1
|
Adhikari B, Barakoti P, Pantcheva MB, Krebs MD. 3D printed gelatin methacryloyl hydrogels for perfusion culture of human trabecular meshwork cells and glaucoma studies. Biotechnol Bioeng 2025; 122:69-79. [PMID: 39291858 DOI: 10.1002/bit.28849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
Glaucoma, a progressive eye disease leading to irreversible blindness, currently affects over 70 million people globally. Elevated intraocular pressure (IOP) is implicated in its development. IOP is carefully regulated by the trabecular meshwork (TM). However, studying TM behavior has been limited to traditional tissue culture studies or costly ex vivo cultures of animal and donor eyes. Developing novel functional TM models could enhance cell/tissue behavior understanding and aid therapeutic development for glaucoma. In this study, we 3D printed a simplified and reproducible model of the human TM (hTM) and studied hTM cell behavior under static and dynamic cultures. Gelatin Methacryloyl bioinks proved suitable for printing with viable and proliferative hTM cells expressing crucial marker genes in response to glucocorticoid induction. This, to our knowledge, is the first functional 3D printed hTM model and aims to facilitate TM research. Moreover, this easily reproducible model could also be applicable in the study of numerous other cell types throughout the body.
Collapse
Affiliation(s)
- Bikram Adhikari
- Colorado School of Mines, Quantitative Biosciences and Engineering, Golden, Colorado, USA
| | - Prasanga Barakoti
- Colorado School of Mines, Quantitative Biosciences and Engineering, Golden, Colorado, USA
| | - Mina B Pantcheva
- Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Melissa D Krebs
- Colorado School of Mines, Quantitative Biosciences and Engineering, Golden, Colorado, USA
- Colorado School of Mines, Chemical and Biological Engineering, Golden, Colorado, USA
| |
Collapse
|
2
|
Lu H, Li Z, Zhu L, Xu P, Wang H, Li Y, Zhao W. Fabrication and Temporal Dependency Osteogenic Regulation of Dual-Scale Hierarchical Microstructures on Medical Metal Surface. Adv Healthc Mater 2024; 13:e2402369. [PMID: 39175381 DOI: 10.1002/adhm.202402369] [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: 07/13/2024] [Revised: 08/09/2024] [Indexed: 08/24/2024]
Abstract
The structural characteristics at the interface of bone implants can guide biological regulation. In this study, a dual-scale hierarchical microstructure is proposed and customized using hybrid machining to achieve temporal dependency osteogenic regulation. It is observed that osteoblasts induced by dual-scale hierarchical structure exhibit adequate protrusion development and rapid cell attachment through the modulation of mechanical forces in the cell growth environment, and further promot the upregulation of the cell membrane receptor PDGFR-α, which is related to cell proliferation. Afterward, transcriptomic analysis reveals that during the differentiation stage, the DSH structure regulates cellular signaling cascades primarily through integrin adhesion mechanisms and then accelerates osteogenic differentiation by activating the TGF-β pathway and cAMP signaling pathway. Furthermore, the calcium nodules are preferentially deposited within the lower honeycomb-like channels, thereby endowing the proposed dual-scale hierarchical structure with the potential to induce oriented deposition and improve the long-term stability of the implant.
Collapse
Affiliation(s)
- Hao Lu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Zhijun Li
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Lida Zhu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Peihua Xu
- School of Mechanical Engineering and Automation, Northeastern University, Shenyang, 110819, China
| | - Hai Wang
- Shenyang Lebuy Vacuum Tech. Co., Ltd, Shenyang, Liaoning, China
| | - Yonghao Li
- Shenyang Lebuy Vacuum Tech. Co., Ltd, Shenyang, Liaoning, China
| | - Weidong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, and Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
3
|
Shuai Y, Zheng M, Kundu SC, Mao C, Yang M. Bioengineered Silk Protein-Based 3D In Vitro Models for Tissue Engineering and Drug Development: From Silk Matrix Properties to Biomedical Applications. Adv Healthc Mater 2024; 13:e2401458. [PMID: 39009465 DOI: 10.1002/adhm.202401458] [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: 04/21/2024] [Revised: 06/22/2024] [Indexed: 07/17/2024]
Abstract
3D in vitro model has emerged as a valuable tool for studying tissue development, drug screening, and disease modeling. 3D systems can accurately replicate tissue microstructures and physiological features, mirroring the in vivo microenvironment departing from conventional 2D cell cultures. Various 3D in vitro models utilizing biomacromolecules like collagen and synthetic polymers have been developed to meet diverse research needs and address the complex challenges of contemporary research. Silk proteins, bearing structural and functional similarities to collagen, have been increasingly employed to construct advanced 3D in vitro systems, surpassing the limitations of 2D cultures. This review examines silk proteins' composition, structure, properties, and functions, elucidating their role in 3D in vitro models. Furthermore, recent advances in biomedical applications involving silk-based organoid models are discussed. In particular, the unique physiological attributes of silk matrix constituents in in vitro tissue constructs are highlighted, providing a meticulous evaluation of their importance. Additionally, it outlines the current research hurdles and complexities while contemplating future avenues, thereby paving the way for developing complex and biomimetic silk protein-based microtissues.
Collapse
Affiliation(s)
- Yajun Shuai
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Meidan Zheng
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Subhas C Kundu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, P. R. China
| | - Mingying Yang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
4
|
Mai-Thi HN, Nguyen DPH, Le P, Tran NQ, Tran CT, Stoldt VR, Huynh K. Low shear-induced fibrillar fibronectin: comparative analyses of morphologies and cellular effects on bovine aortic endothelial cell adhesion and proliferation. Phys Biol 2024; 21:066002. [PMID: 39366409 DOI: 10.1088/1478-3975/ad838c] [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: 04/12/2024] [Accepted: 10/04/2024] [Indexed: 10/06/2024]
Abstract
Wall shear stress (WSS) is a critical factor in vascular biology, and both high and low WSS are implicated in atherosclerosis. Fibronectin (FN) is a key extracellular matrix protein that plays an important role in cell activities. Under high shear stress, plasma FN undergoes fibrillogenesis; however, its behavior under low shear stress remains unclear. This study aimed to investigate the formation ofin vitrocell-free fibrillar FN (FFN) under low shear rate conditions and its effect on bovine aortic endothelial cell behavior. FN (500µg ml-1) was perfused through slide chambers at three flow rates (0.16 ml h-1, 0.25 ml h-1, and 0.48 ml h-1), corresponding to low shear rates of 0.35 s-1, 0.55 s-1, and 1.05 s-1, respectively, for 4 h at room temperature. The formed FN matrices were observed using fluorescence microscopy and scanning electron microscopy. Under low shear rates, distinct FN matrix structures were observed. FFN0.48 formed immense fibrils with smooth surfaces, FFN0.25 formed a matrix with a rough surface, and FFN16 exhibited nodular structures. FFN0.25 supported cell activities to a greater extent than native FN and other FFN surfaces. Our study suggests that abnormally low shear conditions impact FN structure and function and enhance the understanding of FN fibrillogenesis in vascular biology, particularly in atherosclerosis.
Collapse
Affiliation(s)
- Hoang-Nghi Mai-Thi
- School of Biomedical Engineering, International University, Quarter 6, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- Vietnam National University, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- School of Nursing, Eastern International University, Nam Ky Khoi Nghia Street, Hoa Phu Ward, Thu Dau Mot City, Binh Duong Province, Vietnam
| | - Dang Phu-Hai Nguyen
- School of Biomedical Engineering, International University, Quarter 6, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- School of Biotechnology, International University, Quarter 6, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- Vietnam National University, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
| | - Phong Le
- School of Biomedical Engineering, International University, Quarter 6, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- Vietnam National University, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
| | - Ngoc Quyen Tran
- Graduate University of Science and Technology Viet Nam, Vietnam Academy of Science and Technology, Ho Chi Minh City, Viet Nam; and Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Cam Tu Tran
- Institute of Tropical Biology, Viet Nam Academy of Science and Technology (VAST), Ho Chi Minh City, Vietnam
| | - Volker R Stoldt
- Department of General, Visceral and Pediatric Surgery, University Hospital and Medical Faculty of the Heinrich-Heine University Düsseldorf, Moorenstr. 5, Düsseldorf 40225, Germany
| | - Khon Huynh
- School of Biomedical Engineering, International University, Quarter 6, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
- Vietnam National University, Linh Trung, Thu Duc District, Ho Chi Minh City, Vietnam
| |
Collapse
|
5
|
Dadafarin H, Konkov E, Vali H, Ali I, Omanovic S. Modification of 316L Stainless Steel, Nickel Titanium, and Cobalt Chromium Surfaces by Irreversible Immobilization of Fibronectin: Towards Improving the Coronary Stent Biocompatibility. Molecules 2024; 29:4927. [PMID: 39459295 PMCID: PMC11510294 DOI: 10.3390/molecules29204927] [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: 09/23/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
An extracellular matrix protein, fibronectin (Fn), was covalently immobilized on 316L stainless steel, L605 cobalt chromium (CoCr), and nickel titanium (NiTi) surfaces through an 11-mercaptoundecanoic acid (MUA) self-assembled monolayer (SAM) pre-formed on these surfaces. Polarization modulation infrared reflection adsorption spectroscopy (PM-IRRAS) confirmed the presence of Fn on the surfaces. The Fn monolayer attached to the SAM was found to be stable under fluid shear stress. Deconvolution of the Fn amide I band indicated that the secondary structure of Fn changes significantly upon immobilization to the SAM-functionalized metal substrate. Scanning electron microscopy and energy dispersive X-ray analysis revealed that the spacing between Fn molecules on a modified commercial stent surface is approximately 66 nm, which has been reported to be the most appropriate spacing for cell/surface interactions.
Collapse
Affiliation(s)
- Hesam Dadafarin
- Department of Chemical Engineering, McGill University, 3610 University St., Montreal, QC H3A 0C5, Canada; (H.D.); (E.K.)
| | - Evgeny Konkov
- Department of Chemical Engineering, McGill University, 3610 University St., Montreal, QC H3A 0C5, Canada; (H.D.); (E.K.)
| | - Hojatollah Vali
- Department of Anatomy and Cell Biology, McGill University, 3640 University St., Montreal, QC H3A 0C7, Canada;
| | - Irshad Ali
- Department of Chemical Engineering, Faculty of Mechanical, Chemical and Industrial Engineering, University of Engineering & Technology, Jamrud Road, Peshawar 25000, Pakistan;
| | - Sasha Omanovic
- Department of Chemical Engineering, McGill University, 3610 University St., Montreal, QC H3A 0C5, Canada; (H.D.); (E.K.)
| |
Collapse
|
6
|
Cheong S, Peng Y, Lu F, He Y. Structural extracellular matrix-mediated molecular signaling in wound repair and tissue regeneration. Biochimie 2024:S0300-9084(24)00230-X. [PMID: 39369941 DOI: 10.1016/j.biochi.2024.10.003] [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: 05/04/2024] [Revised: 09/19/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
The extracellular matrix (ECM) is a complex, non-cellular network of molecules that offers structural support for cells and tissues. The ECM is composed of various structural components, including collagen, fibronectin, laminin, perlecan, nidogen, tenascin, and fibulin, which are capable of binding to each other and to cell-to-adhesion receptors, endowing the ECM with unique physical and biochemical properties that are essential for its function in maintaining health and managing disease. Over the past three decades, extensive research has shown that the core of the ECM can significantly impact cellular events at the molecular level. Structural modifications have also been strongly associated with tissue repair. Through interactions with cells, matrix proteins regulate critical processes such as cell proliferation and differentiation, migration, and apoptosis, essential for maintaining tissue homeostasis, formation, and regeneration. This review emphasizes the interlocking networks of ECM macromolecules and their primary roles in tissue regeneration and wound repair. Through studying ECM dynamics, researchers have discovered molecular signaling pathways that demonstrate how the ECM influences protein patterns and open up more possibilities for developing therapeutics that target the ECM to enhance wound repair and tissue regeneration.
Collapse
Affiliation(s)
- Sousan Cheong
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| | - Yujie Peng
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| | - Feng Lu
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| | - Yunfan He
- The Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, 510515, China.
| |
Collapse
|
7
|
Filiz Y, Esposito A, De Maria C, Vozzi G, Yesil-Celiktas O. A comprehensive review on organ-on-chips as powerful preclinical models to study tissue barriers. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2024; 6:042001. [PMID: 39655848 DOI: 10.1088/2516-1091/ad776c] [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: 10/27/2023] [Accepted: 09/04/2024] [Indexed: 12/18/2024]
Abstract
In the preclinical stage of drug development, 2D and 3D cell cultures under static conditions followed by animal models are utilized. However, these models are insufficient to recapitulate the complexity of human physiology. With the developing organ-on-chip (OoC) technology in recent years, human physiology and pathophysiology can be modeled better than traditional models. In this review, the need for OoC platforms is discussed and evaluated from both biological and engineering perspectives. The cellular and extracellular matrix components are discussed from a biological perspective, whereas the technical aspects such as the intricate working principles of these systems, the pivotal role played by flow dynamics and sensor integration within OoCs are elucidated from an engineering perspective. Combining these two perspectives, bioengineering applications are critically discussed with a focus on tissue barriers such as blood-brain barrier, ocular barrier, nasal barrier, pulmonary barrier and gastrointestinal barrier, featuring recent examples from the literature. Furthermore, this review offers insights into the practical utility of OoC platforms for modeling tissue barriers, showcasing their potential and drawbacks while providing future projections for innovative technologies.
