51
|
Heinz F, Proksch J, Schmidt RF, Gradzielski M, Koksch B, Keller BG. How Chromophore Labels Shape the Structure and Dynamics of a Peptide Hydrogel. Biomacromolecules 2024; 25:1262-1273. [PMID: 38288602 PMCID: PMC10865361 DOI: 10.1021/acs.biomac.3c01225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 02/13/2024]
Abstract
Biocompatible and functionalizable hydrogels have a wide range of (potential) medicinal applications. The hydrogelation process, particularly for systems with very low polymer weight percentages (<1 wt %), remains poorly understood, making it challenging to predict the self-assembly of a given molecular building block into a hydrogel. This severely hinders the rational design of self-assembled hydrogels. In this study, we demonstrate the impact of an N-terminal group on the self-assembly and rheology of the peptide hydrogel hFF03 (hydrogelating, fibril forming peptide 03) using molecular dynamics simulations, oscillatory shear rheology, and circular dichroism spectroscopy. We find that the chromophore and even its specific regioisomers have a significant influence on the microscopic structure and dynamics of the self-assembled fibril, and on the macroscopic mechanical properties. This is because the chromophore influences the possible salt bridges, which form and stabilize the fibril formation. Furthermore, we find that the solvation shell fibrils by itself cannot explain the viscoelasticity of hFF03 hydrogels. Our atomistic model of the hFF03 fibril formation enables a more rational design of these hydrogels. In particular, altering the N-terminal chromophore emerges as a design strategy to tune the mechanic properties of these self-assembled peptide hydrogels.
Collapse
Affiliation(s)
- Frederick Heinz
- Department
of Biology, Chemistry and Pharmacy, Freie
Universität Berlin, Arnimallee 22, Berlin 14195, Germany
| | - Jonas Proksch
- Department
of Biology, Chemistry and Pharmacy, Freie
Universität Berlin, Arnimallee 22, Berlin 14195, Germany
| | - Robert F. Schmidt
- Stranski-Laboratorium
für Physikalische und Theoretische Chemie, Institut für
Chemie, Technische Universität Berlin, Straße des 17. Juni 124, Berlin 10623, Germany
| | - Michael Gradzielski
- Stranski-Laboratorium
für Physikalische und Theoretische Chemie, Institut für
Chemie, Technische Universität Berlin, Straße des 17. Juni 124, Berlin 10623, Germany
| | - Beate Koksch
- Department
of Biology, Chemistry and Pharmacy, Freie
Universität Berlin, Arnimallee 22, Berlin 14195, Germany
| | - Bettina G. Keller
- Department
of Biology, Chemistry and Pharmacy, Freie
Universität Berlin, Arnimallee 22, Berlin 14195, Germany
| |
Collapse
|
52
|
Golebiowska AA, Intravaia JT, Sathe VM, Kumbar SG, Nukavarapu SP. Decellularized extracellular matrix biomaterials for regenerative therapies: Advances, challenges and clinical prospects. Bioact Mater 2024; 32:98-123. [PMID: 37927899 PMCID: PMC10622743 DOI: 10.1016/j.bioactmat.2023.09.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Tissue engineering and regenerative medicine have shown potential in the repair and regeneration of tissues and organs via the use of engineered biomaterials and scaffolds. However, current constructs face limitations in replicating the intricate native microenvironment and achieving optimal regenerative capacity and functional recovery. To address these challenges, the utilization of decellularized tissues and cell-derived extracellular matrix (ECM) has emerged as a promising approach. These biocompatible and bioactive biomaterials can be engineered into porous scaffolds and grafts that mimic the structural and compositional aspects of the native tissue or organ microenvironment, both in vitro and in vivo. Bioactive dECM materials provide a unique tissue-specific microenvironment that can regulate and guide cellular processes, thereby enhancing regenerative therapies. In this review, we explore the emerging frontiers of decellularized tissue-derived and cell-derived biomaterials and bio-inks in the field of tissue engineering and regenerative medicine. We discuss the need for further improvements in decellularization methods and techniques to retain structural, biological, and physicochemical characteristics of the dECM products in a way to mimic native tissues and organs. This article underscores the potential of dECM biomaterials to stimulate in situ tissue repair through chemotactic effects for the development of growth factor and cell-free tissue engineering strategies. The article also identifies the challenges and opportunities in developing sterilization and preservation methods applicable for decellularized biomaterials and grafts and their translation into clinical products.
Collapse
Affiliation(s)
| | - Jonathon T. Intravaia
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
| | - Vinayak M. Sathe
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| | - Syam P. Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, 06269, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, 06032, USA
| |
Collapse
|
53
|
Carrara SC, Davila-Lezama A, Cabriel C, Berenschot EJ, Krol S, Gardeniers J, Izeddin I, Kolmar H, Susarrey-Arce A. 3D topographies promote macrophage M2d-Subset differentiation. Mater Today Bio 2024; 24:100897. [PMID: 38169974 PMCID: PMC10758855 DOI: 10.1016/j.mtbio.2023.100897] [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: 07/19/2023] [Revised: 11/11/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
In vitro cellular models denote a crucial part of drug discovery programs as they aid in identifying successful drug candidates based on their initial efficacy and potency. While tremendous headway has been achieved in improving 2D and 3D culture techniques, there is still a need for physiologically relevant systems that can mimic or alter cellular responses without the addition of external biochemical stimuli. A way forward to alter cellular responses is using physical cues, like 3D topographical inorganic substrates, to differentiate macrophage-like cells. Herein, protein secretion and gene expression markers for various macrophage subsets cultivated on a 3D topographical substrate are investigated. The results show that macrophages differentiate into anti-inflammatory M2-type macrophages, secreting increased IL-10 levels compared to the controls. Remarkably, these macrophage cells are differentiated into the M2d subset, making up the main component of tumour-associated macrophages (TAMs), as measured by upregulated Il-10 and Vegf mRNA. M2d subset differentiation is attributed to the topographical substrates with 3D fractal-like geometries arrayed over the surface, else primarily achieved by tumour-associated factors in vivo. From a broad perspective, this work paves the way for implementing 3D topographical inorganic surfaces for drug discovery programs, harnessing the advantages of in vitro assays without external stimulation and allowing the rapid characterisation of therapeutic modalities in physiologically relevant environments.
Collapse
Affiliation(s)
- Stefania C. Carrara
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Amanda Davila-Lezama
- Facultad de Ciencias de la Salud (FACISALUD), Universidad Autónoma de Baja California, Blvd. Universitario 1000, Valle de las Palmas, 22260 Tijuana, Mexico
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, P.O. Box 217, 7500AE Enschede, the Netherlands
| | - Clément Cabriel
- Institut Langevin, ESPCI Paris, CNRS, Université PSL, 75005 Paris, France
| | - Erwin J.W. Berenschot
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, P.O. Box 217, 7500AE Enschede, the Netherlands
| | - Silke Krol
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, P.O. Box 217, 7500AE Enschede, the Netherlands
- Encytos B.V., Piet Heinstraat 12, Enschede, the Netherlands
| | - J.G.E. Gardeniers
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, P.O. Box 217, 7500AE Enschede, the Netherlands
| | - Ignacio Izeddin
- Institut Langevin, ESPCI Paris, CNRS, Université PSL, 75005 Paris, France
| | - Harald Kolmar
- Institute for Organic Chemistry and Biochemistry, Technical University of Darmstadt, Alarich-Weiss-Strasse 4, D-64287 Darmstadt, Germany
- Centre for Synthetic Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Arturo Susarrey-Arce
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, P.O. Box 217, 7500AE Enschede, the Netherlands
| |
Collapse
|
54
|
Wen F, Wang Y, Tu B, Cui L. Superfast Gelation of Spider Silk-Based Artificial Silk Protein. Gels 2024; 10:69. [PMID: 38247791 PMCID: PMC10815891 DOI: 10.3390/gels10010069] [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: 11/20/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
Spider silk proteins (spidroins) have garnered attention in biomaterials research due to their ability to self-assemble into hydrogels. However, reported spidroin hydrogels require high protein concentration and prolonged gelation time. Our study engineered an artificial spidroin that exhibits unprecedented rapid self-assembly into hydrogels at physiologically relevant conditions, achieving gelation at a low concentration of 6 mg/mL at 37 °C without external additives. Remarkably, at a 30 mg/mL concentration, our engineered protein forms hydrogels within 30 s, a feature we termed "superfast gelation". This rapid formation is modulated by ions, pH, and temperature, offering versatility in biomedical applications. The hydrogel's capacity to encapsulate proteins and support E. coli growth while inducing RFP expression provides a novel platform for drug delivery and bioengineering applications. Our findings introduce a superfast, highly adaptable, and cytocompatible hydrogel that self-assembles under mild conditions, underscoring the practical implication of rapid gelation in biomedical research and clinical applications.
Collapse
Affiliation(s)
- Fan Wen
- CCZU-JITRI Joint Bio-X Lab, School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, China
| | - Yu Wang
- CCZU-JITRI Joint Bio-X Lab, School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, China
| | - Bowen Tu
- Pathogenic Biological Laboratory, Changzhou Disease Control and Prevention Centre, Changzhou Medical Centre, Nanjing Medical University, Changzhou 213000, China
| | - Lun Cui
- CCZU-JITRI Joint Bio-X Lab, School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, China
- Biomaterials Lab, Changzhou AiRiBio Healthcare Co., Ltd., Changzhou 213164, China
| |
Collapse
|
55
|
Ladero M, Reche-Sainz JA, Gallardo ME. Hereditary Optic Neuropathies: A Systematic Review on the Interplay between Biomaterials and Induced Pluripotent Stem Cells. Bioengineering (Basel) 2024; 11:52. [PMID: 38247929 PMCID: PMC10813088 DOI: 10.3390/bioengineering11010052] [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: 11/27/2023] [Revised: 12/29/2023] [Accepted: 12/30/2023] [Indexed: 01/23/2024] Open
Abstract
Hereditary optic neuropathies (HONs) such as dominant optic atrophy (DOA) and Leber Hereditary Optic Neuropathy (LHON) are mitochondrial diseases characterized by a degenerative loss of retinal ganglion cells (RGCs) and are a cause of blindness worldwide. To date, there are only limited disease-modifying treatments for these disorders. The discovery of induced pluripotent stem cell (iPSC) technology has opened several promising opportunities in the field of HON research and the search for therapeutic approaches. This systematic review is focused on the two most frequent HONs (LHON and DOA) and on the recent studies related to the application of human iPSC technology in combination with biomaterials technology for their potential use in the development of RGC replacement therapies with the final aim of the improvement or even the restoration of the vision of HON patients. To this purpose, the combination of natural and synthetic biomaterials modified with peptides, neurotrophic factors, and other low- to medium-molecular weight compounds, mimicking the ocular extracellular matrices, with human iPSC or iPSC-derived cell retinal progenitors holds enormous potential to be exploited in the near future for the generation of transplantable RGC populations.
