
Abstract
After skin tissue injury or pathological removal, vascularization timing is paramount in graft survival. As full thickness skin grafts often fail to become perfused over larger surfaces, split-thickness grafts are preferred and can be used together with biomaterials, which themselves are non-angiogenic. One way of promoting vascular ingrowth is to "pre-vascularize" an engineered substitute by introducing endothelial cells (ECs). Since it has been previously demonstrated that surface structured biomaterials have an effect on wound healing, skin regeneration, and fibrosis reduction, we proposed that a microvascular-rich lipoconstruct with anisotropic topographical cues could be a clinically translatable vascularization approach. Murine lipofragments were formed with three polydimethylsiloxane molds (flat, 5 µm, and 50 µm parallel gratings) and implanted into the dorsal skinfold chamber of male C57BL/6 mice. Vascular ingrowth was observed through intravital microscopy over 21 days and further assessed by histology and protein identification. Our investigation revealed that topographical feature size influenced the commencement of neovascular ingrowth, with 5 µm gratings exhibiting early construct perfusion at 3 days post-operation, and 50 µm being delayed until day 5. We therefore postulate that surface structured lipoconstructs may serve as an easily obtained and produced construct suitable for providing soft tissue and ECs to tissue defects. STATEMENT OF SIGNIFICANCE: Skin graft failures due to inadequate or uneven perfusion frequently occur and can be even more complicated in deep, difficult to heal wounds, or bone coverage. In complex injuries, biomaterials are often used to cover bone structures with a standard split thickness graft; however, perfusion can take up to 3 weeks. Thus, any means to promote faster and uniform vascularization could significantly reduce healing time, as well as lower patient down-time. As pre-vascularized constructs have reported success in research, we created a cost-efficient, translatable method with no additional laboratory time as adipose tissue can be harvested and used immediately. We further used surface topography as an aspect to modulate construct perfusion, which has been reported for the first time here.
Collapse
|
44
|
Narayanan N, Jiang C, Wang C, Uzunalli G, Whittern N, Chen D, Jones OG, Kuang S, Deng M. Harnessing Fiber Diameter-Dependent Effects of Myoblasts Toward Biomimetic Scaffold-Based Skeletal Muscle Regeneration. Front Bioeng Biotechnol 2020; 8:203. [PMID: 32266234 PMCID: PMC7105569 DOI: 10.3389/fbioe.2020.00203] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 03/02/2020] [Indexed: 12/28/2022] Open
Abstract
Regeneration of skeletal muscles is limited in cases of volumetric muscle loss and muscle degenerative diseases. Therefore, there is a critical need for developing strategies that provide cellular and structural support for skeletal muscle regeneration. In the present work, a bioengineered cell niche composed of mechanically competent aligned polyester fiber scaffolds is developed to mimic the oriented muscle fiber microenvironment by electrospinning poly(lactide-co-glycolide) (PLGA) using a custom-designed rotating collector with interspaced parallel blades. Aligned fiber scaffolds with fiber diameters ranging from 335 ± 154 nm to 3013 ± 531 nm are characterized for their bioactivities in supporting growth and differentiation of myoblasts. During in vitro culture, polymeric scaffolds with larger fiber diameter support enhanced alignment, growth, and differentiation of myoblasts associated with phosphorylation of p38 MAPK and upregulated expression of myogenin and myosin heavy chain. In vivo studies using a dystrophin-deficient mdx mouse model show that optimized fiber scaffolds seeded with primary myoblasts result in formation of dystrophin-positive myofibers network in tibialis anterior muscles. Collectively, these experiments provide critical insights on harnessing interactions between muscle cells and engineered fiber matrices to develop effective biomaterials for accelerated muscle regeneration.