Collapse
Affiliation(s)
- Yagmur Filiz
- Department of Development and Regeneration, Faculty of Medicine, KU Leuven, 8500 Kortrijk, Belgium
| | - Alessio Esposito
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Carmelo De Maria
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Giovanni Vozzi
- Research Center E. Piaggio and Department of Information Engineering, University of Pisa, Largo L. Lazzarino 1, Pisa 56126, Italy
| | - Ozlem Yesil-Celiktas
- Department of Bioengineering, Faculty of Engineering, Ege University, 35100 Izmir, Turkey
- EgeSAM-Ege University Translational Pulmonary Research Center, Bornova, Izmir, Turkey
- ODTÜ MEMS Center, Ankara, Turkey
| |
Collapse
|
8
|
Di Francesco D, Marcello E, Casarella S, Copes F, Chevallier P, Carmagnola I, Mantovani D, Boccafoschi F. Characterization of a decellularized pericardium extracellular matrix hydrogel for regenerative medicine: insights on animal-to-animal variability. Front Bioeng Biotechnol 2024; 12:1452965. [PMID: 39205858 PMCID: PMC11350490 DOI: 10.3389/fbioe.2024.1452965] [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: 06/21/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
In the past years, the use of hydrogels derived from decellularized extracellular matrix (dECM) for regenerative medicine purposes has significantly increased. The intrinsic bioactive and immunomodulatory properties indicate these materials as promising candidates for therapeutical applications. However, to date, limitations such as animal-to-animal variability still hinder the clinical translation. Moreover, the choice of tissue source, decellularization and solubilization protocols leads to differences in dECM-derived hydrogels. In this context, detailed characterization of chemical, physical and biological properties of the hydrogels should be performed, with attention to how these properties can be affected by animal-to-animal variability. Herein, we report a detailed characterization of a hydrogel derived from the decellularized extracellular matrix of bovine pericardium (dBP). Protein content, rheological properties, injectability, surface microstructure, in vitro stability and cytocompatibility were evaluated, with particular attention to animal-to-animal variability. The gelation process showed to be thermoresponsive and the obtained dBP hydrogels are injectable, porous, stable up to 2 weeks in aqueous media, rapidly degrading in enzymatic environment and cytocompatible, able to maintain cell viability in human mesenchymal stromal cells. Results from proteomic analysis proved that dBP hydrogels are highly rich in composition, preserving bioactive proteoglycans and glycoproteins in addition to structural proteins such as collagen. With respect to the chemical composition, animal-to-animal variability was shown, but the biological properties were not affected, which remained consistent in different batches. Taken together these results show that dBP hydrogels are excellent candidates for regenerative medicine applications.
Collapse
Affiliation(s)
- Dalila Di Francesco
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair Tier I for the Innovation in Surgery, Department of Min-Met-Materials Engineering and Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec, QC, Canada
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Elena Marcello
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Torino, Italy
- Polito BioMed Lab, Politecnico di Torino, Torino, Italy
| | - Simona Casarella
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
| | - Francesco Copes
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair Tier I for the Innovation in Surgery, Department of Min-Met-Materials Engineering and Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec, QC, Canada
| | - Pascale Chevallier
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair Tier I for the Innovation in Surgery, Department of Min-Met-Materials Engineering and Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec, QC, Canada
| | - Irene Carmagnola
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Torino, Italy
- Polito BioMed Lab, Politecnico di Torino, Torino, Italy
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, Canada Research Chair Tier I for the Innovation in Surgery, Department of Min-Met-Materials Engineering and Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec, QC, Canada
| | - Francesca Boccafoschi
- Department of Health Sciences, University of Piemonte Orientale “A. Avogadro”, Novara, Italy
| |
Collapse
|
9
|
Zumbo B, Guagnini B, Medagli B, Porrelli D, Turco G. Fibronectin Functionalization: A Way to Enhance Dynamic Cell Culture on Alginate/Hydroxyapatite Scaffolds. J Funct Biomater 2024; 15:222. [PMID: 39194660 DOI: 10.3390/jfb15080222] [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: 07/08/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
Bone defects are a global health concern; bone tissue engineering (BTE) is the most promising alternative to reduce patient morbidity and overcome the inherent drawbacks of autograft and allograft bone. Three-dimensional scaffolds are pivotal in this field due to their potential to provide structural support and mimic the natural bone microenvironment. Following an already published protocol, a 3D porous structure consisting of alginate and hydroxyapatite was prepared after a gelation step and a freezing-drying step. Despite the frequent use of alginate in tissue regeneration, the biological inertness of this polysaccharide hampers proper cell colonization and proliferation. Therefore, the purpose of this work was to enhance the biological properties by promoting the interaction and adhesion between cells and biomaterial with the use of Fibronectin. This extracellular matrix protein was physically adsorbed on the scaffold, and its presence was evaluated with environmental scanning electron microscopy (eSEM) and the Micro-Bicinchoninic Acid (μBCA) protein assay. The MG-63 cell line was used for both static and dynamic (i.e., in bioreactor) 3D cell culturing on the scaffolds. The use of the bioreactor allowed for a better exchange of nutrients and oxygen and a better removal of cell catabolites from the inner portion of the construct, mimicking the physiological environment. The functionalized scaffolds showed an improvement in cell proliferation and colonization compared to non-functionalized ones; the effect of the addition of Fibronectin was more evident in the dynamic culturing conditions, where the cells clearly adhered on the surface of functionalized scaffolds.
Collapse
Affiliation(s)
- Bianca Zumbo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34129 Trieste, Italy
| | - Benedetta Guagnini
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34129 Trieste, Italy
| | - Barbara Medagli
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34129 Trieste, Italy
| | - Davide Porrelli
- Department of Life Sciences, University of Trieste, Via Alexander Fleming 31/B, 34127 Trieste, Italy
| | - Gianluca Turco
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Piazza dell'Ospitale 1, 34129 Trieste, Italy
| |
Collapse
|
10
|
Xu Y, Liu X, Ahmad MA, Ao Q, Yu Y, Shao D, Yu T. Engineering cell-derived extracellular matrix for peripheral nerve regeneration. Mater Today Bio 2024; 27:101125. [PMID: 38979129 PMCID: PMC11228803 DOI: 10.1016/j.mtbio.2024.101125] [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: 03/05/2024] [Revised: 05/28/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
Extracellular matrices (ECMs) play a key role in nerve repair and are recognized as the natural source of biomaterials. In parallel to extensively studied tissue-derived ECMs (ts-ECMs), cell-derived ECMs (cd-ECMs) also have the capability to partially recapitulate the complicated regenerative microenvironment of native nerve tissues. Notably, cd-ECMs can avoid the shortcomings of ts-ECMs. Cd-ECMs can be prepared by culturing various cells or even autologous cells in vitro under pathogen-free conditions. And mild decellularization can achieve efficient removal of immunogenic components in cd-ECMs. Moreover, cd-ECMs are more readily customizable to achieve the desired functional properties. These advantages have garnered significant attention for the potential of cd-ECMs in neuroregenerative medicine. As promising biomaterials, cd-ECMs bring new hope for the effective treatment of peripheral nerve injuries. Herein, this review comprehensively examines current knowledge about the functional characteristics of cd-ECMs and their mechanisms of interaction with cells in nerve regeneration, with a particular focus on the preparation, engineering optimization, and scalability of cd-ECMs. The applications of cd-ECMs from distinct cell sources reported in peripheral nerve tissue engineering are highlighted and summarized. Furthermore, current limitations that should be addressed and outlooks related to clinical translation are put forward as well.
Collapse
Affiliation(s)
- Yingxi Xu
- Department of Clinical Nutrition, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xianbo Liu
- Department of Orthodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | | | - Qiang Ao
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, Institute of Regulatory Science for Medical Device, National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, China
| | - Yang Yu
- Health Sciences Institute, Key Laboratory of Obesity and Glucose/Lipid Associated Metabolic Diseases, China Medical University, Shenyang, China
| | - Dan Shao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangdong, Guangzhou, China
| | - Tianhao Yu
- The VIP Department, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| |
Collapse
|
11
|
Popescu F, Titorencu I, Albu Kaya M, Miculescu F, Tutuianu R, Coman AE, Danila E, Marin MM, Ancuta DL, Coman C, Barbilian A. Development of Innovative Biocomposites with Collagen, Keratin and Hydroxyapatite for Bone Tissue Engineering. Biomimetics (Basel) 2024; 9:428. [PMID: 39056869 PMCID: PMC11275084 DOI: 10.3390/biomimetics9070428] [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: 05/21/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
This study follows the process for the development of an innovative biomimetic composite derived from bovine collagen with keratin, with hydroxyapatite being hybridized into its architecture, and it builds a comprehensive evaluation of the composite's characteristics. The novel biomimetic materials are tailored with special traits to be achieved for the repair of osteochondral defects (OCDs). The purpose of the present research is to create a reliable effective alternative to existing bone graft materials while leveraging the intrinsic properties of the components for enhanced osteoinduction and integration. The composites were characterized based on their morphological properties, including water absorption, through scanning electron microscopy (SEM), and their structural properties were characterized by Fourier-Transform Infrared Spectroscopy (FTIR). Biological performance was assessed in vitro using human bone marrow mesenchymal stem cells (BMSCs), focusing on cytotoxicity, cell viability, and the ability to support cell colonization with forthcoming results. This in vivo study illustrates the real potential that this class of novel composites exhibits in regard to bone and cartilage tissue engineering and encourages further exploration and development for future clinical applications.
Collapse
Affiliation(s)
- Florin Popescu
- Department of Orthopedics and Traumatology, Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 8 Eroii Sanitari Bvd., 050474 Bucharest, Romania; (F.P.); (A.B.)
| | - Irina Titorencu
- Institute of Cellular Biology and Pathology ‘’Nicolae Simionescu’’, 8 B. P. Hasdeu Street, District 5, 050568 Bucharest, Romania; (I.T.); (R.T.)
| | - Madalina Albu Kaya
- INCDTP—Division Leather and Footwear Research Institute, 93 Ion Minulescu Str., 031215 Bucharest, Romania;
| | - Florin Miculescu
- Department of Metallic Materials Science, Physical Metallurgy, National University of Science and Technology Politehnica Bucharest, 313 Independenței Spl., 060042 Bucharest, Romania;
| | - Raluca Tutuianu
- Institute of Cellular Biology and Pathology ‘’Nicolae Simionescu’’, 8 B. P. Hasdeu Street, District 5, 050568 Bucharest, Romania; (I.T.); (R.T.)
| | - Alina Elena Coman
- INCDTP—Division Leather and Footwear Research Institute, 93 Ion Minulescu Str., 031215 Bucharest, Romania;
| | - Elena Danila
- INCDTP—Division Leather and Footwear Research Institute, 93 Ion Minulescu Str., 031215 Bucharest, Romania;
| | - Minodora Maria Marin
- Advanced Polymer Materials Group, National University of Science and Technology Politehnica Bucharest, 1-7 Polizu Street, 011061 Bucharest, Romania;
| | - Diana-Larisa Ancuta
- “Cantacuzino” National Medical-Military Institute for Research and Development, 103 Independenței Spl., 050096 Bucharest, Romania; (D.-L.A.); (C.C.)
| | - Cristin Coman
- “Cantacuzino” National Medical-Military Institute for Research and Development, 103 Independenței Spl., 050096 Bucharest, Romania; (D.-L.A.); (C.C.)
| | - Adrian Barbilian
- Department of Orthopedics and Traumatology, Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 8 Eroii Sanitari Bvd., 050474 Bucharest, Romania; (F.P.); (A.B.)
| |
Collapse
|
12
|
Rima M, Villeneuve-Faure C, Soumbo M, El Garah F, Pilloux L, Roques C, Makasheva K. Towards a better understanding of the effect of protein conditioning layers on microbial adhesion: a focused investigation of fibronectin and bovine serum albumin layers on SiO 2 surfaces. Biomater Sci 2024; 12:3086-3099. [PMID: 38716803 DOI: 10.1039/d4bm00099d] [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: 06/12/2024]
Abstract
The interaction of foreign implants with their surrounding environment is significantly influenced by the adsorption of proteins on the biomaterial surfaces, playing a role in microbial adhesion. Therefore, understanding protein adsorption on solid surfaces and its effect on microbial adhesion is essential to assess the associated risk of infection. The aim of this study is to evaluate the effect of conditioning by fibronectin (Fn) or bovine serum albumin (BSA) protein layers of silica (SiO2) surfaces on the adhesion and detachment of two pathogenic microorganisms: Pseudomonas aeruginosa PAO1-Tn7-gfp and Candida albicans CIP 48.72. Experiments are conducted under both static and hydrodynamic conditions using a shear stress flow chamber. Through the use of very low wall shear stresses, the study brings the link between the static and dynamic conditions of microbial adhesion. The results reveal that the microbial adhesion critically depends on: (i) the presence of a protein layer conditioning the SiO2 surface, (ii) the type of protein and (iii) the protein conformation and organization in the conditioning layer. In addition, a very distinct adhesion behaviour of P. aeruginosa is observed towards the two tested proteins, Fn and BSA. This effect is reinforced by the amount of proteins adsorbed on the surface and their organization in the layer. The results are discussed in the light of atomic force microscopy analysis of the organization and conformation of proteins in the layers after adsorption on the SiO2 surface, as well as the specificity in bacterial behaviour when interacting with these protein layers. The study also demonstrates the very distinctive behaviours of the prokaryote P. aeruginosa PAO1-Tn7-gfp compared to the eukaryote C. albicans CIP 48.72. This underscores the importance of considering species-specific interactions between the protein conditioning layer and different pathogenic microorganisms, which appear crucial in designing tailored anti-adhesive surfaces.