Collapse
Affiliation(s)
- Miguel Ladero
- FQPIMA Group, Materials and Chemical Engineering Department, Chemical Sciences School, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jose Alberto Reche-Sainz
- Ophthalmology Unit, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
- Translational Research with iPS Cells Group, Research Institute of Hospital 12 de Octubre, imas12, 28041 Madrid, Spain
| | - M. Esther Gallardo
- Translational Research with iPS Cells Group, Research Institute of Hospital 12 de Octubre, imas12, 28041 Madrid, Spain
| |
Collapse
|
56
|
Huang J, Liu Y, Wu J, Dong F, Liu C, Luo J, Liu X, Wang N, Wang L, Xu H. An extracellular matrix-mimicking magnetic microrobot for targeted elimination of circulating cancer cells. NANOSCALE 2024; 16:624-634. [PMID: 38086673 DOI: 10.1039/d3nr03799a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2024]
Abstract
Cancer cells disseminate through the bloodstream, leading to metastasis in distant sites within the body. One promising strategy to prevent metastasis is to eliminate circulating tumor cells. However, this remains challenging due to the lack of an active and targeted biomedical tool for efficient cancer cell elimination. Here, we developed a magnetic microrobot by using natural materials derived from the extracellular matrix (ECM) to mimic the ligand-receptor interaction between cancer cells and the ECM, offering targeted elimination of cancer cells. The ECM-mimicking microrobot is designed with a biodegradable hydrogel matrix, incorporating a cancer cell ligand and magnetic microparticles for cancer cell capture and active locomotion. This microrobot was fabricated based on an interface-shearing method, enabling controllable magnetic response and size scalability (30 μm-500 μm). The presented ECM-mimicking microrobot can actively approach and capture single cancer cells and cell clusters under the control of specific magnetic fields. The experiment was conducted in a blood vessel-mimicking simulator. The microrobot demonstrates an outstanding elimination efficacy of 92.3% on MDA-MB-231 cancer cells and a stable transport capability of the captured cells over long distances to a designed recycling site, inhibiting cell metastasis. This magnetic ECM-mimicking microrobot based on a bioinspired binding mechanism represents a promising candidate for the efficient elimination of cancer cells and other biological waste in the blood.
Collapse
Affiliation(s)
- Jing Huang
- Department of Polymer Materials and Engineering, College of Materials and Metallurgy, Guizhou University, Guiyang, 550025, China.
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China.
| | - Yuan Liu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China.
| | - Jiandong Wu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China.
| | - Fuping Dong
- Department of Polymer Materials and Engineering, College of Materials and Metallurgy, Guizhou University, Guiyang, 550025, China.
| | - Chu Liu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China.
| | - Jiawei Luo
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China.
| | - Xiangchao Liu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China.
| | - Ning Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China.
| | - Lei Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China.
| | - Haifeng Xu
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, 518055, China.
| |
Collapse
|
57
|
Khan MUA, Stojanović GM, Abdullah MFB, Dolatshahi-Pirouz A, Marei HE, Ashammakhi N, Hasan A. Fundamental properties of smart hydrogels for tissue engineering applications: A review. Int J Biol Macromol 2024; 254:127882. [PMID: 37951446 DOI: 10.1016/j.ijbiomac.2023.127882] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 10/22/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023]
Abstract
Tissue engineering is an advanced and potential biomedical approach to treat patients suffering from lost or failed an organ or tissue to repair and regenerate damaged tissues that increase life expectancy. The biopolymers have been used to fabricate smart hydrogels to repair damaged tissue as they imitate the extracellular matrix (ECM) with intricate structural and functional characteristics. These hydrogels offer desired and controllable qualities, such as tunable mechanical stiffness and strength, inherent adaptability and biocompatibility, swellability, and biodegradability, all crucial for tissue engineering. Smart hydrogels provide a superior cellular environment for tissue engineering, enabling the generation of cutting-edge synthetic tissues due to their special qualities, such as stimuli sensitivity and reactivity. Numerous review articles have presented the exceptional potential of hydrogels for various biomedical applications, including drug delivery, regenerative medicine, and tissue engineering. Still, it is essential to write a comprehensive review article on smart hydrogels that successfully addresses the essential challenging issues in tissue engineering. Hence, the recent development on smart hydrogel for state-of-the-art tissue engineering conferred progress, highlighting significant challenges and future perspectives. This review discusses recent advances in smart hydrogels fabricated from biological macromolecules and their use for advanced tissue engineering. It also provides critical insight, emphasizing future research directions and progress in tissue engineering.
Collapse
Affiliation(s)
- Muhammad Umar Aslam Khan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar.
| | - Goran M Stojanović
- Department of Electronics, Faculty of Technical Sciences, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Mohd Faizal Bin Abdullah
- Oral and Maxillofacial Surgery Unit, School of Dental Sciences, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kota Bharu, Kelantan, Malaysia; Oral and Maxillofacial Surgery Unit, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Health Campus, 16150, Kubang Kerian, Kota Bharu, Kelantan, Malaysia.
| | | | - Hany E Marei
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering (IQ), Department of Biomedical Engineering, College of Engineering and Human Medicine, Michigan State University, East Lansing, MI 48824, USA.
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar
| |
Collapse
|
58
|
Santilli F, Fabrizi J, Santacroce C, Caissutti D, Spinello Z, Candelise N, Lancia L, Pulcini F, Delle Monache S, Mattei V. Analogies and Differences Between Dental Stem Cells: Focus on Secretome in Combination with Scaffolds in Neurological Disorders. Stem Cell Rev Rep 2024; 20:159-174. [PMID: 37962698 PMCID: PMC10799818 DOI: 10.1007/s12015-023-10652-9] [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] [Accepted: 11/02/2023] [Indexed: 11/15/2023]
Abstract
Mesenchymal stem cells (MSCs) are well known for their beneficial effects, differentiation capacity and regenerative potential. Dental-derived MSCs (DSCs) are more easily accessible and have a non-invasive isolation method rather than MSCs isolated from other sources (umbilical cord, bone marrow, and adipose tissue). In addition, DSCs appear to have a relevant neuro-regenerative potential due to their neural crest origin. However, it is now known that the beneficial effects of MSCs depend, at least in part, on their secretome, referring to all the bioactive molecules (neurotrophic factors) released in the conditioned medium (CM) or in the extracellular vesicles (EVs) in particular exosomes (Exos). In this review, we described the similarities and differences between various DSCs. Our focus was on the secretome of DSCs and their applications in cell therapy for neurological disorders. For neuro-regenerative purposes, the secretome of different DSCs has been tested. Among these, the secretome of dental pulp stem cells and stem cells from human exfoliated deciduous teeth have been the most widely studied. Both CM and Exos obtained from DSCs have been shown to promote neurite outgrowth and neuroprotective effects as well as their combination with scaffold materials (to improve their functional integration in the tissue). For these reasons, the secretome obtained from DSCs in combination with scaffold materials may represent a promising tissue engineering approach for neuroprotective and neuro-regenerative treatments.
Collapse
Affiliation(s)
- Francesca Santilli
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Via A.M. Ricci 35/A, 02100, Rieti, Italy
| | - Jessica Fabrizi
- Department of Experimental Medicine, "Sapienza" University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Costantino Santacroce
- Biomedicine and Advanced Technologies Rieti Center, "Sabina Universitas", Via A.M. Ricci 35/A, 02100, Rieti, Italy
| | - Daniela Caissutti
- Department of Experimental Medicine, "Sapienza" University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Zaira Spinello
- Department of Experimental Medicine, "Sapienza" University, Viale Regina Elena 324, 00161, Rome, Italy
| | - Niccolò Candelise
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Viale Regina Elena, 29900161, Rome, Italy
| | - Loreto Lancia
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy
| | - Fanny Pulcini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy
| | - Simona Delle Monache
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, Via Vetoio, 67100, L'Aquila, Italy.
| | - Vincenzo Mattei
- Dipartimento di Scienze della Vita, della Salute e delle Professioni Sanitarie, Link Campus University, Via del Casale di San Pio V 44, 00165, Rome, Italy.
| |
Collapse
|
59
|
Aazmi A, Zhang D, Mazzaglia C, Yu M, Wang Z, Yang H, Huang YYS, Ma L. Biofabrication methods for reconstructing extracellular matrix mimetics. Bioact Mater 2024; 31:475-496. [PMID: 37719085 PMCID: PMC10500422 DOI: 10.1016/j.bioactmat.2023.08.018] [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: 05/09/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023] Open
Abstract
In the human body, almost all cells interact with extracellular matrices (ECMs), which have tissue and organ-specific compositions and architectures. These ECMs not only function as cellular scaffolds, providing structural support, but also play a crucial role in dynamically regulating various cellular functions. This comprehensive review delves into the examination of biofabrication strategies used to develop bioactive materials that accurately mimic one or more biophysical and biochemical properties of ECMs. We discuss the potential integration of these ECM-mimics into a range of physiological and pathological in vitro models, enhancing our understanding of cellular behavior and tissue organization. Lastly, we propose future research directions for ECM-mimics in the context of tissue engineering and organ-on-a-chip applications, offering potential advancements in therapeutic approaches and improved patient outcomes.