Collapse
Affiliation(s)
- Naagarajan Narayanan
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
| | - Chunhui Jiang
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
| | - Chao Wang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Gözde Uzunalli
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
| | - Nicole Whittern
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
| | - Da Chen
- Department of Food Sciences, Purdue University, West Lafayette, IN, United States
| | - Owen G. Jones
- Department of Food Sciences, Purdue University, West Lafayette, IN, United States
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - Meng Deng
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Bindley Bioscience Center, Purdue University, West Lafayette, IN, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
- Department of Materials Engineering, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
45
|
Natale CF, Angrisano T, Pistelli L, Falco G, Calabrò V, Netti PA, Ventre M. Topographic Cues Impact on Embryonic Stem Cell Zscan4-Metastate. Front Bioeng Biotechnol 2020; 8:178. [PMID: 32211397 PMCID: PMC7069379 DOI: 10.3389/fbioe.2020.00178] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
The extracellular microenvironment proved to exert a potent regulatory effect over different aspects of Embryonic Stem Cells (ESCs) behavior. In particular, the employment of engineered culture surfaces aimed at modulating ESC self-organization resulted effective in directing ESCs toward specific fate decision. ESCs fluctuate among different levels of functional potency and in this context the Zscan4 gene marks the so-called "metastate," a cellular state in which ESCs retain both self-renewal and pluripotency capabilities. Here we investigated the impact of topographic cues on ESCs pluripotency, differentiation and organization capabilities. To this aim, we engineered culturing platforms of nanograted surfaces with different features size and we investigated their impact on ESCs multicellular organization and Zscan4 gene expression. We showed that the morphology of ESC-derived aggregates and Zscan4 expression are strictly intertwined. Our data suggest that ESC Zscan4 metastate can be promoted if the adhesive surface conditions guide cellular self-aggregation into 3D dome-like structure, in which both cell-material interactions and cell-cell contact are supportive for Zscan4 expression.
Collapse
Affiliation(s)
- Carlo F. Natale
- Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy
| | - Tiziana Angrisano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Luigi Pistelli
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Geppino Falco
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Viola Calabrò
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Paolo A. Netti
- Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| | - Maurizio Ventre
- Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| |
Collapse
|
46
|
Chopra P, Logun MT, White EM, Lu W, Locklin J, Karumbaiah L, Boons GJ. Fully Synthetic Heparan Sulfate-Based Neural Tissue Construct That Maintains the Undifferentiated State of Neural Stem Cells. ACS Chem Biol 2019; 14:1921-1929. [PMID: 31389687 DOI: 10.1021/acschembio.9b00401] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heparin and heparan sulfate (HS) are attractive components for constructing biomaterials due to their ability to recruit and regulate the activity of growth factors. The structural and functional heterogeneity of naturally derived heparin and HS is, however, an impediment for the preparation of biomaterials for regenerative medicine. To address this problem, we have prepared hydrogels modified by well-defined synthetic HS-derived disaccharides. Human induced pluripotent cell-derived neural stem cells (HIP-NSCs) encapsulated in a polyethylene glycol-based hydrogel modified by a pendent HS disaccharide that is a known ligand for fibroblast growth factor-2 (FGF-2) exhibited a significant increase in proliferation and self-renewal. This observation is important because evidence is emerging that undifferentiated stems cells can yield significant therapeutic benefits via their paracrine signaling mechanisms. Our data indicate that the HS disaccharide protects FGF-2, which has a very short biological half-live, from degradation. It is anticipated that, by careful selection of a synthetic HS oligosaccharide, it will be possible to control retention and release of specific growth factor, which in turn will provide control over cell fate.