Collapse
Affiliation(s)
- Maya Rima
- LGC, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| | | | - Marvine Soumbo
- LGC, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
- LAPLACE, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| | - Fatima El Garah
- LGC, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| | - Ludovic Pilloux
- LGC, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| | - Christine Roques
- LGC, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| | - Kremena Makasheva
- LAPLACE, University of Toulouse, CNRS, UTIII, INPT, Toulouse, France.
| |
Collapse
|
13
|
Ghezzi B, Meglioli M, Salvaterra Toffoli A, Mergoni G, Rossi F, Manfredi M, Lumetti S, Manfredi E. Polishing methods for composites restoration: the influence on human gingival fibroblasts behaviour. BMC Oral Health 2024; 24:651. [PMID: 38831398 PMCID: PMC11149303 DOI: 10.1186/s12903-024-04418-z] [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: 02/23/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024] Open
Abstract
BACKGROUND Carious/Non-carious cervical lesions with gingival recessions may require both dental and periodontal reconstructive therapy, where flaps/grafts may be placed in contact with a dental filling material. Human Gingival Fibroblasts (HGF-1) response during the early phase of healing could vary according to the procedures employed to cure the dental composite. Moreover, oxygen diffusion into dental composite inhibits the polymerization reaction, creating an oxygen-inhibited layer (OIL) that presents residual unreacted monomers. The aim of this study was to assess the effect of different polishing techniques and OIL on HGF-1. METHODS Composite discs polished with different techniques (diamond rubber, abrasive discs and tungsten carbide burr) were used. An additional not polished smooth group obtained with and without OIL was used as control. Samples were physically characterized through the analysis of their hydrophilicity and surface topography through contact angle measurement and SEM, respectively; afterwards the biologic response of HGF-1 when cultured on the different substrates was analyzed in terms of cytotoxicity and gene expression. RESULTS The finishing systems caused alterations to the wettability, even if without a proportional relation towards the results of the proliferation essay, from which emerges a greater proliferation on surfaces polished with one-step diamond rubber and with abrasive discs as well as a direct effect of the glycerin layer, confirming that surface roughness can heavily influence the biological response of HGF-1. CONCLUSIONS Surfaces wettability as well as cellular behavior seem to be affected by the selection of the finishing system used to lastly shape the restoration. Especially, the presence of OIL act as a negative factor in the regards of human gingival fibroblasts. The present study may provide the first clinical instruction regarding the best polishing system of composite material when the restoration is placed directly in contact with soft tissue cells. Understanding HGF-1 behavior can help identifying the polishing treatment for direct restoration of carious/non-carious cervical lesions associated with gingival recessions.
Collapse
Affiliation(s)
- Benedetta Ghezzi
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy.
- IMEM-CNR, Institute of Materials for Electronics and Magnetism, National Research Council, Parco Area delle Scienze 37/A, Parma, 43124, Italy.
| | - Matteo Meglioli
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Andrea Salvaterra Toffoli
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Giovanni Mergoni
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Francesca Rossi
- IMEM-CNR, Institute of Materials for Electronics and Magnetism, National Research Council, Parco Area delle Scienze 37/A, Parma, 43124, Italy
| | - Maddalena Manfredi
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| | - Simone Lumetti
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
- IMEM-CNR, Institute of Materials for Electronics and Magnetism, National Research Council, Parco Area delle Scienze 37/A, Parma, 43124, Italy
| | - Edoardo Manfredi
- Department of Medicine and Surgery, Centre for Dental Medicine, University of Parma, Via Gramsci 14, Parma, 43126, Italy
| |
Collapse
|
14
|
Phuphanitcharoenkun S, Louis F, Sowa Y, Matsusaki M, Palaga T. Improving stability of human three dimensional skin equivalents using plasma surface treatment. Biotechnol Bioeng 2024; 121:1950-1960. [PMID: 38470332 DOI: 10.1002/bit.28690] [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: 10/21/2023] [Revised: 01/10/2024] [Accepted: 02/19/2024] [Indexed: 03/13/2024]
Abstract
In developing three-dimensional (3D) human skin equivalents (HSEs), preventing dermis and epidermis layer distortion due to the contraction of hydrogels by fibroblasts is a challenging issue. Previously, a fabrication method of HSEs was tested using a modified solid scaffold or a hydrogel matrix in combination with the natural polymer coated onto the tissue culture surface, but the obtained HSEs exhibited skin layer contraction and loss of the skin integrity and barrier functions. In this study, we investigated the method of HSE fabrication that enhances the stability of the skin model by using surface plasma treatment. The results showed that plasma treatment of the tissue culture surface prevented dermal layer shrinkage of HSEs, in contrast to the HSE fabrication using fibronectin coating. The HSEs from plasma-treated surface showed significantly higher transepithelial electrical resistance compared to the fibronectin-coated model. They also expressed markers of epidermal differentiation (keratin 10, keratin 14 and loricrin), epidermal tight junctions (claudin 1 and zonula occludens-1), and extracellular matrix proteins (collagen IV), and exhibited morphological characteristics of the primary human skins. Taken together, the use of plasma surface treatment significantly improves the stability of 3D HSEs with well-defined dermis and epidermis layers and enhanced skin integrity and the barrier functions.
Collapse
Affiliation(s)
- Suphanun Phuphanitcharoenkun
- Graduate Program in Biotechnology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Materials and Bio-Interfaces, Chulalongkorn University, Bangkok, Thailand
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Yoshihiro Sowa
- Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto, Japan
- Department of Plastic Surgery, Jichi Medical University, Tochigi, Japan
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Tanapat Palaga
- Center of Excellence in Materials and Bio-Interfaces, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
15
|
Benmelech S, Le T, McKay M, Nam J, Subramaniam K, Tellez D, Vlasak G, Mak M. Biophysical and biochemical aspects of immune cell-tumor microenvironment interactions. APL Bioeng 2024; 8:021502. [PMID: 38572312 PMCID: PMC10990568 DOI: 10.1063/5.0195244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/19/2024] [Indexed: 04/05/2024] Open
Abstract
The tumor microenvironment (TME), composed of and influenced by a heterogeneous set of cancer cells and an extracellular matrix, plays a crucial role in cancer progression. The biophysical aspects of the TME (namely, its architecture and mechanics) regulate interactions and spatial distributions of cancer cells and immune cells. In this review, we discuss the factors of the TME-notably, the extracellular matrix, as well as tumor and stromal cells-that contribute to a pro-tumor, immunosuppressive response. We then discuss the ways in which cells of the innate and adaptive immune systems respond to tumors from both biochemical and biophysical perspectives, with increased focus on CD8+ and CD4+ T cells. Building upon this information, we turn to immune-based antitumor interventions-specifically, recent biophysical breakthroughs aimed at improving CAR-T cell therapy.
Collapse
Affiliation(s)
- Shoham Benmelech
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Thien Le
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Maggie McKay
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Jungmin Nam
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Krupakar Subramaniam
- Department of Molecular, Cellular, and Developmental Biology, Yale University, New Haven, Connecticut 06511, USA
| | - Daniela Tellez
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Grace Vlasak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| | - Michael Mak
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut 06511, USA
| |
Collapse
|
16
|
Akhtar H, Alhamoudi FH, Marshall J, Ashton T, Darr JA, Rehman IU, Chaudhry AA, Reilly G. Synthesis of cerium, zirconium, and copper doped zinc oxide nanoparticles as potential biomaterials for tissue engineering applications. Heliyon 2024; 10:e29150. [PMID: 38601679 PMCID: PMC11004213 DOI: 10.1016/j.heliyon.2024.e29150] [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: 11/06/2023] [Revised: 04/02/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
A novel eco-friendly high throughput continuous hydrothermal flow system was used to synthesise phase pure ZnO and doped ZnO in order to explore their properties for tissue engineering applications. Cerium, zirconium, and copper were introduced as dopants during flow synthesis of ZnO nanoparticles, Zirconium doped ZnO were successfully synthesised, however secondary phases of CeO and CuO were detected in X-ray diffraction (XRD). The nanoparticles were characterised using X-ray diffraction, Brunauer-Emmett-Teller (BET), Dynamic Light scattering Measurements, Scanning Electron Microscopy (SEM), Transmission Electron Microscopy (TEM), Fourier transform infrared spectroscopy (FT-IR) and RAMAN spectroscopy was used to evaluate physical, chemical, and structural properties. The change in BET surface area was also significant, the surface area increased from 11.35 (ZnO_2) to 26.18 (ZrZnO_5). However. In case of CeZnO_5 and CuZnO_5 was not significant 13.68 (CeZnO_5) and 12.16 (CuZnO_5) respectively. Cell metabolic activity analysis using osteoblast-like cells (MG63) and human embryonic derived mesenchymal stem cells (hES-MP) demonstrated that doped ZnO nanoparticles supported higher cell metabolic activity compared to cells grown in standard media with no nanoparticles added, or pure zinc oxide nanoparticles. The ZrZnO_5 demonstrated the highest cell metabolic activity and non-cytotoxicity over the duration of 28 days as compared to un doped or Ce or Cu incorporated nanoparticles. The current data suggests that Zirconium doping positively enhances the properties of ZnO nanoparticles by increasing the surface area and cell proliferation. Therefore, are potential additives within biomaterials or for tissue engineering applications.
Collapse
Affiliation(s)
- Hafsah Akhtar
- Department of Materials Science and Engineering, Pam Liversidge Building, Mappin Street, Sheffield, United Kingdom
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS University Islamabad, Lahore Campus, Pakistan
| | - Fahad Hussain Alhamoudi
- Dental Technology Department, Applied Medical Science, King Khalid University, Abha 62529, Kingdom of Saudi Arabia
| | - Julie Marshall
- Department of Materials Science and Engineering, Pam Liversidge Building, Mappin Street, Sheffield, United Kingdom
| | | | | | - Ihtesham Ur Rehman
- Research and Enterprise, School of Medicine,University of Central Lancashire, Preston, United Kingdom
| | - Aqif Anwar Chaudhry
- Interdisciplinary Research Centre in Biomedical Materials (IRCBM), COMSATS University Islamabad, Lahore Campus, Pakistan
| | - Gwendolen Reilly
- Department of Materials Science and Engineering, Pam Liversidge Building, Mappin Street, Sheffield, United Kingdom
| |
Collapse
|
17
|
Shi T, Shah I, Dang Q, Taylor L, Jagannath A. Sex-specific regulation of the cortical transcriptome in response to sleep deprivation. Front Neurosci 2024; 17:1303727. [PMID: 38504908 PMCID: PMC10948409 DOI: 10.3389/fnins.2023.1303727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 12/07/2023] [Indexed: 03/21/2024] Open
Abstract
Multiple studies have documented sex differences in sleep behaviour, however, the molecular determinants of such differences remain unknown. Furthermore, most studies addressing molecular mechanisms have been performed only in males, leaving the current state of knowledge biased towards the male sex. To address this, we studied the differences in the transcriptome of the cerebral cortex of male and female C57Bl/6 J mice after 6 h of sleep deprivation. We found that several genes, including the neurotrophin growth factor Bdnf, immediate early genes Fosb and Fosl2, and the adenylate cyclase Adcy7 are differentially upregulated in males compared to females. We identified the androgen-receptor activating transcription factor EZH2 as the upstream regulatory element specifying sex differences in the sleep deprivation transcriptome. We propose that the pathways downstream of these transcripts, which impact on cellular re-organisation, synaptic signalling, and learning may underpin the differential response to sleep deprivation in the two sexes.
Collapse
Affiliation(s)
- Tianyi Shi
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, Oxford, United Kingdom
| | - Ishani Shah
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom
| | - Quang Dang
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, Oxford, United Kingdom
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
| | - Lewis Taylor
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, Oxford, United Kingdom
| | - Aarti Jagannath
- Sleep and Circadian Neuroscience Institute (SCNi), Nuffield Department of Clinical Neurosciences, New Biochemistry Building, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
18
|
Crossley RM, Johnson S, Tsingos E, Bell Z, Berardi M, Botticelli M, Braat QJS, Metzcar J, Ruscone M, Yin Y, Shuttleworth R. Modeling the extracellular matrix in cell migration and morphogenesis: a guide for the curious biologist. Front Cell Dev Biol 2024; 12:1354132. [PMID: 38495620 PMCID: PMC10940354 DOI: 10.3389/fcell.2024.1354132] [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: 12/11/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The extracellular matrix (ECM) is a highly complex structure through which biochemical and mechanical signals are transmitted. In processes of cell migration, the ECM also acts as a scaffold, providing structural support to cells as well as points of potential attachment. Although the ECM is a well-studied structure, its role in many biological processes remains difficult to investigate comprehensively due to its complexity and structural variation within an organism. In tandem with experiments, mathematical models are helpful in refining and testing hypotheses, generating predictions, and exploring conditions outside the scope of experiments. Such models can be combined and calibrated with in vivo and in vitro data to identify critical cell-ECM interactions that drive developmental and homeostatic processes, or the progression of diseases. In this review, we focus on mathematical and computational models of the ECM in processes such as cell migration including cancer metastasis, and in tissue structure and morphogenesis. By highlighting the predictive power of these models, we aim to help bridge the gap between experimental and computational approaches to studying the ECM and to provide guidance on selecting an appropriate model framework to complement corresponding experimental studies.
Collapse
Affiliation(s)
- Rebecca M. Crossley
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Samuel Johnson
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | - Erika Tsingos
- Computational Developmental Biology Group, Institute of Biodynamics and Biocomplexity, Utrecht University, Utrecht, Netherlands
| | - Zoe Bell
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Massimiliano Berardi
- LaserLab, Department of Physics and Astronomy, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Optics11 life, Amsterdam, Netherlands
| | | | - Quirine J. S. Braat
- Department of Applied Physics and Science Education, Eindhoven University of Technology, Eindhoven, Netherlands
| | - John Metzcar
- Department of Intelligent Systems Engineering, Indiana University, Bloomington, IN, United States
- Department of Informatics, Indiana University, Bloomington, IN, United States
| | | | - Yuan Yin
- Wolfson Centre for Mathematical Biology, Mathematical Institute, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
19
|
Melo-Fonseca F, Gasik M, Cruz A, Moreira D, S. Silva F, Miranda G, Mendes Pinto I. Engineering a Hybrid Ti6Al4V-Based System for Responsive and Consistent Osteogenesis. ACS OMEGA 2024; 9:8985-8994. [PMID: 38434873 PMCID: PMC10905591 DOI: 10.1021/acsomega.3c07232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/17/2023] [Accepted: 01/05/2024] [Indexed: 03/05/2024]
Abstract
As the aging population increases worldwide, the incidence of musculoskeletal diseases and the need for orthopedic implants also arise. One of the most desirable goals in orthopedic reconstructive therapies is de novo bone formation. Yet, reproducible, long-lasting, and cost-effective strategies for implants that strongly induce osteogenesis are still in need. Nanoengineered titanium substrates (and their alloys) are among the most used materials in orthopedic implants. Although having high biocompatibility, titanium alloys hold a low bioactivity profile. The osteogenic capacity and osseointegration of Ti-based implantable systems are limited, as they critically depend on the body-substrate interactions defined by blood proteins adsorbed into implant surfaces that ultimately lead to the recruitment, proliferation, and differentiation of mesenchymal stem cells (MSCs) to comply bone formation and regeneration. In this work, a hybrid Ti6Al4V system combining micro- and nanoscale modifications induced by hydrothermal treatment followed by functionalization with a bioactive compound (fibronectin derived from human plasma) is proposed, aiming for bioactivity improvement. An evaluation of the biological activity and cellular responses in vitro with respect to bone regeneration indicated that the integration of morphological and chemical modifications into Ti6Al4V surfaces induces the osteogenic differentiation of MSCs to improve bone regeneration by an enhancement of mineral matrix formation that accelerates the osseointegration process. Overall, this hybrid system has numerous competitive advantages over more complex treatments, including reproducibility, low production cost, and potential for improved long-term maintenance of the implant.