Collapse
Affiliation(s)
- Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Duo Zhang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 51817, China
| | - Corrado Mazzaglia
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
60
|
Mauduit O, Delcroix V, Wong A, Ivanova A, Miles L, Lee HS, Makarenkova H. A closer look into the cellular and molecular biology of myoepithelial cells across various exocrine glands. Ocul Surf 2024; 31:63-80. [PMID: 38141817 PMCID: PMC10855576 DOI: 10.1016/j.jtos.2023.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/02/2023] [Accepted: 12/09/2023] [Indexed: 12/25/2023]
Abstract
Myoepithelial cells (MECs) are a unique subset of epithelial cells that possess several smooth muscle cell characteristics, such as a high number of actin-myosin filaments and the ability to contract. These cells are primarily located around the secretory cells of exocrine glands, including the salivary, mammary, lacrimal, and sweat glands. Their primary functions involve the construction of the basement membrane and help with secretion of gland products through contraction. So far, no comparative analysis of MECs in different exocrine glands had ever evaluated their differences. In this review, we took advantage of the various publicly available scRNAseq data from mouse exocrine glands to identify their shared and unique characteristics. The aim of this review is to compare the role of MECs in maintaining healthy glandular function, their involvement in disease states, and their regenerative capacity, with a particular emphasis on the latest research findings in these areas.
Collapse
Affiliation(s)
- Olivier Mauduit
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Vanessa Delcroix
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Andrew Wong
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Anastasiia Ivanova
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Lindsey Miles
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Hyun Soo Lee
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA; Department of Ophthalmology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Helen Makarenkova
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
61
|
Anjum S, Li T, Saeed M, Ao Q. Exploring polysaccharide and protein-enriched decellularized matrix scaffolds for tendon and ligament repair: A review. Int J Biol Macromol 2024; 254:127891. [PMID: 37931866 DOI: 10.1016/j.ijbiomac.2023.127891] [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: 07/17/2023] [Revised: 10/07/2023] [Accepted: 11/02/2023] [Indexed: 11/08/2023]
Abstract
Tissue engineering (TE) has become a primary research topic for the treatment of diseased or damaged tendon/ligament (T/L) tissue. T/L injuries pose a severe clinical burden worldwide, necessitating the development of effective strategies for T/L repair and tissue regeneration. TE has emerged as a promising strategy for restoring T/L function using decellularized extracellular matrix (dECM)-based scaffolds. dECM scaffolds have gained significant prominence because of their native structure, relatively high bioactivity, low immunogenicity, and ability to function as scaffolds for cell attachment, proliferation, and differentiation, which are difficult to imitate using synthetic materials. Here, we review the recent advances and possible future prospects for the advancement of dECM scaffolds for T/L tissue regeneration. We focus on crucial scaffold properties and functions, as well as various engineering strategies employed for biomaterial design in T/L regeneration. dECM provides both the physical and mechanical microenvironments required by cells to survive and proliferate. Various decellularization methods and sources of allogeneic and xenogeneic dECM in T/L repair and regeneration are critically discussed. Additionally, dECM hydrogels, bio-inks in 3D bioprinting, and nanofibers are briefly explored. Understanding the opportunities and challenges associated with dECM-based scaffold development is crucial for advancing T/L repairs in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Shabnam Anjum
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang 110122, China; 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 610064, China
| | - Ting Li
- Department of Laboratory Medicine, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Mohammad Saeed
- Dr. A.P.J Abdul Kalam Technical University, Lucknow 226031, India
| | - Qiang Ao
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, Shenyang 110122, China; 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 610064, China.
| |
Collapse
|
62
|
Molina-Peña R, Ferreira NH, Roy C, Roncali L, Najberg M, Avril S, Zarur M, Bourgeois W, Ferreirós A, Lucchi C, Cavallieri F, Hindré F, Tosi G, Biagini G, Valzania F, Berger F, Abal M, Rousseau A, Boury F, Alvarez-Lorenzo C, Garcion E. Implantable SDF-1α-loaded silk fibroin hyaluronic acid aerogel sponges as an instructive component of the glioblastoma ecosystem: Between chemoattraction and tumor shaping into resection cavities. Acta Biomater 2024; 173:261-282. [PMID: 37866725 DOI: 10.1016/j.actbio.2023.10.022] [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: 05/25/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
In view of inevitable recurrences despite resection, glioblastoma (GB) is still an unmet clinical need. Dealing with the stromal-cell derived factor 1-alpha (SDF-1α)/CXCR4 axis as a hallmark of infiltrative GB tumors and with the resection cavity situation, the present study described the effects and relevance of a new engineered micro-nanostructured SF-HA-Hep aerogel sponges, made of silk fibroin (SF), hyaluronic acid (HA) and heparin (Hep) and loaded with SDF-1α, to interfere with the GB ecosystem and residual GB cells, attracting and confining them in a controlled area before elimination. 70 µm-pore sponges were designed as an implantable scaffold to trap GB cells. They presented shape memory and fit brain cavities. Histological results after implantation in brain immunocompetent Fischer rats revealed that SF-HA-Hep sponges are well tolerated for more than 3 months while moderately and reversibly colonized by immuno-inflammatory cells. The use of human U87MG GB cells overexpressing the CXCR4 receptor (U87MG-CXCR4+) and responding to SDF-1α allowed demonstrating directional GB cell attraction and colonization of the device in vitro and in vivo in orthotopic resection cavities in Nude rats. Not modifying global survival, aerogel sponge implantation strongly shaped U87MG-CXCR4+ tumors in cavities in contrast to random infiltrative growth in controls. Overall, those results support the interest of SF-HA-Hep sponges as modifiers of the GB ecosystem dynamics acting as "cell meeting rooms" and biocompatible niches whose properties deserve to be considered toward the development of new clinical procedures. STATEMENT OF SIGNIFICANCE: Brain tumor glioblastoma (GB) is one of the worst unmet clinical needs. To prevent the relapse in the resection cavity situation, new implantable biopolymer aerogel sponges loaded with a chemoattractant molecule were designed and preclinically tested as a prototype targeting the interaction between the initial tumor location and its attraction by the peritumoral environment. While not modifying global survival, biocompatible SDF1-loaded hyaluronic acid and silk fibroin sponges induce directional GB cell attraction and colonization in vitro and in rats in vivo. Interestingly, they strongly shaped GB tumors in contrast to random infiltrative growth in controls. These results provide original findings on application of exogenous engineered niches that shape tumors and serve as cell meeting rooms for further clinical developments.
Collapse
Affiliation(s)
- Rodolfo Molina-Peña
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, F-49000 Angers, France
| | | | - Charlotte Roy
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, F-49000 Angers, France
| | - Loris Roncali
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, F-49000 Angers, France
| | - Mathie Najberg
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, F-49000 Angers, France
| | - Sylvie Avril
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, F-49000 Angers, France
| | - Mariana Zarur
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, ID Farma (GI-1645), Facultad de Farmacia, iMATUS, and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - William Bourgeois
- Inserm UMR1205, Brain Tech Lab, Grenoble Alpes University Hospital (CHUGA), Grenoble, 38000, France
| | - Alba Ferreirós
- NASASBIOTECH S.L., Cantón Grande nº 9, 15003, A Coruña, Spain
| | - Chiara Lucchi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Francesco Cavallieri
- Neurology Unit, Neuromotor and Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - François Hindré
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, F-49000 Angers, France
| | - Giovani Tosi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Giuseppe Biagini
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Franco Valzania
- Neurology Unit, Neuromotor and Rehabilitation Department, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - François Berger
- Inserm UMR1205, Brain Tech Lab, Grenoble Alpes University Hospital (CHUGA), Grenoble, 38000, France
| | - Miguel Abal
- NASASBIOTECH S.L., Cantón Grande nº 9, 15003, A Coruña, Spain
| | - Audrey Rousseau
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, F-49000 Angers, France
| | - Frank Boury
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, F-49000 Angers, France
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, ID Farma (GI-1645), Facultad de Farmacia, iMATUS, and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain.
| | - Emmanuel Garcion
- Univ Angers, Nantes Université, Inserm, CNRS, CRCI2NA, SFR ICAT, F-49000 Angers, France.
| |
Collapse
|
63
|
Enoch K, Somasundaram AA. Rheological insights on Carboxymethyl cellulose hydrogels. Int J Biol Macromol 2023; 253:127481. [PMID: 37865366 DOI: 10.1016/j.ijbiomac.2023.127481] [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: 06/08/2023] [Revised: 10/05/2023] [Accepted: 10/15/2023] [Indexed: 10/23/2023]
Abstract
Hydrogels are copiously studied for tissue engineering, drug delivery, and bone regeneration owing to their water content, mechanical strength, and elastic behaviour. The preparation of stable and mechanically strengthened hydrogels without using toxic crosslinkers and expensive approaches is immensely challenging. In this study, we prepared Carboxymethyl cellulose based hydrogels with different polymer concentration via a less expensive physical crosslinking approach without using any toxic crosslinkers and evaluated their mechanical strength. In this hydrogel system, the carbopol concentration was fixed at 1 wt/v% and the Carboxymethyl cellulose concentration was varied between 1 and 5 wt/v%. In this hydrogel system, Carbopol serves as the crosslinker to bridge Carboxymethyl cellulose polymer through hydrogen bonds. Rheological analysis was employed in assessing the mechanical properties of the prepared hydrogel, in particular, the viscoelastic behaviour of the hydrogels. The viscoelastic nature and mechanical strength of the hydrogels increased with an increase in the Carboxymethyl cellulose polymer concentration. Further, our results suggested that gels with Carboxymethyl cellulose concentration between 3 wt/v % and 4 wt/v % with yield stresses of 58.83 Pa and 81.47 Pa, respectively, are potential candidates for use in transdermal drug delivery. The prepared hydrogels possessed high thermal stability and retained their gel network structure even at 50 °C. These findings are beneficial for biomedical applications in transdermal drug delivery and tissue engineering owing to the biocompatibility, stability, and mechanical strength of the prepared hydrogels.