Collapse
Affiliation(s)
- Pradeep Chopra
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
| | - Meghan T. Logun
- Regenerative Bioscience Center, ADS Complex, University of Georgia, 422 River Road, Athens, Georgia 30602, United States
| | - Evan M. White
- New Material Institute, University of Georgia, 220 Riverbend Road, Athens, Georgia 30602, United States
| | - Weigang Lu
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
- Department of Chemistry, University of Georgia, 140 Cedar Street, Athens, Georgia 30602, United States
| | - Jason Locklin
- New Material Institute, University of Georgia, 220 Riverbend Road, Athens, Georgia 30602, United States
- Department of Chemistry, University of Georgia, 140 Cedar Street, Athens, Georgia 30602, United States
| | - Lohitash Karumbaiah
- Regenerative Bioscience Center, ADS Complex, University of Georgia, 422 River Road, Athens, Georgia 30602, United States
| | - Geert-Jan Boons
- Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, Georgia 30602, United States
- Department of Chemistry, University of Georgia, 140 Cedar Street, Athens, Georgia 30602, United States
- Department of Chemical Biology and Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, and Bijvoet Center for Biomolecular Research, Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| |
Collapse
|
47
|
d'Angelo M, Benedetti E, Tupone MG, Catanesi M, Castelli V, Antonosante A, Cimini A. The Role of Stiffness in Cell Reprogramming: A Potential Role for Biomaterials in Inducing Tissue Regeneration. Cells 2019; 8:E1036. [PMID: 31491966 PMCID: PMC6770247 DOI: 10.3390/cells8091036] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/30/2019] [Accepted: 09/04/2019] [Indexed: 01/12/2023] Open
Abstract
The mechanotransduction is the process by which cells sense mechanical stimuli such as elasticity, viscosity, and nanotopography of extracellular matrix and translate them into biochemical signals. The mechanotransduction regulates several aspects of the cell behavior, including migration, proliferation, and differentiation in a time-dependent manner. Several reports have indicated that cell behavior and fate are not transmitted by a single signal, but rather by an intricate network of many signals operating on different length and timescales that determine cell fate. Since cell biology and biomaterial technology are fundamentals in cell-based regenerative therapies, comprehending the interaction between cells and biomaterials may allow the design of new biomaterials for clinical therapeutic applications in tissue regeneration. In this work, we present the most relevant mechanism by which the biomechanical properties of extracellular matrix (ECM) influence cell reprogramming, with particular attention on the new technologies and materials engineering, in which are taken into account not only the biochemical and biophysical signals patterns but also the factor time.
Collapse
Affiliation(s)
- Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Maria Grazia Tupone
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| |
Collapse
|
48
|
Pennacchio FA, Caliendo F, Iaccarino G, Langella A, Siciliano V, Santoro F. Three-dimensionally Patterned Scaffolds Modulate the Biointerface at the Nanoscale. NANO LETTERS 2019; 19:5118-5123. [PMID: 31268343 DOI: 10.1021/acs.nanolett.9b01468] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The main aim of cell instructive materials is to guide in a controlled way cellular behavior by fine-tuning cell-material crosstalk. In the last decades, several efforts have been spent in elucidating the relations between material cues and cellular fate at the nanoscale and in the development of novel strategies for gaining a superior control over cellular function modulation. In this context, a particular attention has been recently paid to the role played by cellular membrane rearrangement in triggering specific molecular pathways linked to the regulation of different cellular functions. Here, we characterize the effect of linear microtopographies upon cellular behavior in three-dimensional (3D) environments, with particular focus on the relations linking cytoskeleton structuration to membrane rearrangement and internalization tuning. The performed analysis shown that, by altering the cellular adhesion processes at the micro- and nanoscale, it is possible to alter the membrane physical state and cellular internalization capability. More specifically, our findings pointed out that an increased cytoskeletal structuration influences the formation of nanoinvagination membrane process at the cell-material interface and the expression of clathrin and caveolin, two of the main proteins involved in the endocytosis regulation. Moreover, we proved that such topographies enhance the engulfment of inert polystyrene nanoparticles attached on 3D patterned surfaces. Our results could give new guidelines for the design of innovative and more efficient 3D cell culture systems usable for diagnostic, therapeutic, and tissue engineering purposes.
Collapse
Affiliation(s)
- Fabrizio A Pennacchio
- Center for Advanced Biomaterials for Healthcare , Istituto Italiano di Tecnologia , 80125 Naples , Italy
| | - Fabio Caliendo
- Center for Advanced Biomaterials for Healthcare , Istituto Italiano di Tecnologia , 80125 Naples , Italy
| | - Giulia Iaccarino
- Center for Advanced Biomaterials for Healthcare , Istituto Italiano di Tecnologia , 80125 Naples , Italy
| | - Angela Langella
- Center for Advanced Biomaterials for Healthcare , Istituto Italiano di Tecnologia , 80125 Naples , Italy
| | - Velia Siciliano
- Center for Advanced Biomaterials for Healthcare , Istituto Italiano di Tecnologia , 80125 Naples , Italy
| | - Francesca Santoro
- Center for Advanced Biomaterials for Healthcare , Istituto Italiano di Tecnologia , 80125 Naples , Italy
| |
Collapse
|
49
|
Lim MS, Ko SH, Kim MS, Lee B, Jung HS, Kim K, Park CH. Hybrid Nanofiber Scaffold-Based Direct Conversion of Neural Precursor Cells/Dopamine Neurons. Int J Stem Cells 2019; 12:340-346. [PMID: 31023000 PMCID: PMC6657951 DOI: 10.15283/ijsc18123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/18/2019] [Accepted: 03/10/2019] [Indexed: 12/22/2022] Open
Abstract
The concept of cellular reprogramming was developed to generate induced neural precursor cells (iNPCs)/dopaminergic (iDA) neurons using diverse approaches. Here, we investigated the effects of various nanoscale scaffolds (fiber, dot, and line) on iNPC/iDA differentiation by direct reprogramming. The generation and maturation of iDA neurons (microtubule-associated protein 2-positive and tyrosine hydroxylase-positive) and iNPCs (NESTIN-positive and SOX2-positive) increased on fiber and dot scaffolds as compared to that of the flat (control) scaffold. This study demonstrates that nanotopographical environments are suitable for direct differentiation methods and may improve the differentiation efficiency.