Collapse
Affiliation(s)
- Francisca Melo-Fonseca
- Center
for MicroElectroMechanical Systems (CMEMS-UMinho), University of Minho, Guimarães 4800-058, Portugal
- LABBELS—Associate
Laboratory, Braga, Guimarães 4710-057, Portugal
- International
Iberian Nanotechnology Laboratory (INL), Braga 4715-330, Portugal
| | - Michael Gasik
- School
of Chemical Engineering, Aalto University
Foundation, Espoo 00076, Finland
| | - Andrea Cruz
- International
Iberian Nanotechnology Laboratory (INL), Braga 4715-330, Portugal
| | - Daniel Moreira
- Institute
for Research and Innovation in Health (i3S), Porto 4200-135, Portugal
| | - Filipe S. Silva
- Center
for MicroElectroMechanical Systems (CMEMS-UMinho), University of Minho, Guimarães 4800-058, Portugal
- LABBELS—Associate
Laboratory, Braga, Guimarães 4710-057, Portugal
| | - Georgina Miranda
- CICECO, Aveiro
Institute of Materials, Department of Materials and Ceramic Engineering, University of Aveiro, Aveiro 3810-193, Portugal
| | - Inês Mendes Pinto
- International
Iberian Nanotechnology Laboratory (INL), Braga 4715-330, Portugal
- Institute
for Research and Innovation in Health (i3S), Porto 4200-135, Portugal
| |
Collapse
|
20
|
Hoveidaei AH, Sadat-Shojai M, Mosalamiaghili S, Salarikia SR, Roghani-Shahraki H, Ghaderpanah R, Ersi MH, Conway JD. Nano-hydroxyapatite structures for bone regenerative medicine: Cell-material interaction. Bone 2024; 179:116956. [PMID: 37951520 DOI: 10.1016/j.bone.2023.116956] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/04/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
Bone tissue engineering holds great promise for the regeneration of damaged or severe bone defects. However, several challenges hinder its translation into clinical practice. To address these challenges, interdisciplinary efforts and advances in biomaterials, cell biology, and bioengineering are required. In recent years, nano-hydroxyapatite (nHA)-based scaffolds have emerged as a promising approach for the development of bone regenerative agents. The unique similarity of nHA with minerals found in natural bones promotes remineralization and stimulates bone growth, which are crucial factors for efficient bone regeneration. Moreover, nHA exhibits desirable properties, such as strong chemical interactions with bone and facilitation of tissue growth, without inducing inflammation or toxicity. It also promotes osteoblast survival, adhesion, and proliferation, as well as increasing alkaline phosphatase activity, osteogenic differentiation, and bone-specific gene expression. However, it is important to note that the effect of nHA on osteoblast behavior is dose-dependent, with cytotoxic effects observed at higher doses. Additionally, the particle size of nHA plays a crucial role, with smaller particles having a more significant impact. Therefore, in this review, we highlighted the potential of nHA for improving bone regeneration processes and summarized the available data on bone cell response to nHA-based scaffolds. In addition, an attempt is made to portray the current status of bone tissue engineering using nHA/polymer hybrids and some recent scientific research in the field.
Collapse
Affiliation(s)
- Amir Human Hoveidaei
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Mehdi Sadat-Shojai
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran
| | - Seyedarad Mosalamiaghili
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Rezvan Ghaderpanah
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hamed Ersi
- Evidence Based Medicine Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Clinical Research Development Center of Shahid Mohammadi Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Janet D Conway
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA.
| |
Collapse
|
21
|
Li X, Mao X, Cong J, Zhang Q, Chen W, Yan K, Huang Y, Su D, Xiang Q. Recombinantly expressed rhFEB remodeled the skin defect of db/db mice. Appl Microbiol Biotechnol 2024; 108:183. [PMID: 38285241 PMCID: PMC10824822 DOI: 10.1007/s00253-024-13021-9] [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: 08/03/2023] [Revised: 01/04/2024] [Accepted: 01/18/2024] [Indexed: 01/30/2024]
Abstract
Fibronectin (FN) and collagen are vital components of the extracellular matrix (ECM). These proteins are essential for tissue formation and cell alignment during the wound healing stage. In particular, FN interacts with collagens to activate various intracellular signaling pathways to maintain ECM stability. A novel recombinant extra domain-B fibronectin (EDB-FN)-COL3A1 fusion protein (rhFEB) was designed to mimic the ECM to promote chronic and refractory skin ulcer wound healing. rhFEB significantly enhanced cell adhesion and migration, vascular ring formation, and the production of new collagen I (COL1A1) in vitro. rhFEB decreased M1 macrophages and further modulated the wound microenvironment, which was confirmed by the treatment of db/db mice with rhFEB. Accelerated wound healing was shown during the initial stages in rhFEB-treated db/db mice, as was enhanced follicle regeneration, re-epithelialization, collagen deposition, granulation, inflammation, and angiogenesis. The wound chronicity of diabetic foot ulcers (DFUs) remains the main challenge in current and future treatment. rhFEB may be a candidate molecule for regulating M1 macrophages during DFU healing. KEY POINTS: • A recombinant protein EDB-FN-collagen III (rhFEB) was highly expressed in Escherichia coli • rhFEB protein induces COL1A1 secretion in human skin fibroblasts • rhFEB protein accelerates diabetic wound healing.
Collapse
Affiliation(s)
- Xiaomin Li
- Perfect Life and Health Institute Co., Ltd, Zhongshan, China
| | - Xinliang Mao
- Perfect Life and Health Institute Co., Ltd, Zhongshan, China
| | - Jianhang Cong
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
| | - Qirong Zhang
- Biopharmaceutical R&D Center, Jinan University, Guangzhou, China
| | - Wenjie Chen
- Perfect Life and Health Institute Co., Ltd, Zhongshan, China
| | - Kunjun Yan
- Biopharmaceutical R&D Center, Jinan University, Guangzhou, China
| | - Yadong Huang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China
- Biopharmaceutical R&D Center, Jinan University, Guangzhou, China
| | - Dun Su
- Perfect Life and Health Institute Co., Ltd, Zhongshan, China.
| | - Qi Xiang
- Institute of Biomedicine and Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou, China.
- Biopharmaceutical R&D Center, Jinan University, Guangzhou, China.
| |
Collapse
|
22
|
Mahzarnia A, Lutz MW, Badea A. A Continuous Extension of Gene Set Enrichment Analysis Using the Likelihood Ratio Test Statistics Identifies Vascular Endothelial Growth Factor as a Candidate Pathway for Alzheimer's Disease via ITGA5. J Alzheimers Dis 2024; 97:635-648. [PMID: 38160360 PMCID: PMC10836573 DOI: 10.3233/jad-230934] [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: 11/01/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) involves brain neuropathologies such as amyloid plaque and hyperphosphorylated tau tangles and is accompanied by cognitive decline. Identifying the biological mechanisms underlying disease onset and progression based on quantifiable phenotypes will help understand disease etiology and devise therapies. OBJECTIVE Our objective was to identify molecular pathways associated with hallmark AD biomarkers and cognitive status, accounting for variables such as age, sex, education, and APOE genotype. METHODS We introduce a pathway-based statistical approach, extending the gene set likelihood ratio test to continuous phenotypes. We first analyzed independently each of the three phenotypes (amyloid-β, tau, cognition) using continuous gene set likelihood ratio tests to account for covariates, including age, sex, education, and APOE genotype. The analysis involved 634 subjects with data available for all three phenotypes, allowing for the identification of common pathways. RESULTS We identified 14 pathways significantly associated with amyloid-β; 5 associated with tau; and 174 associated with cognition, which showed a larger number of pathways compared to biomarkers. A single pathway, vascular endothelial growth factor receptor binding (VEGF-RB), exhibited associations with all three phenotypes. Mediation analysis showed that among the VEGF-RB family genes, ITGA5 mediates the relationship between cognitive scores and pathological biomarkers. CONCLUSIONS We presented a new statistical approach linking continuous phenotypes, gene expression across pathways, and covariates like sex, age, and education. Our results reinforced VEGF RB2's role in AD cognition and demonstrated ITGA5's significant role in mediating the AD pathology-cognition connection.
Collapse
Affiliation(s)
- Ali Mahzarnia
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
| | - Michael W. Lutz
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Alexandra Badea
- Department of Radiology, Duke University School of Medicine, Durham, NC, USA
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
- Biomedical Engineering, Duke University, Durham, NC, USA
- Brain Imaging and Analysis Center, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
23
|
Lin CH, Tang X, Chen P, Luo SC. Unraveling the Adhesion Behavior of Different Cell Lines on Biomimetic PEDOT Interfaces: The Role of Surface Morphology and Antifouling Properties. ACS APPLIED BIO MATERIALS 2023; 6:5695-5707. [PMID: 37971532 DOI: 10.1021/acsabm.3c00833] [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] [Indexed: 11/19/2023]
Abstract
The poly(3,4-ethylenedioxythiophene) (PEDOT) interface, renowned for its biocompatibility and intrinsic conductivity, holds substantial potential in biosensing and cellular modulation. Through strategic functionalization, PEDOT derivatives can be adaptable for multifaceted applications. Notably, integrating phosphorylcholine (PC) groups into PEDOT, mimicking the hydrophilic headgroups from cell membranes, confers exceptional antifouling properties on the coating. This study systematically investigated biomolecule interactions with distinct forms of PEDOT, incorporating variations in surface modifications and structure. Zwitterionic PEDOT-PC was electropolymerized on smooth and nanostructured surfaces using various feeding ratios in electrolytes to finely control the antifouling properties of the interface. Precise electropolymerization conditions governed the attainment of smooth and nanostructured filamentous surfaces. The study employed a quartz crystal microbalance with dissipation (QCM-D) to assess protein binding behavior. Bovine serum albumin (BSA), lysozyme (LYZ), cytochrome c (cyt c), and fibronectin (FN) were used to evaluate their binding affinities for PEDOT films. FN, a pivotal extracellular matrix component, was included for connecting to cell adhesion behavior. Furthermore, the cellular adhesion behaviors on PEDOT interfaces were evaluated. Three cell lines─MG-63 osteosarcoma, HeLa cervical cancer, and fibroblast NIH/3T3 were examined. The presence of PC moieties significantly altered the adhesive response, including the number of attached cells, their morphologies, and nucleus shrinkage. MG-63 cells exhibited the highest tolerance for PC moieties. A feeding ratio of PEDOT-PC exceeding 70% resulted in cell apoptosis. This study contributes to understanding biomolecule adsorption on PEDOT surfaces of diverse morphologies and degrees of the antifouling moiety. Meanwhile, it also sheds light on the responses of various cell types.
Collapse
Affiliation(s)
- Chia-Hsin Lin
- Department of Materials Science and Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Xiaofang Tang
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Peilin Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
- Institute of Physics, Academia Sinica, Taipei 11529, Taiwan
| | - Shyh-Chyang Luo
- Department of Materials Science and Engineering, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes (NHRI), Miaoli County 35053, Taiwan
| |
Collapse
|
24
|
Atkin L. Small intestinal submucosa extracellular matrix: advantages, evidence and application. J Wound Care 2023; 32:S4-S7. [PMID: 38175770 DOI: 10.12968/jowc.2023.32.sup12a.s4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Affiliation(s)
- Leanne Atkin
- Vascular Nurse Consultant/Research Fellow, University of Huddersfield and Mid Yorkshire Hospitals NHS Trust, UK
| |
Collapse
|
25
|
Hulme CH, Mennan C, McCarthy HS, Davies R, Lan T, Rix L, Perry J, Wright K. A comprehensive review of quantum bioreactor cell manufacture: Research and clinical applications. Cytotherapy 2023; 25:1017-1026. [PMID: 37162433 DOI: 10.1016/j.jcyt.2023.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/15/2023] [Accepted: 04/10/2023] [Indexed: 05/11/2023]
Abstract
The Quantum cell expansion system manufactured by Terumo-BCT is perhaps the most widely reported Good Manufacturing Practice-compliant bioreactor used for the expansion of adherent cell populations, both for research purposes and clinical cell-based therapies/trials. Although the system was originally designed for adherent cell expansion, more recently suspension cultures and extracellular vesicle manufacturing protocols have been published using the Quantum system. Cell therapy research and regenerative medicine in general is a rapidly expanding field and as such it is likely that the use of this system will become even more widespread and perhaps mandatory, for both research and development and in the clinic. The purpose of this review is to describe, compare and discuss the diverse range of research and clinical applications currently using the Quantum system, which to our knowledge has not previously been reviewed. In addition, current and future challenges will also be discussed.