Collapse
Affiliation(s)
- Karolinekersin Enoch
- Soft Matter Laboratory, Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur - 603203, Tamil Nadu, India
| | - Anbumozhi Angayarkanni Somasundaram
- Soft Matter Laboratory, Department of Physics and Nanotechnology, SRM Institute of Science and Technology, Kattankulathur - 603203, Tamil Nadu, India.
| |
Collapse
|
64
|
Kim JH, Lee HJ, Song HJ, Park JB. Impact of 17β-Estradiol on the Shape, Survival, Osteogenic Transformation, and mRNA Expression of Gingiva-Derived Stem Cell Spheroids. MEDICINA (KAUNAS, LITHUANIA) 2023; 60:60. [PMID: 38256321 PMCID: PMC10817649 DOI: 10.3390/medicina60010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 12/05/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024]
Abstract
Background and Objectives: Mesenchymal stem cells hold promise for tissue regeneration, given their robust growth and versatile differentiation capabilities. An analysis of bone marrow-sourced mesenchymal stem cell proliferation showed that 17β-estradiol could enhance their growth. This study aims to investigate the influence of 17β-estradiol on the shape, survival, osteogenic differentiation, and mineralization of human mesenchymal stem cells. Materials and Methods: Spheroids made from human gingiva-derived stem cells were cultivated with varying concentrations of 17β-estradiol: 0, 0.01, 0.1, 1, and 10 nM. Morphology was assessed on days 1, 3, and 5. The live/dead kit assay was employed on day 3 for qualitative cell viability, while cell counting kit-8 was used for quantitative viability assessments on days 1, 3, and 5. To evaluate the osteogenic differentiation of the spheroids, a real-time polymerase chain reaction assessed the expressions of RUNX2 and COL1A1 on day 7. Results: The stem cells formed cohesive spheroids, and the inclusion of 17β-estradiol did not noticeably alter their shape. The spheroid diameter remained consistent across concentrations of 0, 0.01, 0.1, 1, and 10 nM of 17β-estradiol. However, cellular viability was boosted with the addition of 1 and 10 nM of 17β-estradiol. The highest expression levels for RUNX2 and COL1A1 were observed with the introduction of 17β-estradiol at 0.1 nM. Conclusions: In conclusion, from the results obtained, it can be inferred that 17β-estradiol can be utilized for differentiating stem cell spheroids. Furthermore, the localized and controlled use, potentially through localized delivery systems or biomaterials, can be an area of active research. While 17β-estradiol holds promise for enhancing stem cell applications, any clinical use requires a thorough understanding of its mechanisms, careful control of its dosage and delivery, and extensive testing to ensure safety and efficacy.
Collapse
Affiliation(s)
- Ju-Hwan Kim
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-H.K.); (H.-J.L.)
| | - Hyun-Jin Lee
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-H.K.); (H.-J.L.)
| | - Hye-Jung Song
- Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (J.-H.K.); (H.-J.L.)
- Dental Implantology, Graduate School of Clinical Dental Science, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Department of Medicine, Graduate School, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
65
|
Taborda M, Catalan KN, Orellana N, Bezjak D, Enrione J, Acevedo CA, Corrales TP. Micropatterned Nanofiber Scaffolds of Salmon Gelatin, Chitosan, and Poly(vinyl alcohol) for Muscle Tissue Engineering. ACS OMEGA 2023; 8:47883-47896. [PMID: 38144088 PMCID: PMC10733945 DOI: 10.1021/acsomega.3c06436] [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: 08/28/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 12/26/2023]
Abstract
The development of scaffolds that mimic the aligned fibrous texture of the extracellular matrix has become an important requirement in muscle tissue engineering. Electrospinning is a widely used technique to fabricate biomimetic scaffolds. Therefore, a biopolymer blend composed of salmon gelatin (SG), chitosan (Ch), and poly(vinyl alcohol) (PVA) was developed by electrospinning onto a micropatterned (MP) collector, resulting in a biomimetic scaffold for seeding muscle cells. Rheology and surface tension studies were performed to determine the optimum solution concentration and viscosity for electrospinning. The scaffold microstructure was analyzed using SEM to determine the nanofiber's diameter and orientation. Blends of SG/Ch/PVA exhibited better electrospinnability and handling properties than pure PVA. The resulting scaffolds consist of a porous surface (∼46%), composed of a random fiber distribution, for a flat collector and scaffolds with regions of aligned nanofibers for the MP collector. The nanofiber diameters are 141 ± 2 and 151 ± 2 nm for the flat and MP collector, respectively. In vitro studies showed that myoblasts cultured on scaffold SG/Ch/PVA presented a high rate of cell growth. Furthermore, the aligned nanofibers on the SG/Ch/PVA scaffold provide a suitable platform for myoblast alignment.
Collapse
Affiliation(s)
- María
I. Taborda
- Centro
de Biotecnología, Universidad Técnica
Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
- Programa
de doctorado en Biotecnología, Pontificia
Universidad Católica de Valparaíso−Universidad
Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
| | - Karina N. Catalan
- Departamento
de Física, Universidad Técnica
Federico Santa María, Av. España 1680, Valparaíso 2340000, Chile
| | - Nicole Orellana
- Centro
de Biotecnología, Universidad Técnica
Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
| | - Dragica Bezjak
- Centro
de Biotecnología, Universidad Técnica
Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
- Programa
de doctorado en Biotecnología, Pontificia
Universidad Católica de Valparaíso−Universidad
Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
| | - Javier Enrione
- Escuela
de Nutrición y Dietética, Facultad de Medicina, Universidad de los Andes, Monseñor Álvaro del Portillo 12455, Las Condes, Santiago 7550000, Chile
| | - Cristian A. Acevedo
- Centro
de Biotecnología, Universidad Técnica
Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
- Departamento
de Física, Universidad Técnica
Federico Santa María, Av. España 1680, Valparaíso 2340000, Chile
- Centro
Científico Tecnológico de Valparaíso (CCTVAL), Universidad Técnica Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
| | - Tomas P. Corrales
- Centro
de Biotecnología, Universidad Técnica
Federico Santa María, Avenida España 1680, Valparaíso 2340000, Chile
- Departamento
de Física, Universidad Técnica
Federico Santa María, Av. España 1680, Valparaíso 2340000, Chile
- Millenium
Nucleus in NanoBioPhysics (NNBP), Valparaíso 2340000, Chile
| |
Collapse
|
66
|
Semitela Â, Pinto SC, Capitão A, Marques PAAP, Completo A. Fabrication of Customizable and Reproducible 3D Chondrocyte-Laden Nanofibrous Architectures: Effect of Specific Fiber Alignments and Porosities on Chondrocyte Response under Cyclic Compression. ACS APPLIED BIO MATERIALS 2023; 6:5541-5554. [PMID: 37947854 DOI: 10.1021/acsabm.3c00737] [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: 11/12/2023]
Abstract
Electrospinning has been widely employed to fabricate complex extracellular matrix-like microenvironments for tissue engineering due to its ability to replicate structurally biomimetic micro- and nanotopographic cues. Nevertheless, these nanofibrous structures are typically either confined to bidimensional systems or confined to three-dimensional ones that are unable to provide controlled multiscale patterns. Thus, an electrospinning modality was used in this work to fabricate chondrocyte-laden nanofibrous scaffolds with highly customizable three-dimensional (3D) architectures in an automated manner, with the ultimate goal of recreating a suitable 3D scaffold for articular cartilage tissue engineering. Three distinct architectures were designed and fabricated by combining multiple nanofibrous and chondrocyte-laden hydrogel layers and tested in vitro in a compression bioreactor system. Results demonstrated that it was possible to precisely control the placement and alignment of electrospun polycaprolactone and gelatin nanofibers, generating three unique architectures with distinctive macroscale porosity, water absorption capacity, and mechanical properties. The architecture organized in a lattice-like fashion was highly porous with substantial pore interconnectivity, resulting in a high-water absorption capacity but a poor compression modulus and relatively weaker energy dissipation capacity. The donut-like 3D geometry was the densest, with lower swelling, but the highest compression modulus and improved energy dissipation ability. The third architecture combined a lattice and donut-like fibrous arrangement, exhibiting intermediary behavior in terms of porosity, water absorption, compression modulus, and energy dissipation capacity. The properties of the donut-like 3D architecture demonstrated great potential for articular cartilage tissue engineering, as it mimicked key topographic, chemical, and mechanical characteristics of chondrocytes' surrounding environment. In fact, the combination of these architectural features with a dynamically compressive mechanical stimulus triggered the best in vitro results in terms of viability and biosynthetic production.