Collapse
Affiliation(s)
- Mi-Sun Lim
- Research and Development Center, Jeil Pharmaceutical Company, Yongin, Korea
| | - Seung Hwan Ko
- Graduate School of Biomedical Science & Engineering, Hanyang University, Seoul, Korea
| | - Min Sung Kim
- School of Mechanical & Aerospace Engineering, Seoul National University, Seoul, Korea
| | - Byungjun Lee
- School of Mechanical & Aerospace Engineering, Seoul National University, Seoul, Korea
| | - Ho-Sup Jung
- Center for Food and Bioconvergence, Department of Food Science and Biotechnology, Seoul National University, Seoul, Korea
| | - Keesung Kim
- Research Institute of Advanced Materials, Seoul National University, Seoul, Korea
| | - Chang-Hwan Park
- Graduate School of Biomedical Science & Engineering, Hanyang University, Seoul, Korea.,Department of Microbiology, College of Medicine, Hanyang University, Seoul, Korea
| |
Collapse
|
50
|
Ventre M, Coppola V, Natale CF, Netti PA. Aligned fibrous decellularized cell derived matrices for mesenchymal stem cell amplification. J Biomed Mater Res A 2019; 107:2536-2546. [PMID: 31325203 DOI: 10.1002/jbm.a.36759] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Accepted: 07/15/2019] [Indexed: 01/08/2023]
Abstract
Biochemical and biophysical stimuli of stem cell niches finely regulate the self-renewal/differentiation equilibrium. Replicating this in vitro is technically challenging, making the control of stem cell functions difficult. Cell derived matrices capture certain aspect of niches that influence fate decisions. Here, aligned fibrous matrices synthesized by MC3T3 cells were produced and the role of matrix orientation and stiffness on the maintenance of stem cell characteristics and adipo- or osteo-genic differentiation of murine mesenchymal stem cells (mMSCs) was investigated. Decellularized matrices promoted mMSC proliferation. Fibrillar alignment and matrix stiffness work in concert in defining cell fate. Soft matrices preserve stemness, whereas stiff ones, in presence of biochemical supplements, promptly induce differentiation. Matrix alignment impacts the homogeneity of the cell population, that is, soft aligned matrices ameliorate the spontaneous adipogenic differentiation, whereas stiff aligned matrices reduce cross-differentiation. We infer that mechanical signaling is a dominant factor in mMSC fate decision and the matrix alignment contributes to produce a more homogeneous environment, which results in a uniform response of cells to biophysical environment. Matrix thus produced can be obtained in vitro in a facile and consistent manner and can be used for homogeneous stem cell amplification or for mechanotransduction-related studies.
Collapse
Affiliation(s)
- Maurizio Ventre
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy.,Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy.,Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| | - Valerio Coppola
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy
| | - Carlo F Natale
- Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy
| | - Paolo A Netti
- Department of Chemical, Materials and Industrial Production Engineering, University of Naples Federico II, Naples, Italy.,Interdisciplinary Research Centre on Biomaterials, University of Naples Federico II, Naples, Italy.,Center for Advanced Biomaterials for Healthcare@CRIB, Istituto Italiano di Tecnologia, Naples, Italy
| |
Collapse
|