Collapse
Affiliation(s)
- Charlotte H Hulme
- Centre for Regenerative Medicine Research, School of Pharmacy and Bioengineering, Keele University, Keele, Newcastle, United Kingdom; Robert Jones and Agnes Hunt Orthopaedic Hospital, Gobowen, Oswestry, Shropshire, United Kingdom
| | - Claire Mennan
- Centre for Regenerative Medicine Research, School of Pharmacy and Bioengineering, Keele University, Keele, Newcastle, United Kingdom; Robert Jones and Agnes Hunt Orthopaedic Hospital, Gobowen, Oswestry, Shropshire, United Kingdom
| | - Helen S McCarthy
- Centre for Regenerative Medicine Research, School of Pharmacy and Bioengineering, Keele University, Keele, Newcastle, United Kingdom; Robert Jones and Agnes Hunt Orthopaedic Hospital, Gobowen, Oswestry, Shropshire, United Kingdom
| | - Rebecca Davies
- Centre for Regenerative Medicine Research, School of Pharmacy and Bioengineering, Keele University, Keele, Newcastle, United Kingdom; Robert Jones and Agnes Hunt Orthopaedic Hospital, Gobowen, Oswestry, Shropshire, United Kingdom
| | - Tian Lan
- Centre for Regenerative Medicine Research, School of Pharmacy and Bioengineering, Keele University, Keele, Newcastle, United Kingdom; Robert Jones and Agnes Hunt Orthopaedic Hospital, Gobowen, Oswestry, Shropshire, United Kingdom
| | - Larissa Rix
- Centre for Regenerative Medicine Research, School of Pharmacy and Bioengineering, Keele University, Keele, Newcastle, United Kingdom; Robert Jones and Agnes Hunt Orthopaedic Hospital, Gobowen, Oswestry, Shropshire, United Kingdom
| | - Jade Perry
- Centre for Regenerative Medicine Research, School of Pharmacy and Bioengineering, Keele University, Keele, Newcastle, United Kingdom; Robert Jones and Agnes Hunt Orthopaedic Hospital, Gobowen, Oswestry, Shropshire, United Kingdom
| | - Karina Wright
- Centre for Regenerative Medicine Research, School of Pharmacy and Bioengineering, Keele University, Keele, Newcastle, United Kingdom; Robert Jones and Agnes Hunt Orthopaedic Hospital, Gobowen, Oswestry, Shropshire, United Kingdom.
| |
Collapse
|
26
|
Lewis M, David G, Jacobs D, Kuczwara P, Woessner AE, Kim JW, Quinn KP, Song Y. Neuro-regenerative behavior of adipose-derived stem cells in aligned collagen I hydrogels. Mater Today Bio 2023; 22:100762. [PMID: 37600354 PMCID: PMC10433000 DOI: 10.1016/j.mtbio.2023.100762] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 07/16/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023] Open
Abstract
Peripheral nerve injuries persist as a major clinical issue facing the US population and can be caused by stretch, laceration, or crush injuries. Small nerve gaps are simple to treat, and the nerve stumps can be reattached with sutures. In longer nerve gaps, traditional treatment options consist of autografts, hollow nerve guidance conduits, and, more recently, manufactured fibrous scaffolds. These manufactured scaffolds often incorporate stem cells, growth factors, and/or extracellular matrix (ECM) proteins to better mimic the native environment but can have issues with homogenous cell distribution or uniformly oriented neurite outgrowth in scaffolds without fibrous alignment. Here, we utilize a custom device to fabricate collagen I hydrogels with aligned fibers and encapsulated adipose-derived mesenchymal stem cells (ASCs) for potential use as a peripheral nerve repair graft. Initial results of our scaffold system revealed significantly less cell viability in higher collagen gel concentrations; 3 mg/mL gels showed 84.8 ± 7.3% viable cells, compared to 6 mg/mL gels viability of 76.7 ± 9.5%. Mechanical testing of the 3 mg/mL gels showed a Young's modulus of 6.5 ± 0.8 kPa nearly matching 7.45 kPa known to support Schwann cell migration. Further analysis of scaffolds coupled with stretching in vitro revealed heightened angiogenic and factor secretion, ECM deposition, fiber alignment, and dorsal root ganglia (DRG) neurite outgrowth along the axis of fiber alignment. Our platform serves as an in vitro testbed to assess neuro-regenerative potential of ASCs in aligned collagen fiber scaffolds and may provide guidance on next-generation nerve repair scaffold design.
Collapse
Affiliation(s)
- Mackenzie Lewis
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Gabriel David
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Danielle Jacobs
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Patrick Kuczwara
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
- Department of Biological & Agricultural Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Alan E. Woessner
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Jin-Woo Kim
- Department of Biological & Agricultural Engineering; University of Arkansas, Fayetteville, AR, USA
- Materials Science & Engineering Program; University of Arkansas, Fayetteville, AR, USA
| | - Kyle P. Quinn
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| | - Younghye Song
- Department of Biomedical Engineering; University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|
27
|
Mitchell J, Lo KWH. The Use of Small-Molecule Compounds for Cell Adhesion and Migration in Regenerative Medicine. Biomedicines 2023; 11:2507. [PMID: 37760948 PMCID: PMC10525671 DOI: 10.3390/biomedicines11092507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Cell adhesion is essential for cell survival, communication, and regulation, and it is of fundamental importance in the development and maintenance of tissues. Cell adhesion has been widely explored due to its many important roles in the fields of tissue regenerative engineering and cell biology. This is because the mechanical interactions between a cell and its extracellular matrix (ECM) can influence and control cell behavior and function. Currently, biomaterials for regenerative medicine have been heavily investigated as substrates for promoting a cells' adhesive properties and subsequent proliferation, tissue differentiation, and maturation. Specifically, the manipulation of biomaterial surfaces using ECM coatings such as fibronectin extracted from animal-derived ECM have contributed significantly to tissue regenerative engineering as well as basic cell biology research. Additionally, synthetic and natural bioadhesive agents with pronounced abilities to enhance adhesion in numerous biological components and molecules have also been assessed in the field of tissue regeneration. Research into the use of facilitative bioadhesives has aimed to further optimize the biocompatibility, biodegradability, toxicity levels, and crosslinking duration of bioadhesive materials for improved targeted delivery and tissue repair. However, the restrictive drawbacks of some of these bioadhesive and animal-derived materials include the potential risk of disease transmission, immunogenicity, poor reproducibility, impurities, and instability. Therefore, it is necessary for alternative strategies to be sought out to improve the quality of cell adhesion to biomaterials. One promising strategy involves the use of cell-adhesive small molecules. Small molecules are relatively inexpensive, stable, and low-molecular-weight (<1000 Da) compounds with great potential to serve as efficient alternatives to conventional bioadhesives, ECM proteins, and other derived peptides. Over the past few years, a number of cell adhesive small molecules with the potential for tissue regeneration have been reported. In this review, we discuss the current progress using cell adhesive small molecules to regulate tissue regeneration.
Collapse
Affiliation(s)
- Juan Mitchell
- School of Dental Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA;
| | - Kevin W.-H. Lo
- Connecticut Convergence Institute for Translation in Regenerative Engineering, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Medicine, Division of Endocrinology, School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA
- Department of Biomedical Engineering, School of Engineering, University of Connecticut, Storrs, CT 06268, USA
- Institute of Materials Science (IMS), School of Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
28
|
Galitsyna EV, Buianova AA, Kozhukhov VI, Domogatsky SP, Bukharova TB, Goldshtein DV. Cytocompatibility and Osteoinductive Properties of Collagen-Fibronectin Hydrogel Impregnated with siRNA Targeting Glycogen Synthase Kinase 3β: In Vitro Study. Biomedicines 2023; 11:2363. [PMID: 37760805 PMCID: PMC10525875 DOI: 10.3390/biomedicines11092363] [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: 07/30/2023] [Revised: 08/16/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
In this study, we developed an osteoplastic material based on collagen-fibronectin hydrogel impregnated with siRNA molecules targeting glycogen synthase kinase 3β (GSK3β), which inhibits the osteogenic differentiation of mesenchymal stem cells. The hydrogel impregnated with polyplexes containing siRNA GSK3β and polyethylenimine has been shown to have no cytotoxic effect: there was no statistically significant change in the cell's viability after 7 days of incubation in its presence compared to the control group. On days 2 and 7, an increase in the level of expression of markers of osteogenic differentiation was observed, which confirms the osteoinductive qualities of the material. It has been demonstrated that the hydrogel maintains cell adhesion. Our results obtained in vitro indicate cytocompatibility and osteoinductive properties of collagen-fibronectin hydrogel impregnated with siRNA GSK3β molecules.
Collapse
Affiliation(s)
- Elena V. Galitsyna
- Research Centre for Medical Genetics, 1 Moskvorechye Str., 115522 Moscow, Russia
| | | | - Vadim I. Kozhukhov
- Federal State Budgetary Institution, National Medical Research Centre of Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Sergey P. Domogatsky
- Federal State Budgetary Institution, National Medical Research Centre of Cardiology, Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Tatiana B. Bukharova
- Research Centre for Medical Genetics, 1 Moskvorechye Str., 115522 Moscow, Russia
| | | |
Collapse
|
29
|
Zhang J, Xu X, Liang Y, Wu X, Qian Z, Zhang L, Wang T. Particulate matter promotes the epithelial to mesenchymal transition in human lung epithelial cells via the ROS pathway. Am J Transl Res 2023; 15:5159-5167. [PMID: 37692935 PMCID: PMC10492054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 07/04/2023] [Indexed: 09/12/2023]
Abstract
OBJECTS Epidemiologic studies have linked exposure to airborne pollutant particulate matter (PM) with increased rates of chronic cardiopulmonary diseases, including asthma and idiopathic pulmonary fibrosis (IPF). Several investigations have suggested that the epithelial-to-mesenchymal transition (EMT) may contribute to the complex pathobiology of environmental exposure-mediated pulmonary fibrosis. The present study was designed to characterize the mechanisms of PM-mediated EMT in human lung epithelial cells (HBECs). METHODS AND RESULTS PM induced significant dose (0-100 μg/ml) and time (0-72 h)-dependent increases in transforming growth factor β (TGFβ) and fibronectin (FN) protein levels in HBECs lysates. PM-activated TGFβ and FN protein production in HBECs was prevented by the antioxidant N-acetyl-cysteine (NAC, 5 mM). Furthermore, the NF-κB inhibitor BAY11-7082 (5 μM) abolished PM-induced FN production in HBECs. Biomarkers of EMT (ACTA2, SNAIL1 and SNAIL2) in PM-treated HBECs were significantly increased at the mRNA level compared to control cells. CONCLUSIONS These results demonstrate that PM increases protein levels of TGFβ and FN via reactive oxygen species (ROS)-dependent pathways. In addition, PM exposure induces EMT in human lung epithelial cells, supporting a novel mechanism for PM-induced pulmonary fibrosis.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Medicine, University of ArizonaTucson, AZ, USA
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical UniversityBeijing, China
| | - Xiaoyan Xu
- Department of Medicine, University of ArizonaTucson, AZ, USA
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical UniversityBeijing, China
| | - Ying Liang
- Department of Medicine, University of ArizonaTucson, AZ, USA
| | - Xiaomin Wu
- Department of Medicine, University of ArizonaTucson, AZ, USA
| | - Zhongqing Qian
- Anhui Provincial Key Laboratory of Immunology in Chronic Diseases, Anhui Provincial Key Laboratory of Infection and Immunology, Department of Laboratory Medicine, Bengbu Medical CollegeBengbu, Anhui, China
| | - Liming Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine and Beijing Chao-Yang Hospital, Capital Medical UniversityBeijing, China
| | - Ting Wang
- Department of Medicine, University of ArizonaTucson, AZ, USA
- Center of Translational Science, Florida International University11350 SW Village Parkway, Port St. Lucie, FL, USA
| |
Collapse
|
30
|
Izac JR, Kwee EJ, Tian L, Elsheikh E, Gaigalas AK, Elliott JT, Wang L. Development of a Cell-Based SARS-CoV-2 Pseudovirus Neutralization Assay Using Imaging and Flow Cytometry Analysis. Int J Mol Sci 2023; 24:12332. [PMID: 37569707 PMCID: PMC10418775 DOI: 10.3390/ijms241512332] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/28/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
COVID-19 is an ongoing, global pandemic caused by the novel, highly infectious SARS-CoV-2 virus. Efforts to mitigate the effects of SARS-CoV-2, such as mass vaccination and development of monoclonal therapeutics, require precise measurements of correlative, functional neutralizing antibodies that block virus infection. The development of rapid, safe, and easy-to-use neutralization assays is essential for faster diagnosis and treatment. Here, we developed a vesicular stomatitis virus (VSV)-based neutralization assay with two readout methods, imaging and flow cytometry, that were capable of quantifying varying degrees of neutralization in patient serum samples. We tested two different spike-pseudoviruses and conducted a time-course assay at multiple multiplicities of infection (MOIs) to optimize the assay workflow. The results of this assay correlate with the results of previously developed serology and surrogate neutralization assays. The two pseudovirus readout methods produced similar values of 50% neutralization titer values. Harvest-free in situ readouts for live-cell imaging and high-throughput analysis results for flow cytometry can provide unique capabilities for fast evaluation of neutralization, which is critical for the mitigation of future pandemics.
Collapse
Affiliation(s)
- Jerilyn R. Izac
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (L.T.); (E.E.); (A.K.G.); (J.T.E.); (L.W.)
| | - Edward J. Kwee
- Biosystems and Biomaterials Division, National Institute of Standards and Technology (NIST), Gaithersburg, MD 20899, USA; (L.T.); (E.E.); (A.K.G.); (J.T.E.); (L.W.)
| | | | | | | | | | | |
Collapse
|
31
|
Rezvanian P, Álvarez-López A, Tabraue-Rubio R, Daza R, Colchero L, Elices M, Guinea GV, González-Nieto D, Pérez-Rigueiro J. Modulation of Cell Response through the Covalent Binding of Fibronectin to Titanium Substrates. J Funct Biomater 2023; 14:342. [PMID: 37504837 PMCID: PMC10381834 DOI: 10.3390/jfb14070342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/16/2023] [Accepted: 06/26/2023] [Indexed: 07/29/2023] Open
Abstract
Titanium (Ti-6Al-4V) substrates were functionalized through the covalent binding of fibronectin, and the effect of the existence of this extracellular matrix protein on the surface of the material was assessed by employing mesenchymal stem cell (MSC) cultures. The functionalization process comprised the usage of the activation vapor silanization (AVS) technique to deposit a thin film with a high surface density of amine groups on the material, followed by the covalent binding of fibronectin to the amine groups using the N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride/N-hydroxysuccinimide (EDC/NHS) crosslinking chemistry. The biological effect of the fibronectin on murine MSCs was assessed in vitro. It was found that functionalized samples not only showed enhanced initial cell adhesion compared with bare titanium, but also a three-fold increase in the cell area, reaching values comparable to those found on the polystyrene controls. These results provide compelling evidence of the potential to modulate the response of the organism to an implant through the covalent binding of extracellular matrix proteins on the prosthesis.