Collapse
Affiliation(s)
- Ângela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Susana C Pinto
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Ana Capitão
- Centre for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal
| |
Collapse
|
67
|
Liu Y, Liu X, Zhang Y, Cao Y, Luo B, Wang Z, Pei R. Interpenetrating Polymer Network HA/Alg-RGD Hydrogel: An Equilibrium of Macroscopic Stability and Microscopic Adaptability for 3D Cell Growth and Vascularization. Biomacromolecules 2023; 24:5977-5988. [PMID: 37939799 DOI: 10.1021/acs.biomac.3c01022] [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: 11/10/2023]
Abstract
Two-dimensional (2D) cell culture methods dominate the current research. However, the inherent responsiveness of cells to their native three-dimensional (3D) microenvironment necessitates a paradigm shift toward the development of advanced hydrogels that faithfully mimic the intricacies of the extracellular matrix (ECM) and enable continuous cell-ECM interactions. To address the constraints of traditional static hydrogel networks that impede effective cell-matrix and cell-cell interactions, and to tackle the inherent stability issues associated with dynamically cross-linked hydrogels, which have become a pressing concern. Herein, we present an interpenetrating polymer network (IPN) hydrogel (HA/Alg-RGD hydrogel) that combines a physically cross-linked network between alginate and calcium ions (Alg-Ca2+) for the enhanced cell growth adaptability with a chemically cross-linked hyaluronic acid (HA) network to ensure macroscopic stability during cell culture. The incorporation of arginine-glycine-aspartic peptide modified alginate (Alg-RGD) further facilitates cell adhesion and improves the cell-hydrogel interaction. Notably, this IPN hydrogel demonstrates mechanical stability and enables cell spreading and growth within its structural framework. Leveraging the reversible characteristics of the ionically cross-linked Alg-Ca2+ network within IPN hydrogels, we demonstrate the feasibility of the gelatin sacrificial solution for 3D printing purposes within the hydrogel matrix. Subsequent UV-induced covalent cross-linking enables the fabrication of vascularized microfluidic channels within the resulting construct. Our results demonstrate endothelial cell spreading and spontaneous cell sprouting within the hydrogel matrix, thus highlighting the efficacy of this IPN hydrogel system in facilitating 3D cell growth. Additionally, our study emphasizes the potential of 3D printed constructs as a promising approach for vascularization in tissue engineering. The importance of RGD peptides in promoting favorable cell-hydrogel scaffold interactions is also highlighted, emphasizing their critical role in optimizing biomaterial-cell interfaces.
Collapse
Affiliation(s)
- Yuanshan Liu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xingzhu Liu
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yajie Zhang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yi Cao
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Bingqing Luo
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Zheng Wang
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory for Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| |
Collapse
|
68
|
Ghasemzadeh-Hasankolaei M, Pinto CA, Jesus D, Saraiva JA, Mano JF. Effect of high cyclic hydrostatic pressure on osteogenesis of mesenchymal stem cells cultured in liquefied micro-compartments. Mater Today Bio 2023; 23:100861. [PMID: 38058695 PMCID: PMC10696388 DOI: 10.1016/j.mtbio.2023.100861] [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: 02/23/2023] [Revised: 11/04/2023] [Accepted: 11/09/2023] [Indexed: 12/08/2023] Open
Abstract
Bone resident cells are constantly subjected to a range of distinct mechanical loadings, which generates a complex microenvironment. In particular, hydrostatic pressure (HP) has a key impact on modulation of cell function and fate determination. Although HP is a constant mechanical stimulus, its role in regulating the osteogenesis process within a defined 3D microenvironment has not been comprehensively elucidated. Perceiving how environmental factors regulate the differentiation of stem cells is essential for expanding their regenerative potential. Inspired by the mechanical environment of bone, this study attempted to investigate the influence of different ranges of cyclic HP on human adipose-derived mesenchymal stem cells (MSCs) encapsulated within a compartmentalized liquefied microenvironment. Taking advantage of the liquefied environment of microcapsules, MSCs were exposed to cyclic HP of 5 or 50 MPa, 3 times/week at 37 °C. Biological tests using fluorescence staining of F-actin filaments showed a noticeable improvement in cell-cell interactions and cellular network formation of MSCs. These observations were more pronounced in osteogenic (OST) condition, as confirmed by fluorescent staining of vinculin. More interestingly, there was a significant increase in alkaline phosphatase activity of MSCs exposed to 50 MPa magnitude of HP, even in the absence of osteoinductive factors. In addition, a greater staining area of both osteopontin and hydroxyapatite was detected in the 50 MPa/OST group. These findings highlight the benefit of hydrostatic pressure to regulate osteogenesis of MSCs as well as the importance of employing simultaneous biochemical and mechanical stimulation to accelerate the osteogenic potential of MSCs for biomedical purposes.
Collapse
Affiliation(s)
| | - Carlos A. Pinto
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Diana Jesus
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Jorge A. Saraiva
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - João F. Mano
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| |
Collapse
|
69
|
Septiana WL, Ayudyasari W, Gunardi H, Pawitan JA, Balachander GM, Yu H, Antarianto RD. Liver organoids cocultured on decellularized native liver scaffolds as a bridging therapy improves survival from liver failure in rabbits. In Vitro Cell Dev Biol Anim 2023; 59:747-763. [PMID: 38110841 DOI: 10.1007/s11626-023-00817-8] [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: 07/18/2023] [Accepted: 09/28/2023] [Indexed: 12/20/2023]
Abstract
The present study aimed to develop viable liver organoids using decellularized native liver scaffolds and evaluate the efficacy of human liver organoid transplantation in a rabbit model of cirrhosis. Liver organoids were formed by coculture of hepatocyte-like cells derived from the human-induced pluripotent stem cells with three other cell types. Twelve 3-mo-old New Zealand White Rabbits underwent a sham operation, bile duct ligation, or biliary duct ligation followed by liver organoid transplantation. Liver organoid structure and function before and after transplantation were evaluated using histological and molecular analyses. A survival analysis using the Kaplan-Meier method was performed to determine the cumulative probability of survival according to liver organoid transplantation with significantly greater overall survival observed in rabbits that underwent liver organoid transplantation (P = 0.003, log-rank test). The short-term group had higher hepatic expression levels of ALB and CYP3A mRNA and lower expression levels of AST mRNA compared to the long-term group. The short-term group also had lower collagen deposition in liver tissues. Transplantation of human liver organoids cocultured in decellularized native liver scaffold into rabbits that had undergone bile duct ligation improved short-term survival and hepatic function. The results of the present study highlight the potential of liver organoid transplantation as a bridging therapy in liver failure; however, rejection and poor liver organoid function may limit the long-term efficacy of this therapeutic approach.
Collapse
Affiliation(s)
- Wahyunia Likhayati Septiana
- Program Doktor Ilmu Biomedik, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Histology, Faculty of Medicine, Universitas Gunadarma, Depok, Indonesia
| | - Wulan Ayudyasari
- Department of Surgery, Fakultas Kedokteran Universitas Indonesia, Jakarta, Indonesia
| | - Hardian Gunardi
- Department of Surgery, Fakultas Kedokteran Universitas Indonesia, Jakarta, Indonesia
| | - Jeanne Adiwinata Pawitan
- Department of Histology, Fakultas Kedokteran Universitas Indonesia, Jl Salemba Raya No 6. Jakarta Pusat 10430, Jakarta, Indonesia
- Stem Cell and Tissue Engineering Research Cluster, (IMERI) Indonesian Medical Education and Research Institute, Jakarta, Indonesia
- Integrated Service Unit of Stem Cell Medical Technology (IPT TK Sel Punca), Dr. Cipto Mangunkusumo General Hospital (RSCM), Jakarta, Indonesia
| | - Gowri Manohari Balachander
- Department of Physiology, The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, India, 221005
| | - Hanry Yu
- Department of Physiology, The Institute for Digital Medicine (WisDM), Yong Loo Lin School of Medicine, MD9-04-11, 2 Medical Drive, Singapore, 117593, Singapore
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, India, 221005
- Institute of Bioengineering & Bioimaging, A*STAR, 31 Biopolis Way, #07-01, Singapore, 138669, Singapore
- CAMP, Singapore-MIT Alliance for Research and Technology, 1 CREATE Way, Level 4 Enterprise Wing, Singapore, 138602, Singapore
- Mechanobiology Institute, National University of Singapore, T-Lab, #05-01, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Radiana Dhewayani Antarianto
- Department of Histology, Fakultas Kedokteran Universitas Indonesia, Jl Salemba Raya No 6. Jakarta Pusat 10430, Jakarta, Indonesia.
- Stem Cell and Tissue Engineering Research Cluster, (IMERI) Indonesian Medical Education and Research Institute, Jakarta, Indonesia.
| |
Collapse
|
70
|
Roberto de Barros N, Wang C, Maity S, Peirsman A, Nasiri R, Herland A, Ermis M, Kawakita S, Gregatti Carvalho B, Hosseinzadeh Kouchehbaghi N, Donizetti Herculano R, Tirpáková Z, Mohammad Hossein Dabiri S, Lucas Tanaka J, Falcone N, Choroomi A, Chen R, Huang S, Zisblatt E, Huang Y, Rashad A, Khorsandi D, Gangrade A, Voskanian L, Zhu Y, Li B, Akbari M, Lee J, Remzi Dokmeci M, Kim HJ, Khademhosseini A. Engineered organoids for biomedical applications. Adv Drug Deliv Rev 2023; 203:115142. [PMID: 37967768 PMCID: PMC10842104 DOI: 10.1016/j.addr.2023.115142] [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: 04/18/2023] [Revised: 10/03/2023] [Accepted: 11/10/2023] [Indexed: 11/17/2023]
Abstract
As miniaturized and simplified stem cell-derived 3D organ-like structures, organoids are rapidly emerging as powerful tools for biomedical applications. With their potential for personalized therapeutic interventions and high-throughput drug screening, organoids have gained significant attention recently. In this review, we discuss the latest developments in engineering organoids and using materials engineering, biochemical modifications, and advanced manufacturing technologies to improve organoid culture and replicate vital anatomical structures and functions of human tissues. We then explore the diverse biomedical applications of organoids, including drug development and disease modeling, and highlight the tools and analytical techniques used to investigate organoids and their microenvironments. We also examine the latest clinical trials and patents related to organoids that show promise for future clinical translation. Finally, we discuss the challenges and future perspectives of using organoids to advance biomedical research and potentially transform personalized medicine.
Collapse
Affiliation(s)
| | - Canran Wang
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA 91125, USA
| | - Surjendu Maity
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Arne Peirsman
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Plastic and Reconstructive Surgery, Ghent University Hospital, Ghent, Belgium
| | - Rohollah Nasiri
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, 17165 Solna, Sweden
| | - Anna Herland
- Division of Nanobiotechnology, Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, 17165 Solna, Sweden
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Bruna Gregatti Carvalho
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), 13083-970 Campinas, Brazil
| | - Negar Hosseinzadeh Kouchehbaghi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Department of Textile Engineering, Amirkabir University of Technology (Tehran Polytechnic), Hafez Avenue, 1591634311 Tehran, Iran
| | - Rondinelli Donizetti Herculano
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA; São Paulo State University (UNESP), Bioengineering and Biomaterials Group, School of Pharmaceutical Sciences, Araraquara, SP, Brazil
| | - Zuzana Tirpáková
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Department of Biology and Physiology, University of Veterinary Medicine and Pharmacy in Kosice, Komenskeho 73, 04181 Kosice, Slovakia
| | - Seyed Mohammad Hossein Dabiri
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Jean Lucas Tanaka
- Butantan Institute, Viral Biotechnology Laboratory, São Paulo, SP Brazil; University of São Paulo (USP), São Paulo, SP Brazil
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - RunRun Chen
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA
| | - Shuyi Huang
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA
| | - Elisheva Zisblatt
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Yixuan Huang
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Ahmad Rashad
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Ankit Gangrade
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Leon Voskanian
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA
| | - Bingbing Li
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; Autonomy Research Center for STEAHM (ARCS), California State University, Northridge, CA 91324, USA
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | | | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA; College of Pharmacy, Korea University, Sejong 30019, Republic of Korea.
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), Los Angeles, CA 90064, USA.
| |
Collapse
|
71
|
Alhattab DM, Isaioglou I, Alshehri S, Khan ZN, Susapto HH, Li Y, Marghani Y, Alghuneim AA, Díaz-Rúa R, Abdelrahman S, Al-Bihani S, Ahmed F, Felimban RI, Alkhatabi H, Alserihi R, Abedalthagafi M, AlFadel A, Awidi A, Chaudhary AG, Merzaban J, Hauser CAE. Fabrication of a three-dimensional bone marrow niche-like acute myeloid Leukemia disease model by an automated and controlled process using a robotic multicellular bioprinting system. Biomater Res 2023; 27:111. [PMID: 37932837 PMCID: PMC10626721 DOI: 10.1186/s40824-023-00457-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/29/2023] [Indexed: 11/08/2023] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematological malignancy that remains a therapeutic challenge due to the high incidence of disease relapse. To better understand resistance mechanisms and identify novel therapies, robust preclinical models mimicking the bone marrow (BM) microenvironment are needed. This study aimed to achieve an automated fabrication process of a three-dimensional (3D) AML disease model that recapitulates the 3D spatial structure of the BM microenvironment and applies to drug screening and investigational studies. METHODS To build this model, we investigated a unique class of tetramer peptides with an innate ability to self-assemble into stable hydrogel. An automated robotic bioprinting process was established to fabricate a 3D BM (niche-like) multicellular AML disease model comprised of leukemia cells and the BM's stromal and endothelial cellular fractions. In addition, monoculture and dual-culture models were also fabricated. Leukemia cell compatibility, functionalities (in vitro and in vivo), and drug assessment studies using our model were performed. In addition, RNAseq and gene expression analysis using TaqMan arrays were also performed on 3D cultured stromal cells and primary leukemia cells. RESULTS The selected peptide hydrogel formed a highly porous network of nanofibers with mechanical properties similar to the BM extracellular matrix. The robotic bioprinter and the novel quadruple coaxial nozzle enabled the automated fabrication of a 3D BM niche-like AML disease model with controlled deposition of multiple cell types into the model. This model supported the viability and growth of primary leukemic, endothelial, and stromal cells and recapitulated cell-cell and cell-ECM interactions. In addition, AML cells in our model possessed quiescent characteristics with improved chemoresistance attributes, resembling more the native conditions as indicated by our in vivo results. Moreover, the whole transcriptome data demonstrated the effect of 3D culture on enhancing BM niche cell characteristics. We identified molecular pathways upregulated in AML cells in our 3D model that might contribute to AML drug resistance and disease relapse. CONCLUSIONS Our results demonstrate the importance of developing 3D biomimicry models that closely recapitulate the in vivo conditions to gain deeper insights into drug resistance mechanisms and novel therapy development. These models can also improve personalized medicine by testing patient-specific treatments.
Collapse
Affiliation(s)
- Dana M Alhattab
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Ioannis Isaioglou
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Salwa Alshehri
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Zainab N Khan
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Hepi H Susapto
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Yanyan Li
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Yara Marghani
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Arwa A Alghuneim
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Rubén Díaz-Rúa
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Sherin Abdelrahman
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Shuroug Al-Bihani
- Core Laboratories, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Farid Ahmed
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Raed I Felimban
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Heba Alkhatabi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Centre, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Raed Alserihi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Malak Abedalthagafi
- Department of Pathology and Laboratory Medicine, Emory School of Medicine, Atlanta, USA
| | - AlShaibani AlFadel
- Division of Hematology, Stem Cell Transplantation & Cellular Therapy, Oncology Center, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Abdalla Awidi
- Cell Therapy Center, The University of Jordan, Amman, Jordan
- Medical School, The University of Jordan, Amman, Jordan
- Jordan University Hospital, Amman, Jordan
| | - Adeel Gulzar Chaudhary
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Innovation in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| | - Jasmeen Merzaban
- Cell Migration and Signaling Laboratory, Bioscience Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Charlotte A E Hauser
- Laboratory for Nanomedicine, Bioengineering Program, Division of Biological & Environmental Science & Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- KAUST Smart Health Initiative (KSHI), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
- Red Sea Research Center (RSRC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
72
|
Wistner SC, Rashad L, Slaughter G. Advances in tissue engineering and biofabrication for in vitro skin modeling. BIOPRINTING (AMSTERDAM, NETHERLANDS) 2023; 35:e00306. [PMID: 38645432 PMCID: PMC11031264 DOI: 10.1016/j.bprint.2023.e00306] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The global prevalence of skin disease and injury is continually increasing, yet conventional cell-based models used to study these conditions do not accurately reflect the complexity of human skin. The lack of inadequate in vitro modeling has resulted in reliance on animal-based models to test pharmaceuticals, biomedical devices, and industrial and environmental toxins to address clinical needs. These in vivo models are monetarily and morally expensive and are poor predictors of human tissue responses and clinical trial outcomes. The onset of three-dimensional (3D) culture techniques, such as cell-embedded and decellularized approaches, has offered accessible in vitro alternatives, using innovative scaffolds to improve cell-based models' structural and histological authenticity. However, these models lack adequate organizational control and complexity, resulting in variations between structures and the exclusion of physiologically relevant vascular and immunological features. Recently, biofabrication strategies, which combine biology, engineering, and manufacturing capabilities, have emerged as instrumental tools to recreate the heterogeneity of human skin precisely. Bioprinting uses computer-aided design (CAD) to yield robust and reproducible skin prototypes with unprecedented control over tissue design and assembly. As the interdisciplinary nature of biofabrication grows, we look to the promise of next-generation biofabrication technologies, such as organ-on-a-chip (OOAC) and 4D modeling, to simulate human tissue behaviors more reliably for research, pharmaceutical, and regenerative medicine purposes. This review aims to discuss the barriers to developing clinically relevant skin models, describe the evolution of skin-inspired in vitro structures, analyze the current approaches to biofabricating 3D human skin mimetics, and define the opportunities and challenges in biofabricating skin tissue for preclinical and clinical uses.
Collapse
Affiliation(s)
- Sarah C. Wistner
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Layla Rashad
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
| | - Gymama Slaughter
- Center for Bioelectronics, Old Dominion University, Norfolk, VA, 23508, USA
- Department of Electrical and Computer Engineering, Old Dominion University, Norfolk, VA, 23508, USA
| |
Collapse
|
73
|
Zengin A, Teixeira FC, Feliciano T, Habibovic P, Mota CD, Baker MB, van Rijt S. Matrix metalloproteinase degradable, in situ photocrosslinked nanocomposite bioinks for bioprinting applications. BIOMATERIALS ADVANCES 2023; 154:213647. [PMID: 37839298 DOI: 10.1016/j.bioadv.2023.213647] [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: 02/16/2023] [Revised: 08/10/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023]
Abstract
The development of suitable bioinks with high printability, mechanical strength, biodegradability, and biocompatibility is a key challenge for the clinical translation of 3D constructs produced with bioprinting technologies. In this work, we developed a new type of nanocomposite bioinks containing thiolated mesoporous silica nanoparticles (MSN) that act as active fillers within norbornene-functionalized hydrogels. The MSNs could rapidly covalently crosslink the hydrogels upon exposure to UV light. The mechanical properties of the gels could be modulated from 9.3 to 19.7 kPa with increasing concentrations of MSN. The ability of the MSN to covalently crosslink polymeric networks was, however, significantly influenced by polymer architecture and the number of functional groups. Modification of the outer surface of MSNs with matrix metalloproteinase (MMP) sensitive peptides (MSN-MMPs) resulted in proteinase K and MMP-9 enzyme responsive biodegradable bioinks. Additional cysteine modified RGD peptide incorporation enhanced cell-matrix interactions and reduced the gelation time for bioprinting. The nanocomposite bioinks could be printed by using extrusion-based bioprinting. Our nanocomposite bioinks preserved their shape during in vitro studies and encapsulated MG63 cells preserved their viability and proliferated within the bioinks. As such, our nanocomposite bioinks are promising bioinks for creating bioprinted constructs with tunable mechanical and degradation properties.