Collapse
Affiliation(s)
- Parsa Rezvanian
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan 8159358686, Iran
| | - Aroa Álvarez-López
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Raquel Tabraue-Rubio
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Bioactive Surfaces S.L. C/Puerto de Navacerrada 18, Galapagar, 28260 Madrid, Spain
| | - Rafael Daza
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Luis Colchero
- Bioactive Surfaces S.L. C/Puerto de Navacerrada 18, Galapagar, 28260 Madrid, Spain
| | - Manuel Elices
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Gustavo V Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, 28040 Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain
- Departamento de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Bioactive Surfaces S.L. C/Puerto de Navacerrada 18, Galapagar, 28260 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, 28040 Madrid, Spain
| |
Collapse
|
32
|
Jia Z, Ma H, Liu J, Yan X, Liu T, Cheng YY, Li X, Wu S, Zhang J, Song K. Preparation and Characterization of Polylactic Acid/Nano Hydroxyapatite/Nano Hydroxyapatite/Human Acellular Amniotic Membrane (PLA/nHAp/HAAM) Hybrid Scaffold for Bone Tissue Defect Repair. MATERIALS (BASEL, SWITZERLAND) 2023; 16:1937. [PMID: 36903052 PMCID: PMC10003763 DOI: 10.3390/ma16051937] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/06/2023] [Accepted: 02/23/2023] [Indexed: 06/18/2023]
Abstract
Bone tissue engineering is a novel and efficient repair method for bone tissue defects, and the key step of the bone tissue engineering repair strategy is to prepare non-toxic, metabolizable, biocompatible, bone-induced tissue engineering scaffolds of suitable mechanical strength. Human acellular amniotic membrane (HAAM) is mainly composed of collagen and mucopolysaccharide; it has a natural three-dimensional structure and no immunogenicity. In this study, a polylactic acid (PLA)/Hydroxyapatite (nHAp)/Human acellular amniotic membrane (HAAM) composite scaffold was prepared and the porosity, water absorption and elastic modulus of the composite scaffold were characterized. After that, the cell-scaffold composite was constructed using newborn Sprague Dawley (SD) rat osteoblasts to characterize the biological properties of the composite. In conclusion, the scaffolds have a composite structure of large and small holes with a large pore diameter of 200 μm and a small pore diameter of 30 μm. After adding HAAM, the contact angle of the composite decreases to 38.7°, and the water absorption reaches 249.7%. The addition of nHAp can improve the scaffold's mechanical strength. The degradation rate of the PLA+nHAp+HAAM group was the highest, reaching 39.48% after 12 weeks. Fluorescence staining showed that the cells were evenly distributed and had good activity on the composite scaffold; the PLA+nHAp+HAAM scaffold has the highest cell viability. The adhesion rate to HAAM was the highest, and the addition of nHAp and HAAM could promote the rapid adhesion of cells to scaffolds. The addition of HAAM and nHAp can significantly promote the secretion of ALP. Therefore, the PLA/nHAp/HAAM composite scaffold can support the adhesion, proliferation and differentiation of osteoblasts in vitro which provide sufficient space for cell proliferation, and is suitable for the formation and development of solid bone tissue.
Collapse
Affiliation(s)
- Zhilin Jia
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
- Department of Hematology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Hailin Ma
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Jiaqi Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Xinyu Yan
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Tianqing Liu
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Xiangqin Li
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shuo Wu
- Department of Medical Oncology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Jingying Zhang
- Key Laboratory of 3D Printing Technology in Stomatology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523808, China
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
33
|
Guagliano G, Volpini C, Sardelli L, Bloise N, Briatico-Vangosa F, Cornaglia AI, Dotti S, Villa R, Visai L, Petrini P. Hep3Gel: A Shape-Shifting Extracellular Matrix-Based, Three-Dimensional Liver Model Adaptable to Different Culture Systems. ACS Biomater Sci Eng 2023; 9:211-229. [PMID: 36525369 PMCID: PMC9832437 DOI: 10.1021/acsbiomaterials.2c01226] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Drug-induced hepatotoxicity is a leading cause of clinical trial withdrawal. Therefore, in vitro modeling the hepatic behavior and functionalities is not only crucial to better understand physiological and pathological processes but also to support drug development with reliable high-throughput platforms. Different physiological and pathological models are currently under development and are commonly implemented both within platforms for standard 2D cultures and within tailor-made chambers. This paper introduces Hep3Gel: a hybrid alginate-extracellular matrix (ECM) hydrogel to produce 3D in vitro models of the liver, aiming to reproduce the hepatic chemomechanical niche, with the possibility of adapting its shape to different manufacturing techniques. The ECM, extracted and powdered from porcine livers by a specifically set-up procedure, preserved its crucial biological macromolecules and was embedded within alginate hydrogels prior to crosslinking. The viscoelastic behavior of Hep3Gel was tuned, reproducing the properties of a physiological organ, according to the available knowledge about hepatic biomechanics. By finely tuning the crosslinking kinetics of Hep3Gel, its dualistic nature can be exploited either by self-spreading or adapting its shape to different culture supports or retaining the imposed fiber shape during an extrusion-based 3D-bioprinting process, thus being a shape-shifter hydrogel. The self-spreading ability of Hep3Gel was characterized by combining empirical and numerical procedures, while its use as a bioink was experimentally characterized through rheological a priori printability evaluations and 3D printing tests. The effect of the addition of the ECM was evident after 4 days, doubling the survival rate of cells embedded within control hydrogels. This study represents a proof of concept of the applicability of Hep3Gel as a tool to develop 3D in vitro models of the liver.
Collapse
Affiliation(s)
- Giuseppe Guagliano
- Department
of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133Milan, Italy
| | - Cristina Volpini
- Molecular
Medicine Department (DMM), Center for Health Technologies (CHT), UdR
INSTM, University of Pavia, 27100Pavia, Italy
| | - Lorenzo Sardelli
- Department
of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133Milan, Italy
| | - Nora Bloise
- Molecular
Medicine Department (DMM), Center for Health Technologies (CHT), UdR
INSTM, University of Pavia, 27100Pavia, Italy
| | - Francesco Briatico-Vangosa
- Department
of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133Milan, Italy
| | - Antonia Icaro Cornaglia
- Department
of Public Health, Experimental and Forensic Medicine, Histology and
Embryology Unit, University of Pavia, 27100Pavia, Italy
| | - Silvia Dotti
- National
Reference Center for Alternative Methods, Welfare and Care of Laboratory
Animals, Istituto Zooprofilattico Sperimentale
della Lomabardia ed Emilia Romagna, 25124Brescia, Italy
| | - Riccardo Villa
- National
Reference Center for Alternative Methods, Welfare and Care of Laboratory
Animals, Istituto Zooprofilattico Sperimentale
della Lomabardia ed Emilia Romagna, 25124Brescia, Italy
| | - Livia Visai
- Molecular
Medicine Department (DMM), Center for Health Technologies (CHT), UdR
INSTM, University of Pavia, 27100Pavia, Italy,Medicina
Clinica-Specialistica, UOR5 Laboratorio Di Nanotecnologie, ICS Maugeri, IRCCS, Pavia, Via Boezio, 28-27100Pavia, Italy,Interuniversity
Center for the Promotion of the 3Rs Principles in Teaching and Research
(Centro 3R), Università di Pavia
Unit, 27100Pavia, Italy
| | - Paola Petrini
- Department
of Chemistry, Materials, and Chemical Engineering “G. Natta”, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133Milan, Italy,Interuniversity
Center for the Promotion of the 3Rs Principles in Teaching and Research
(Centro 3R), Politecnico di Milano Unit, 20133Milan, Italy,
| |
Collapse
|
34
|
Development of Inhalable Spray Dried Nitrofurantoin Formulations for the Treatment of Emphysema. Pharmaceutics 2022; 15:pharmaceutics15010146. [PMID: 36678775 PMCID: PMC9867496 DOI: 10.3390/pharmaceutics15010146] [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: 11/30/2022] [Revised: 12/29/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
A central characteristic of emphysematous progression is the continuous destruction of the lung extracellular matrix (ECM). Current treatments for emphysema have only addressed symptoms rather than preventing or reversing the loss of lung ECM. Nitrofurantoin (NF) is an antibiotic that has the potential to induce lung fibrosis as a side effect upon oral administration. Our study aims to repurpose NF as an inhalable therapeutic strategy to upregulate ECM expression, thereby reversing the disease progression within the emphysematous lung. Spray-dried (SD) formulations of NF were prepared in conjunction with a two-fluid nozzle (2FN) and three-fluid nozzle (3FN) using hydroxypropyl methylcellulose (HPMC) and NF at 1:1 w/w. The formulations were characterized for their physicochemical properties (particle size, morphology, solid-state characteristics, aerodynamic behaviour, and dissolution properties) and characterized in vitro with efficacy studies on human lung fibroblasts. The 2FN formulation displayed a mass mean aerodynamic diameter (MMAD) of 1.8 ± 0.05 µm and fine particle fraction (FPF) of 87.4 ± 2.8% with significantly greater deposition predicted in the lower lung region compared to the 3FN formulation (MMAD: 4.4 ± 0.4 µm; FPF: 40 ± 5.8%). Furthermore, drug dissolution studies showed that NF released from the 2FN formulation after 3 h was significantly higher (55.7%) as compared to the 3FN formulation (42.4%). Importantly, efficacy studies in human lung fibroblasts showed that the 2FN formulation induced significantly enhanced ECM protein expression levels of periostin and Type IV Collagen (203.2% and 84.2% increase, respectively) compared to untreated cells, while 3FN formulations induced only a 172.5% increase in periostin and a 38.1% increase in type IV collagen. In conclusion, our study highlights the influence of nozzle choice in inhalable spray-dried formulations and supports the feasibility of using SD NF prepared using 2FN as a potential inhalable therapeutic agent to upregulate ECM protein production.
Collapse
|
35
|
Harati J, Liu K, Shahsavarani H, Du P, Galluzzi M, Deng K, Mei J, Chen HY, Bonakdar S, Aflatoonian B, Hou G, Zhu Y, Pan H, Wong RCB, Shokrgozar MA, Song W, Wang PY. Defined Physicochemical Cues Steering Direct Neuronal Reprogramming on Colloidal Self-Assembled Patterns (cSAPs). ACS NANO 2022; 17:1054-1067. [PMID: 36583476 DOI: 10.1021/acsnano.2c07473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Direct neuronal reprogramming of somatic cells into induced neurons (iNs) has been recently established as a promising approach to generating neuron cells. Previous studies have reported that the biophysical cues of the in vitro microenvironment are potent modulators in the cell fate decision; thus, the present study explores the effects of a customized pattern (named colloidal self-assembled patterns, cSAPs) on iN generation from human fibroblasts using small molecules. The result revealed that the cSAP, composed of binary particles in a hexagonal-close-packed (hcp) geometry, is capable of improving neuronal reprogramming efficiency and steering the ratio of the iN subtypes. Cells exhibited distinct cell morphology, upregulated cell adhesion markers (i.e., SDC1 and ITGAV), enriched signaling pathways (i.e., Hippo and Wnt), and chromatin remodeling on the cSAP compared to those on the control substrates. The result also showed that the iN subtype specification on cSAP was surface-dependent; therefore, the defined physicochemical cue from each cSAP is exclusive. Our findings show that direct cell reprogramming can be manipulated through specific biophysical cues on the artificial matrix, which is significant in cell transdifferentiation and lineage conversion.
Collapse
Affiliation(s)
- Javad Harati
- Lab Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran1316943551, Iran
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang325000, People's Republic of China
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong518055, People's Republic of China
- University of Chinese Academy of Science, Beijing101408, People's Republic of China
| | - Kun Liu
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong518055, People's Republic of China
| | - Hosein Shahsavarani
- Lab Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran1316943551, Iran
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang325000, People's Republic of China
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran1983969411, Iran
| | - Ping Du
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong518055, People's Republic of China
| | - Massimiliano Galluzzi
- Materials Interfaces Center, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong518055, People's Republic of China
| | - Ke Deng
- School of Food and Bioengineering, Xihua University, Chengdu610097, People's Republic of China
| | - Jei Mei
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang325000, People's Republic of China
| | - Hsien-Yeh Chen
- Department of Chemical Engineering, National Taiwan University, Taipei10617, Taiwan
| | - Shahin Bonakdar
- Lab Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran1316943551, Iran
| | - Behrouz Aflatoonian
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd8916188635, Iran
| | - Guoqiang Hou
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong518055, People's Republic of China
| | - Yingjie Zhu
- Shenzhen Key Laboratory of Drug Addiction, Shenzhen Neher Neural Plasticity Laboratory, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong518055, People's Republic of China
| | - Haobo Pan
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong518055, People's Republic of China
| | - Raymond C B Wong
- Centre for Eye Research Australia, Department of Surgery, University of Melbourne, Parkville, Victoria3002, Australia
| | - Mohammad Ali Shokrgozar
- Lab Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, Tehran1316943551, Iran
| | - Weihong Song
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang325000, People's Republic of China
| | - Peng-Yuan Wang
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang325000, People's Republic of China
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong518055, People's Republic of China
| |
Collapse
|
36
|
Hosseini M, Dalley AJ, Shafiee A. Convergence of Biofabrication Technologies and Cell Therapies for Wound Healing. Pharmaceutics 2022; 14:pharmaceutics14122749. [PMID: 36559242 PMCID: PMC9785239 DOI: 10.3390/pharmaceutics14122749] [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: 10/28/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Cell therapy holds great promise for cutaneous wound treatment but presents practical and clinical challenges, mainly related to the lack of a supportive and inductive microenvironment for cells after transplantation. Main: This review delineates the challenges and opportunities in cell therapies for acute and chronic wounds and highlights the contribution of biofabricated matrices to skin reconstruction. The complexity of the wound healing process necessitates the development of matrices with properties comparable to the extracellular matrix in the skin for their structure and composition. Over recent years, emerging biofabrication technologies have shown a capacity for creating complex matrices. In cell therapy, multifunctional material-based matrices have benefits in enhancing cell retention and survival, reducing healing time, and preventing infection and cell transplant rejection. Additionally, they can improve the efficacy of cell therapy, owing to their potential to modulate cell behaviors and regulate spatiotemporal patterns of wound healing. CONCLUSION The ongoing development of biofabrication technologies promises to deliver material-based matrices that are rich in supportive, phenotype patterning cell niches and are robust enough to provide physical protection for the cells during implantation.