Collapse
Affiliation(s)
- Aygul Zengin
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Filipa Castro Teixeira
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Tony Feliciano
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Carlos Domingues Mota
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Matthew B Baker
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Sabine van Rijt
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands.
| |
Collapse
|
74
|
Abbas ZN, Al-Saffar AZ, Jasim SM, Sulaiman GM. Comparative analysis between 2D and 3D colorectal cancer culture models for insights into cellular morphological and transcriptomic variations. Sci Rep 2023; 13:18380. [PMID: 37884554 PMCID: PMC10603139 DOI: 10.1038/s41598-023-45144-w] [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: 05/05/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023] Open
Abstract
Drug development is a time-consuming and expensive process, given the low success rate of clinical trials. Now, anticancer drug developments have shifted to three-dimensional (3D) models which are more likely to mimic tumor behavior compared to traditional two-dimensional (2D) cultures. A comparative study among different aspects was conducted between 2D and 3D cultures using colorectal cancer (CRC) cell lines, in addition, Formalin-Fixed Paraffin-Embedded (FFPE) block samples of patients with CRC were used for evaluation. Compared to the 2D culture, cells grown in 3D displayed significant (p < 0.01) differences in the pattern of cell proliferation over time, cell death phase profile, expression of tumorgenicity-related genes, and responsiveness to 5-fluorouracil, cisplatin, and doxorubicin. Epigenetically, 3D cultures and FFPE shared the same methylation pattern and microRNA expression, while 2D cells showed elevation in methylation rate and altered microRNA expression. Lastly, transcriptomic study depending on RNA sequencing and thorough bioinformatic analyses showed significant (p-adj < 0.05) dissimilarity in gene expression profile between 2D and 3D cultures involving thousands of genes (up/down-regulated) of multiple pathways for each cell line. Taken together, the study provides insights into variations in cellular morphologies between cells cultured in 2D and 3D models.
Collapse
Affiliation(s)
- Zaid Nsaif Abbas
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Jadriya, Baghdad, Iraq
| | - Ali Z Al-Saffar
- Department of Molecular and Medical Biotechnology, College of Biotechnology, Al-Nahrain University, Jadriya, Baghdad, Iraq.
| | - Saba Mahdi Jasim
- Oncology Teaching Hospital, Medical City, Ministry of Health, Baghdad, Iraq
| | - Ghassan M Sulaiman
- Division of Biotechnology, Department of Applied Sciences, University of Technology, Baghdad, Iraq
| |
Collapse
|
75
|
Godoy-Gallardo M, Cun X, Liu X, Hosta-Rigau L. Silica Replicas Derived from Mammalian Cells as an Innovative Approach to Physically Direct Cell Lineage Decisions of Mesenchymal Stem Cells. ACS APPLIED MATERIALS & INTERFACES 2023; 15:48855-48870. [PMID: 37823476 DOI: 10.1021/acsami.3c05556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
By means of a "live-cell" template strategy, silica replicas displaying the same morphology and topography of the mammalian cells used as templates are fabricated. The replicas are used as substrates to direct the differentiation of mesenchymal stem cells (MSCs) to predefined cell lineages. Upregulation of specific genes shows how the silica replica-based substrates have the ability to induce the molecular characteristics of the mature cell types from which they have been derived from. Thus, MSCs cultured in the presence of silica replicas of human osteoblasts (HObs) differentiate into HObs-like cells, as shown by the upregulation of specific osteogenic genes. Likewise, when MSCs are incubated with silica replicas derived from human chondrocytes, an enhanced expression of chondrogenic markers is observed. Importantly, the effects of the silica replicas are cell type-specific since the incubation of MSCs with HObs silica replicas does not result in upregulation of chondrogenic markers and vice versa. What is more, for both cases, the differentiation rate is enhanced when the silica replicas are used in combination with growth factors, suggesting a potential synergistic effect. These results demonstrate the potential of this innovative method as an efficient and cheap approach with the potential to eliminate, or at least reduce, the use of biochemically soluble compounds in stem cells research.
Collapse
Affiliation(s)
- Maria Godoy-Gallardo
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Produktionstorvet, Building 423, 2800 Kongens Lyngby, Denmark
| | - Xingli Cun
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Produktionstorvet, Building 423, 2800 Kongens Lyngby, Denmark
| | - Xiaoli Liu
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Produktionstorvet, Building 423, 2800 Kongens Lyngby, Denmark
| | - Leticia Hosta-Rigau
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Produktionstorvet, Building 423, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
76
|
Princen K, Marien N, Guedens W, Graulus GJ, Adriaensens P. Hydrogels with Reversible Crosslinks for Improved Localised Stem Cell Retention: A Review. Chembiochem 2023; 24:e202300149. [PMID: 37220343 DOI: 10.1002/cbic.202300149] [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: 02/24/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 05/25/2023]
Abstract
Successful stem cell applications could have a significant impact on the medical field, where many lives are at stake. However, the translation of stem cells to the clinic could be improved by overcoming challenges in stem cell transplantation and in vivo retention at the site of tissue damage. This review aims to showcase the most recent insights into developing hydrogels that can deliver, retain, and accommodate stem cells for tissue repair. Hydrogels can be used for tissue engineering, as their flexibility and water content makes them excellent substitutes for the native extracellular matrix. Moreover, the mechanical properties of hydrogels are highly tuneable, and recognition moieties to control cell behaviour and fate can quickly be introduced. This review covers the parameters necessary for the physicochemical design of adaptable hydrogels, the variety of (bio)materials that can be used in such hydrogels, their application in stem cell delivery and some recently developed chemistries for reversible crosslinking. Implementing physical and dynamic covalent chemistry has resulted in adaptable hydrogels that can mimic the dynamic nature of the extracellular matrix.
Collapse
Affiliation(s)
- Ken Princen
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Neeve Marien
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Wanda Guedens
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Geert-Jan Graulus
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| | - Peter Adriaensens
- Biomolecule Design Group, Institute for Materials Research (IMO-IMOMEC), Hasselt University, Agoralaan-Building D, 3590, Diepenbeek, Belgium
| |
Collapse
|
77
|
Turan Sorhun D, Kuşoğlu A, Öztürk E. Developing Bovine Brain-Derived Extracellular Matrix Hydrogels: a Screen of Decellularization Methods for Their Impact on Biochemical and Mechanical Properties. ACS OMEGA 2023; 8:36933-36947. [PMID: 37841171 PMCID: PMC10569007 DOI: 10.1021/acsomega.3c04064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
Tissue models that recapitulate the key biochemical and physical aspects of the brain have been highly pursued in neural tissue engineering. Decellularization of native organs offers the advantage of preserving the composition of native extracellular matrix (ECM). Brain ECM has distinct features which play a major role in neural cell behavior. Cell instructive ligands and mechanical properties take part in the regulation of cellular processes in homeostasis and diseases. One of the main challenges in decellularization is maintaining mechanical integrity in reconstituted hydrogels and achieving physiologically relevant stiffness. The effect of the decellularization process on different mechanical aspects, particularly the viscoelasticity of brain-derived hydrogels, has not been addressed. In this study, we developed bovine brain-derived hydrogels for the first time. We pursued seven protocols for decellularization and screened their effect on biochemical content, hydrogel formation, and mechanical characteristics. We show that bovine brain offers an easily accessible alternative for in vitro brain tissue modeling. Our data demonstrate that the choice of decellularization method strongly alters gelation as well as the stiffness and viscoelasticity of the resulting hydrogels. Lastly, we investigated the cytocompatibility of brain ECM hydrogels and the effect of modulated mechanical properties on the growth and morphological features of neuroblastoma cells.
Collapse
Affiliation(s)
- Duygu Turan Sorhun
- Engineered
Cancer and Organ Models Laboratory, Koç
University, Istanbul 34450, Turkey
- Research
Center for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
| | - Alican Kuşoğlu
- Engineered
Cancer and Organ Models Laboratory, Koç
University, Istanbul 34450, Turkey
- Research
Center for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
| | - Ece Öztürk
- Engineered
Cancer and Organ Models Laboratory, Koç
University, Istanbul 34450, Turkey
- Research
Center for Translational Medicine (KUTTAM), Koç University, Istanbul 34450, Turkey
- Department
of Medical Biology, School of Medicine, Koç University, Istanbul 34450, Turkey
| |
Collapse
|
78
|
Madappura AP, Madduri S. A comprehensive review of silk-fibroin hydrogels for cell and drug delivery applications in tissue engineering and regenerative medicine. Comput Struct Biotechnol J 2023; 21:4868-4886. [PMID: 37860231 PMCID: PMC10583100 DOI: 10.1016/j.csbj.2023.10.012] [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: 05/15/2023] [Revised: 10/07/2023] [Accepted: 10/08/2023] [Indexed: 10/21/2023] Open
Abstract
Hydrogel scaffolds hold great promise for developing novel treatment strategies in the field of regenerative medicine. Within this context, silk fibroin (SF) has proven to be a versatile material for a wide range of tissue engineering applications owing to its structural and functional properties. In the present review, we report on the design and fabrication of different forms of SF-based scaffolds for tissue regeneration applications, particularly for skin, bone, and neural tissues. In particular, SF hydrogels have emerged as delivery systems for a wide range of bio-actives. Given the growing interest in the field, this review has a primary focus on the fabrication, characterization, and properties of SF hydrogels. We also discuss their potential for the delivery of drugs, stem cells, genes, peptides, and growth factors, including future directions in the field of SF hydrogel scaffolds.
Collapse
Affiliation(s)
- Alakananda Parassini Madappura
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044 Hsinchu, Taiwan, Republic of China
| | - Srinivas Madduri
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
- Department of Surgery, University of Geneva, Geneva, Switzerland
| |
Collapse
|
79
|
Choi J, Lee EJ, Jang WB, Kwon SM. Development of Biocompatible 3D-Printed Artificial Blood Vessels through Multidimensional Approaches. J Funct Biomater 2023; 14:497. [PMID: 37888162 PMCID: PMC10607080 DOI: 10.3390/jfb14100497] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/05/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
Within the human body, the intricate network of blood vessels plays a pivotal role in transporting nutrients and oxygen and maintaining homeostasis. Bioprinting is an innovative technology with the potential to revolutionize this field by constructing complex multicellular structures. This technique offers the advantage of depositing individual cells, growth factors, and biochemical signals, thereby facilitating the growth of functional blood vessels. Despite the challenges in fabricating vascularized constructs, bioprinting has emerged as an advance in organ engineering. The continuous evolution of bioprinting technology and biomaterial knowledge provides an avenue to overcome the hurdles associated with vascularized tissue fabrication. This article provides an overview of the biofabrication process used to create vascular and vascularized constructs. It delves into the various techniques used in vascular engineering, including extrusion-, droplet-, and laser-based bioprinting methods. Integrating these techniques offers the prospect of crafting artificial blood vessels with remarkable precision and functionality. Therefore, the potential impact of bioprinting in vascular engineering is significant. With technological advances, it holds promise in revolutionizing organ transplantation, tissue engineering, and regenerative medicine. By mimicking the natural complexity of blood vessels, bioprinting brings us one step closer to engineering organs with functional vasculature, ushering in a new era of medical advancement.