Collapse
Affiliation(s)
- Motaharesadat Hosseini
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, Queensland University of Technology, Brisbane, QLD 4059, Australia
- ARC Industrial Transformation Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing (M3D), Queensland University of Technology, Brisbane, QLD 4059, Australia
| | - Andrew J. Dalley
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia
- Royal Brisbane and Women’s Hospital, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia
| | - Abbas Shafiee
- Herston Biofabrication Institute, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia
- Royal Brisbane and Women’s Hospital, Metro North Hospital and Health Service, Brisbane, QLD 4029, Australia
- Frazer Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD 4102, Australia
- Correspondence: or
| |
Collapse
|
37
|
Wang T, Yang J, Mao J, Zhu L, Luo X, Cheng C, Zhang L. ITGA5 inhibition in pancreatic stellate cells re-educates the in vitro tumor-stromal crosstalk. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 40:39. [PMID: 36469173 DOI: 10.1007/s12032-022-01902-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/16/2022] [Indexed: 12/07/2022]
Abstract
The interaction between pancreatic cancer cells (PCCs) and pancreatic stellate cells (PSCs) promotes aggressive progression of pancreatic cancer, and disrupting the tumor-stromal crosstalk is a promising therapeutic strategy. Integrin α5 (ITGA5) is specifically overexpressed in pancreatic cancer stroma and activated PSCs. ITGA5 acts as a mediator in PCCs-PSCs interaction, but its role in regulating biological behaviors of PSCs and PCCs is still not quite clear. In this study, ITGA5 in PSCs was inhibited using its specific inhibitor AV3 peptide or siRNA knockdown technique. Pancreatic cancer SW1990 cells conditioned medium (SW1990-CM) and an indirect co-culture system were used to mimic the environment of the in vitro tumor-stromal crosstalk. Our results showed that ITGA5 inhibition impaired the proliferation and migration of PSCs, but enhanced autophagy. After co-culture with PSCs, SW1990 cells gained some cancer stem cells (CSCs)-like characteristics, such as increased drug resistance, migration and invasion ability, but PSCs with ITGA5 knockdown were incapable of producing these effects. The present results suggested that ITGA5 was involved in the development of the malignant biological behaviors of PSCs and PCCs, and ITGA5 inhibition in PSCs might benefit the treatment of pancreatic cancer by re-educating PCCs-PSCs interaction.
Collapse
Affiliation(s)
- Tao Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Jian Yang
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Juanli Mao
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Lizhi Zhu
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Xiu Luo
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Chao Cheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| | - Lu Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
38
|
Sklenářová R, Akla N, Latorre MJ, Ulrichová J, Franková J. Collagen as a Biomaterial for Skin and Corneal Wound Healing. J Funct Biomater 2022; 13:jfb13040249. [PMID: 36412890 PMCID: PMC9680244 DOI: 10.3390/jfb13040249] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022] Open
Abstract
The cornea and the skin are two organs that form the outer barrier of the human body. When either is injured (e.g., from surgery, physical trauma, or chemical burns), wound healing is initiated to restore integrity. Many cells are activated during wound healing. In particular, fibroblasts that are stimulated often transition into repair fibroblasts or myofibroblasts that synthesize extracellular matrix (ECM) components into the wound area. Control of wound ECM deposition is critical, as a disorganized ECM can block restoration of function. One of the most abundant structural proteins in the mammalian ECM is collagen. Collagen type I is the main component in connective tissues. It can be readily obtained and purified, and short analogs have also been developed for tissue engineering applications, including modulating the wound healing response. This review discusses the effect of several current collagen implants on the stimulation of corneal and skin wound healing. These range from collagen sponges and hydrogels to films and membranes.
Collapse
Affiliation(s)
- Renáta Sklenářová
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University in Olomouc, 775 15 Olomouc, Czech Republic
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC H1T 2M4, Canada
| | - Naoufal Akla
- Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | | | - Jitka Ulrichová
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University in Olomouc, 775 15 Olomouc, Czech Republic
| | - Jana Franková
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine and Dentistry, Palacký University in Olomouc, 775 15 Olomouc, Czech Republic
- Correspondence:
| |
Collapse
|
39
|
Shirbhate U, Bajaj P, Pandher J, Durge K. Fibronectin and Its Applications in Dentistry and Periodontics: A Cell Behaviour Conditioner. Cureus 2022; 14:e30702. [DOI: 10.7759/cureus.30702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/26/2022] [Indexed: 11/05/2022] Open
|
40
|
Chaber P, Tylko G, Włodarczyk J, Nitschke P, Hercog A, Jurczyk S, Rech J, Kubacki J, Adamus G. Surface Modification of PHBV Fibrous Scaffold via Lithium Borohydride Reduction. MATERIALS (BASEL, SWITZERLAND) 2022; 15:7494. [PMID: 36363086 PMCID: PMC9653721 DOI: 10.3390/ma15217494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 10/17/2022] [Accepted: 10/22/2022] [Indexed: 06/16/2023]
Abstract
In this study, lithium borohydride (LiBH4) reduction was used to modify the surface chemistry of poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV) fibers. Although the most common reaction employed in the surface treatment of polyester materials is hydrolysis, it is not suitable for fiber modification of bacterial polyesters, which are highly resistant to this type of reaction. The use of LiBH4 allowed the formation of surface hydroxyl groups under very mild conditions, which was crucial for maintaining the fibers' integrity. The presence of these groups resulted in a noticeable improvement in the surface hydrophilicity of PHBV, as revealed by contact angle measurements. After the treatment with a LiBH4 solution, the electrospun PHBV fibrous mat had a significantly greater number of viable osteoblast-like cells (SaOS-2 cell line) than the untreated mat. Moreover, the results of the cell proliferation measurements correlated well with the observed cell morphology. The most flattened SaOS-2 cells were found on the surface that supported the best cell attachment. Most importantly, the results of our study indicated that the degree of surface modification could be controlled by changing the degradation time and concentration of the borohydride solution. This was of great importance since it allowed optimization of the surface properties to achieve the highest cell-proliferation capacity.
Collapse
Affiliation(s)
- Paweł Chaber
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Grzegorz Tylko
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Gronostajowa 9, 30-387 Kraków, Poland
| | - Jakub Włodarczyk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Paweł Nitschke
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Anna Hercog
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Sebastian Jurczyk
- Institute for Engineering of Polymer Materials and Dyes, Łukasiewicz Research Network, Marii Skłodowskiej-Curie 55, 87-100 Toruń, Poland
| | - Jakub Rech
- Department of Biotechnology and Genetic Engineering, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Poniatowskiego 15, 40-055 Katowice, Poland
| | - Jerzy Kubacki
- Faculty of Science and Technology, Institute of Physics, University of Silesia in Katowice, 75 Pułku Piechoty 1, 41-500 Chorzów, Poland
| | - Grażyna Adamus
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| |
Collapse
|
41
|
Lee A, Sousa de Almeida M, Milinkovic D, Septiadi D, Taladriz-Blanco P, Loussert-Fonta C, Balog S, Bazzoni A, Rothen-Rutishauser B, Petri-Fink A. Substrate stiffness reduces particle uptake by epithelial cells and macrophages in a size-dependent manner through mechanoregulation. NANOSCALE 2022; 14:15141-15155. [PMID: 36205559 PMCID: PMC9585528 DOI: 10.1039/d2nr03792k] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/30/2022] [Indexed: 05/23/2023]
Abstract
Cells continuously exert forces on their environment and respond to changes in mechanical forces by altering their behaviour. Many pathologies such as cancer and fibrosis are hallmarked by dysregulation in the extracellular matrix, driving aberrant behaviour through mechanotransduction pathways. We demonstrate that substrate stiffness can be used to regulate cellular endocytosis of particles in a size-dependent fashion. Culture of A549 epithelial cells and J774A.1 macrophages on polystyrene/glass (stiff) and polydimethylsiloxane (soft) substrates indicated that particle uptake is increased up to six times for A549 and two times for macrophages when cells are grown in softer environments. Furthermore, we altered surface characteristics through the attachment of submicron-sized particles as a method to locally engineer substrate stiffness and topography to investigate the biomechanical changes which occurred within adherent epithelial cells, i.e. characterization of A549 cell spreading and focal adhesion maturation. Consequently, decreasing substrate rigidity and particle-based topography led to a reduction of focal adhesion size. Moreover, expression levels of Yes-associated protein were found to correlate with the degree of particle endocytosis. A thorough appreciation of the mechanical cues may lead to improved solutions to optimize nanomedicine approaches for treatment of cancer and other diseases with abnormal mechanosignalling.
Collapse
Affiliation(s)
- Aaron Lee
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Mauro Sousa de Almeida
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Daela Milinkovic
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Dedy Septiadi
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Patricia Taladriz-Blanco
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
- International Iberian Nanotechnology Laboratory (INL), Water Quality Group, Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - Céline Loussert-Fonta
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Sandor Balog
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | - Amelie Bazzoni
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
| | | | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, 1700 Fribourg, Switzerland.
- Department of Chemistry, University of Fribourg, Chemin du Musée 9, 1700 Fribourg, Switzerland
| |
Collapse
|
42
|
Nashchekina Y, Nikonov P, Prasolov N, Sulatsky M, Chabina A, Nashchekin A. The Structural Interactions of Molecular and Fibrillar Collagen Type I with Fibronectin and Its Role in the Regulation of Mesenchymal Stem Cell Morphology and Functional Activity. Int J Mol Sci 2022; 23:ijms232012577. [PMID: 36293432 PMCID: PMC9604100 DOI: 10.3390/ijms232012577] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/14/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
The observed differences in the structure of native tissue and tissue formed in vitro cause the loss of functional activity of cells cultured in vitro. The lack of fundamental knowledge about the protein mechanism interactions limits the ability to effectively create in vitro native tissue. Collagen is able to spontaneously assemble into fibrils in vitro, but in vivo, other proteins, for example fibronectin, have a noticeable effect on this process. The molecular or fibrillar structure of collagen plays an equally important role. Therefore, we studied the interaction of the molecular and fibrillar structure of collagen with fibronectin. Atomic force and transmission electron microscopy showed that the presence of fibronectin does not affect the native structure and diameter of collagen fibrils. Confocal microscopy demonstrated that the collagen structure affects the cell morphology. Cells are better spread on molecular collagen compared with cells cultured on fibrillar collagen. Fibronectin promotes the formation of a large number of focal contacts, while in combination with collagen of both forms, its effect is leveled. Thus, understanding the mechanisms of the relationship between the protein structure and composition will effectively manage the creation in vitro of a new tissue with native properties.
Collapse
Affiliation(s)
- Yuliya Nashchekina
- Institute of Cytology of the Russian Academy of Sciences, Center of Cell Technologies, Tikhoretsky pr. 4, St. Petersburg 194064, Russia
- Correspondence:
| | - Pavel Nikonov
- Institute of Cytology of the Russian Academy of Sciences, Center of Cell Technologies, Tikhoretsky pr. 4, St. Petersburg 194064, Russia
| | - Nikita Prasolov
- Laboratory «Characterization of Materials and Structures of Solid State Electronics», Ioffe Institute, Polytekhnicheskaya Str. 26, St. Petersburg 194021, Russia
| | - Maksim Sulatsky
- Institute of Cytology of the Russian Academy of Sciences, Center of Cell Technologies, Tikhoretsky pr. 4, St. Petersburg 194064, Russia
| | - Alina Chabina
- Institute of Cytology of the Russian Academy of Sciences, Center of Cell Technologies, Tikhoretsky pr. 4, St. Petersburg 194064, Russia
| | - Alexey Nashchekin
- Laboratory «Characterization of Materials and Structures of Solid State Electronics», Ioffe Institute, Polytekhnicheskaya Str. 26, St. Petersburg 194021, Russia
| |
Collapse
|
43
|
Si H, Zhao N, Pedroza A, Zaske AM, Rosen JM, Creighton CJ, Roarty K. Noncanonical Wnt/Ror2 signaling regulates cell-matrix adhesion to prompt directional tumor cell invasion in breast cancer. Mol Biol Cell 2022; 33:ar103. [PMID: 36001375 PMCID: PMC9582800 DOI: 10.1091/mbc.e22-02-0055] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 07/25/2022] [Accepted: 08/18/2022] [Indexed: 12/03/2022] Open
Abstract
Cell-extracellular matrix (ECM) interactions represent fundamental exchanges during tumor progression, yet how particular signal-transduction factors prompt the conversion of tumor cells into migratory populations capable of systemic spread during metastasis remains elusive. We demonstrate that the noncanonical Wnt receptor, Ror2, regulates tumor cell-driven matrix remodeling and invasion in breast cancer. Ror2 loss-of-function (LOF) triggers the disruption of E-cadherin within tumor cells, accompanied by an increase in tumor cell invasion and collagen realignment in three-dimensional cultures. RNA sequencing of Ror2-deficient organoids further uncovered alterations in actin cytoskeleton, cell adhesion, and collagen cross-linking gene expression programs. Spatially, we pinpoint the up-regulation and redistribution of α5 and β3 integrins together with the production of fibronectin in areas of invasion downstream of Ror2 loss. Wnt/β-catenin-dependent and Wnt/Ror2 alternative Wnt signaling appear to regulate distinct functions for tumor cells regarding their ability to modify cell-ECM exchanges during invasion. Furthermore, blocking either integrin or focal adhesion kinase (FAK), a downstream mediator of integrin-mediated signal transduction, abrogates the enhanced migration observed upon Ror2 loss. These results reveal a critical function for the alternative Wnt receptor, Ror2, as a determinant of tumor cell-driven ECM exchanges during cancer invasion and metastasis.
Collapse
Affiliation(s)
- Hongjiang Si
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Na Zhao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Andrea Pedroza
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
| | - Ana-Maria Zaske
- University of Texas Health Science Center at Houston, Houston, TX 77054
| | - Jeffrey M. Rosen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Breast Cancer Program, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| | - Chad J. Creighton
- Breast Cancer Program, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030
| | - Kevin Roarty
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030
- Breast Cancer Program, Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
44
|
Lim R, Banerjee A, Biswas R, Chari AN, Raghavan S. Mechanotransduction through adhesion molecules: Emerging roles in regulating the stem cell niche. Front Cell Dev Biol 2022; 10:966662. [PMID: 36172276 PMCID: PMC9511051 DOI: 10.3389/fcell.2022.966662] [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: 06/11/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Stem cells have been shown to play an important role in regenerative medicine due to their proliferative and differentiation potential. The challenge, however, lies in regulating and controlling their potential for this purpose. Stem cells are regulated by growth factors as well as an array of biochemical and mechanical signals. While the role of biochemical signals and growth factors in regulating stem cell homeostasis is well explored, the role of mechanical signals has only just started to be investigated. Stem cells interact with their niche or to other stem cells via adhesion molecules that eventually transduce mechanical cues to maintain their homeostatic function. Here, we present a comprehensive review on our current understanding of the influence of the forces perceived by cell adhesion molecules on the regulation of stem cells. Additionally, we provide insights on how this deeper understanding of mechanobiology of stem cells has translated toward therapeutics.
Collapse
Affiliation(s)
- Ryan Lim
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Avinanda Banerjee
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Ritusree Biswas
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
- Sastra University, Thanjavur, TN, India
| | - Anana Nandakumar Chari
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Srikala Raghavan
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
| |
Collapse
|
45
|
Leite ML, de Oliveira Ribeiro RA, Soares DG, Hebling J, de Souza Costa CA. Poly(Caprolactone)-Aligned Nanofibers Associated with Fibronectin-loaded Collagen Hydrogel as a Potent Bioactive Scaffold for Cell-Free Regenerative Endodontics. Int Endod J 2022; 55:1359-1371. [PMID: 36036876 DOI: 10.1111/iej.13823] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/29/2022]
Abstract
AIM Guided tissue regeneration has been considered a promising strategy to replace conventional endodontic therapy of teeth with incomplete root formation. Therefore, the objective of this study was to develop a tubular scaffold (TB-SC) with poly (caprolactone)-aligned nanofibers associated with a fibronectin-loaded collagen hydrogel and assess the pulp regeneration potential mediated by human apical papilla cells (hAPCs) using an in vitro model of teeth with incomplete root formation. METHODOLOGY Aligned nanofiber strips based on 10% poly(caprolactone) (PCL) were synthesized with the electrospinning technique to produce the TB-SCs. These were submitted to different treatments, according to the following groups: TB-SC (negative control): TB-SC without treatment; TB-SC+FN (positive control): TB-SC coated with 10 μg/mL of fibronectin; TB-SC+H: TB-SC associated with collagen hydrogel; TB-SC+HFN: TB-SC associated with fibronectin-loaded collagen hydrogel. Then, the biomaterials were inserted into cylindrical devices to mimic the regenerative therapy of teeth with incomplete root formation. The hAPCs were seeded on the upper surface of the TB-SCs associated or not with any treatment, and cell migration/proliferation and the gene expression of markers related to pulp regeneration (ITGA5, ITGAV, COL1A1, and COL1A3) were evaluated. The data were submitted to ANOVA/Tukey's tests (α=5 %). RESULTS Higher values of cell migration/proliferation and gene expression of all markers tested were observed in groups TB-SC+FN, TB-SC+H, and TB-SC+HFN compared with the TB-SC group (p<0.05). The hAPCs in the TB-SC+HFN group showed the highest values of cell proliferation and gene expression of COL1A1 and COL3A1 (p<0.05), as well as superior cell migration results to groups TB-SC and TB-SC+H (p<0.05). CONCLUSION Aligned nanofiber scaffolds associated with the fibronectin-loaded collagen hydrogel enhanced the migration and proliferation of hAPCs, and gene expression of pulp regeneration markers. Therefore, the use of these biomaterials may be considered an interesting strategy for regenerative pulp therapy of teeth with incomplete root formation.
Collapse
Affiliation(s)
- Maria Luísa Leite
- Department of Oral Health Sciences, Faculty of Dentistry, The University of British Columbia. 2199 Wesbrook Mall, Vancouver, BC, Canada
| | - Rafael Antônio de Oliveira Ribeiro
- Department of Dental Materials and Prosthodontics, Araraquara School of Dentistry, Univ. Estadual Paulista - UNESP. 1680 Humaitá Street, Araraquara, SP, Brazil
| | - Diana Gabriela Soares
- Department of Operative Dentistry, Endodontics and Dental Materials, Bauru School of Dentistry, University of São Paulo. 9-75 Alameda Dr. Octávio Pinheiro Brisolla Street, Bauru, SP, Brazil
| | - Josimeri Hebling
- Departament of Orthodontics and Pediatric Dentistry, Araraquara School of Dentistry, São Paulo State University (Unesp). 1680 Humaitá Street, Araraquara, SP, Brazil
| | - Carlos Alberto de Souza Costa
- Department of Physiology and Pathology, Araraquara School of Dentistry, São Paulo State University (Unesp). Humaitá Street, 1680, Araraquara, SP, Brazil
| |
Collapse
|
46
|
Huang Z, Wang G, Yang B, Li P, Yang T, Wu Y, Yang X, Liu J, Li J. Mechanism of ketotifen fumarate inhibiting renal calcium oxalate stone formation in SD rats. Biomed Pharmacother 2022; 151:113147. [PMID: 35643070 DOI: 10.1016/j.biopha.2022.113147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/02/2022] [Accepted: 05/15/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVES To investigate the inhibitory effect of ketotifen fumarate (KFA), a mast cell membrane stabilizer, on renal calcium oxalate stone (CaOx) formation and its possible molecular mechanism. METHODS We used the Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) database for functional and pathway enrichment analyses of osteopontin (OPN), CD44 and fibronectin (FN). Blood biochemistry, reactive oxygen species ratio (ROS), mast cells, proteins (CD44, OPN and FN) and OPN receptor integrin family genes were detected by ELISA, flow cytometry, immunohistochemistry and RT-QPCR, respectively. RESULTS The crystal area of CaOx in the KFA and Control group was significantly smaller than that in the Model group. The number of activated mast cells, the expression levels of OPN and CD44 in the Control and KFA groups were significantly lower than those in the Model group, and the percentage of ROS in the KFA group was also significantly lower than that in the Model group. The mRNA expression levels of ITGB1, ITGA9, ITGAV and ITGA4 genes in the prominent OPN receptor integrin family increased significantly in the Model group. CONCLUSIONS Ketotifen can effectively inhibit the crystal formation of CaOx and reduce the inflammatory response of tissue in SD rats. The mechanism may be to reduce the infiltration and activation of mast cells in renal tissue and down-regulate the expression of OPN, CD44 and FN in renal tubules and renal interstitium. And affect the synthesis of integrins (ITGA9, ITGA4, ITGAV, ITGB1, ITGB3 and ITGB5) and ROS.
Collapse
Affiliation(s)
- Ziye Huang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian-Mian Avenue, Kunming, Yunnan 650101, PR China
| | - Guang Wang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian-Mian Avenue, Kunming, Yunnan 650101, PR China
| | - Bowei Yang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian-Mian Avenue, Kunming, Yunnan 650101, PR China
| | - Pei Li
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian-Mian Avenue, Kunming, Yunnan 650101, PR China
| | - Tongxin Yang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian-Mian Avenue, Kunming, Yunnan 650101, PR China
| | - Yuyun Wu
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian-Mian Avenue, Kunming, Yunnan 650101, PR China
| | - Xing Yang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian-Mian Avenue, Kunming, Yunnan 650101, PR China
| | - Jianhe Liu
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian-Mian Avenue, Kunming, Yunnan 650101, PR China.
| | - Jiongming Li
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, No. 374 Dian-Mian Avenue, Kunming, Yunnan 650101, PR China.
| |
Collapse
|
47
|
Toll-like receptors and damage-associated molecular patterns in the pathogenesis of heart transplant rejection. Mol Cell Biochem 2022; 477:2841-2850. [PMID: 35678986 DOI: 10.1007/s11010-022-04491-4] [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: 02/21/2022] [Accepted: 05/31/2022] [Indexed: 10/18/2022]
Abstract
Significant strides have been made in our understanding of the immune system and its role in cardiac transplant rejection. Despite the growing knowledge of immune responses, the mortality rate following cardiac transplantation remains grim. Related to procedural and pathological complications, toll-like receptor (TLR) and damage-associated molecular pattern (DAMP) signaling is the most direct and earliest interface between tissue integration and the innate immune response. This in turn can activate an adaptive immune response that further damages myocardial tissue. Furthermore, relevant literature on the status of DAMPs in the context of heart-transplantation remains limited, warranting further attention in clinical and translational research. This review aims to critically appraise the perspectives, advances, and challenges on DAMP-mediated innate immune response in the immune-mediated rejection of cardiac transplantation. Detailed analysis of the influence of TLR and DAMP signaling in mounting the immune response against the transplanted heart holds promise for improving outcomes through early detection and prevention of varied forms of organ rejection.
Collapse
|
48
|
Shi Y, Jiao H, Sun J, Lu X, Yu S, Cheng L, Wang Q, Liu H, Biranje S, Wang J, Liu J. Functionalization of nanocellulose applied with biological molecules for biomedical application: A review. Carbohydr Polym 2022; 285:119208. [DOI: 10.1016/j.carbpol.2022.119208] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 01/29/2022] [Accepted: 01/29/2022] [Indexed: 01/21/2023]
|
49
|
Hao D, Lopez JM, Chen J, Iavorovschi AM, Lelivelt NM, Wang A. Engineering Extracellular Microenvironment for Tissue Regeneration. Bioengineering (Basel) 2022; 9:bioengineering9050202. [PMID: 35621480 PMCID: PMC9137730 DOI: 10.3390/bioengineering9050202] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/23/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
The extracellular microenvironment is a highly dynamic network of biophysical and biochemical elements, which surrounds cells and transmits molecular signals. Extracellular microenvironment controls are of crucial importance for the ability to direct cell behavior and tissue regeneration. In this review, we focus on the different components of the extracellular microenvironment, such as extracellular matrix (ECM), extracellular vesicles (EVs) and growth factors (GFs), and introduce engineering approaches for these components, which can be used to achieve a higher degree of control over cellular activities and behaviors for tissue regeneration. Furthermore, we review the technologies established to engineer native-mimicking artificial components of the extracellular microenvironment for improved regenerative applications. This review presents a thorough analysis of the current research in extracellular microenvironment engineering and monitoring, which will facilitate the development of innovative tissue engineering strategies by utilizing different components of the extracellular microenvironment for regenerative medicine in the future.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (D.H.); (J.-M.L.); (J.C.); (A.M.I.); (N.M.L.)
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Juan-Maria Lopez
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (D.H.); (J.-M.L.); (J.C.); (A.M.I.); (N.M.L.)
| | - Jianing Chen
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (D.H.); (J.-M.L.); (J.C.); (A.M.I.); (N.M.L.)
| | - Alexandra Maria Iavorovschi
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (D.H.); (J.-M.L.); (J.C.); (A.M.I.); (N.M.L.)
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
| | - Nora Marlene Lelivelt
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (D.H.); (J.-M.L.); (J.C.); (A.M.I.); (N.M.L.)
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA; (D.H.); (J.-M.L.); (J.C.); (A.M.I.); (N.M.L.)
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
- Correspondence:
| |
Collapse
|
50
|
Azuraini MJ, Vigneswari S, Huong KH, Khairul WM, H.P.S. AK, Ramakrishna S, Amirul AAA. Surface Modification of Sponge-like Porous Poly(3-hydroxybutyrate- co-4-hydroxybutyrate)/Gelatine Blend Scaffolds for Potential Biomedical Applications. Polymers (Basel) 2022; 14:1710. [PMID: 35566880 PMCID: PMC9104733 DOI: 10.3390/polym14091710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/05/2022] [Accepted: 04/12/2022] [Indexed: 12/04/2022] Open
Abstract
In this study, we described the preparation of sponge-like porous scaffolds that are feasible for medical applications. A porous structure provides a good microenvironment for cell attachment and proliferation. In this study, a biocompatible PHA, poly(3-hydroxybutyrate-co-4-hydroxybutyrate) was blended with gelatine to improve the copolymer's hydrophilicity, while structural porosity was introduced into the scaffold via a combination of solvent casting and freeze-drying techniques. Scanning electron microscopy results revealed that the blended scaffolds exhibited higher porosity when the 4HB compositions of P(3HB-co-4HB) ranged from 27 mol% to 50 mol%, but porosity decreased with a high 4HB monomer composition of 82 mol%. The pore size, water absorption capacity, and cell proliferation assay results showed significant improvement after the final weight of blend scaffolds was reduced by half from the initial 0.79 g to 0.4 g. The pore size of 0.79g-(P27mol%G10) increased three-fold while the water absorption capacity of 0.4g-(P50mol%G10) increased to 325%. Meanwhile, the cell proliferation and attachment of 0.4g-(P50mol%G10) and 0.4g-(P82mol%G7.5) increased as compared to the initial seeding number. Based on the overall data obtained, we can conclude that the introduction of a small amount of gelatine into P(3HB-co-4HB) improved the physical and biological properties of blend scaffolds, and the 0.4g-(P50mol%G10) shows great potential for medical applications considering its unique structure and properties.
Collapse
Affiliation(s)
- Mat Junoh Azuraini
- School of Biological Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (M.J.A.); (K.-H.H.)
| | - Sevakumaran Vigneswari
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Malaysia; (S.V.); (W.M.K.)
| | - Kai-Hee Huong
- School of Biological Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (M.J.A.); (K.-H.H.)
- Centre of Chemical Biology, Universiti Sains Malaysia, Penang 11900, Malaysia
- School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta, Midoriku, Yokohama 226-8501, Japan
| | - Wan M. Khairul
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Malaysia; (S.V.); (W.M.K.)
| | - Abdul Khalil H.P.S.
- School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia;
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, Center for Nanotechnology and Sustainability, National University of Singapore, Singapore 119260, Singapore;
| | - Al-Ashraf Abdullah Amirul
- School of Biological Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (M.J.A.); (K.-H.H.)
- Centre of Chemical Biology, Universiti Sains Malaysia, Penang 11900, Malaysia
- Malaysian Institute of Pharmaceuticals and Nutraceuticals, NIBM, Penang 11700, Malaysia
| |
Collapse
|