Collapse
Affiliation(s)
- Jaewoo Choi
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Eun Ji Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Woong Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (J.C.); (E.J.L.)
- Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
80
|
Panda AK, Basu B. Regenerative bioelectronics: A strategic roadmap for precision medicine. Biomaterials 2023; 301:122271. [PMID: 37619262 DOI: 10.1016/j.biomaterials.2023.122271] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/30/2023] [Accepted: 08/06/2023] [Indexed: 08/26/2023]
Abstract
In the past few decades, stem cell-based regenerative engineering has demonstrated its significant potential to repair damaged tissues and to restore their functionalities. Despite such advancement in regenerative engineering, the clinical translation remains a major challenge. In the stance of personalized treatment, the recent progress in bioelectronic medicine likewise evolved as another important research domain of larger significance for human healthcare. Over the last several years, our research group has adopted biomaterials-based regenerative engineering strategies using innovative bioelectronic stimulation protocols based on either electric or magnetic stimuli to direct cellular differentiation on engineered biomaterials with a range of elastic stiffness or functional properties (electroactivity/magnetoactivity). In this article, the role of bioelectronics in stem cell-based regenerative engineering has been critically analyzed to stimulate futuristic research in the treatment of degenerative diseases as well as to address some fundamental questions in stem cell biology. Built on the concepts from two independent biomedical research domains (regenerative engineering and bioelectronic medicine), we propose a converging research theme, 'Regenerative Bioelectronics'. Further, a series of recommendations have been put forward to address the current challenges in bridging the gap in stem cell therapy and bioelectronic medicine. Enacting the strategic blueprint of bioelectronic-based regenerative engineering can potentially deliver the unmet clinical needs for treating incurable degenerative diseases.
Collapse
Affiliation(s)
- Asish Kumar Panda
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bengaluru, 560012, India
| | - Bikramjit Basu
- Laboratory for Biomaterials, Materials Research Centre, Indian Institute of Science, Bengaluru, 560012, India; Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru, 560012, India.
| |
Collapse
|
81
|
Shang Y, Zeng J, Matsusaki M. Construction of enzyme digested holes on hydrogel surface inspired by cell migration processes. Biochem Biophys Res Commun 2023; 674:69-74. [PMID: 37413707 DOI: 10.1016/j.bbrc.2023.06.077] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 06/24/2023] [Indexed: 07/08/2023]
Abstract
The construction of in vitro capillary network models for drug testing and toxicity evaluation has become a major challenge in the field of tissue engineering. Previously, we discovered a novel phenomenon of hole formation by endothelial cell migration on the surface of fibrin gels. Interestingly, the hole characteristics, such as depth and number, were strongly influenced by the gel stiffness, but the details of hole formation are not to be clarified. In this study, we tried to understand the effect of hydrogel stiffness on the hole formation by dropping collagenase solution onto the surface of the hydrogels because the endothelial cell migration was made possible by the metalloproteinases' digestion. We found that smaller hole structures were formed on stiffer fibrin gels, but larger ones were formed on softer fibrin gels after the hydrogel digestion of the collagenase. This is consistent with our previous results in experiments on hole structures formed by endothelial cells. Furthermore, deep and small hole structures were successfully obtained by optimizing the volume of collagenase solution and incubation time. This unique approach inspired by endothelial cell hole formation may provide new methods of fabricating hydrogels with opening hole structures.
Collapse
Affiliation(s)
- Yucheng Shang
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Jinfeng Zeng
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
82
|
Bernava G, Iop L. Advances in the design, generation, and application of tissue-engineered myocardial equivalents. Front Bioeng Biotechnol 2023; 11:1247572. [PMID: 37811368 PMCID: PMC10559975 DOI: 10.3389/fbioe.2023.1247572] [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: 06/26/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Due to the limited regenerative ability of cardiomyocytes, the disabling irreversible condition of myocardial failure can only be treated with conservative and temporary therapeutic approaches, not able to repair the damage directly, or with organ transplantation. Among the regenerative strategies, intramyocardial cell injection or intravascular cell infusion should attenuate damage to the myocardium and reduce the risk of heart failure. However, these cell delivery-based therapies suffer from significant drawbacks and have a low success rate. Indeed, cardiac tissue engineering efforts are directed to repair, replace, and regenerate native myocardial tissue function. In a regenerative strategy, biomaterials and biomimetic stimuli play a key role in promoting cell adhesion, proliferation, differentiation, and neo-tissue formation. Thus, appropriate biochemical and biophysical cues should be combined with scaffolds emulating extracellular matrix in order to support cell growth and prompt favorable cardiac microenvironment and tissue regeneration. In this review, we provide an overview of recent developments that occurred in the biomimetic design and fabrication of cardiac scaffolds and patches. Furthermore, we sift in vitro and in situ strategies in several preclinical and clinical applications. Finally, we evaluate the possible use of bioengineered cardiac tissue equivalents as in vitro models for disease studies and drug tests.
Collapse
Affiliation(s)
| | - Laura Iop
- Department of Cardiac Thoracic Vascular Sciences and Public Health, Padua Medical School, University of Padua, Padua, Italy
| |
Collapse
|
83
|
Ivanov AA, Kuznetsova AV, Popova OP, Danilova TI, Latyshev AV, Yanushevich OO. Influence of Extracellular Matrix Components on the Differentiation of Periodontal Ligament Stem Cells in Collagen I Hydrogel. Cells 2023; 12:2335. [PMID: 37830549 PMCID: PMC10571948 DOI: 10.3390/cells12192335] [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: 07/17/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023] Open
Abstract
Regeneration of periodontal tissues requires an integrated approach to the restoration of the periodontal ligament, cementum, and alveolar bone surrounding the teeth. Current strategies in endogenous regenerative dentistry widely use biomaterials, in particular the decellularized extracellular matrix (dECM), to facilitate the recruitment of populations of resident cells into damaged tissues and stimulate their proliferation and differentiation. The purpose of our study was to evaluate the effect of the exogenous components of the extracellular matrix (hyaluronic acid, laminin, fibronectin) on the differentiation of periodontal ligament stem cells (PDLSCs) cultured with dECM (combinations of decellularized tooth matrices and periodontal ligament) in a 3D collagen I hydrogel. The immunohistochemical expression of various markers in PDLSCs was assessed quantitatively and semi-quantitatively on paraffin sections. The results showed that PDLSCs cultured under these conditions for 14 days exhibited phenotypic characteristics consistent with osteoblast-like and odontoblast-like cells. This potential has been demonstrated by the expression of osteogenic differentiation markers (OC, OPN, ALP) and odontogenic markers (DSPP). This phenomenon corresponds to the in vivo state of the periodontal ligament, in which cells at the interface between bone and cementum tend to differentiate into osteoblasts or cementoblasts. The addition of fibronectin to the dECM most effectively induces the differentiation of PDLSCs into osteoblast-like and odontoblast-like cells under 3D culture conditions. Therefore, this bioengineered construct has a high potential for future use in periodontal tissue regeneration.
Collapse
Affiliation(s)
- Alexey A. Ivanov
- Laboratory of Molecular and Cellular Pathology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (A.V.K.); (O.P.P.); (T.I.D.); (A.V.L.)
| | - Alla V. Kuznetsova
- Laboratory of Molecular and Cellular Pathology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (A.V.K.); (O.P.P.); (T.I.D.); (A.V.L.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Str., 119334 Moscow, Russia
| | - Olga P. Popova
- Laboratory of Molecular and Cellular Pathology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (A.V.K.); (O.P.P.); (T.I.D.); (A.V.L.)
| | - Tamara I. Danilova
- Laboratory of Molecular and Cellular Pathology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (A.V.K.); (O.P.P.); (T.I.D.); (A.V.L.)
| | - Andrey V. Latyshev
- Laboratory of Molecular and Cellular Pathology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia; (A.V.K.); (O.P.P.); (T.I.D.); (A.V.L.)
| | - Oleg O. Yanushevich
- Department of Periodontology, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 20 Delegatskaya Str., 127473 Moscow, Russia;
| |
Collapse
|
84
|
Xiong J, Miller CM, Sharma D. Effect of Bergenin on Human Gingival Fibroblast Response on Zirconia Implant Surfaces: An In Vitro Study. J Funct Biomater 2023; 14:474. [PMID: 37754887 PMCID: PMC10532536 DOI: 10.3390/jfb14090474] [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: 08/11/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
The poor quality of life associated with the loss of teeth can be improved by the placing of dental implants. However, successful implantation relies on integration with soft tissues or peri-implant inflammatory disease that can lead to the loss of the implant. Pharmacological agents, such as antibiotics and antiseptics, can be used as adjunct therapies to facilitate osseointegration; however, they can have a detrimental effect on cells, and resistance is an issue. Alternative treatments are needed. Hence, this study aimed to examine the safety profile of bergenin (at 2.5 μM and 5 μM), a traditional medicine, towards human gingival fibroblasts cultured on acid-etched zirconia implant surfaces. Cellular responses were analysed using SEM, resazurin assay, and scratch wound healing assay. Qualitative assessment was conducted for morphology (day 1) and attachment (early and delayed), and quantitative evaluation for proliferation (day 1, 3, 5 and 7), and migration (0 h, 6 h and 24 h). The concentrations of bergenin at 2.5 μM and 5 μM did not demonstrate a statistically significant effect with regard to any of the cellular responses (p > 0.05) tested. In conclusion, bergenin is non-cytotoxic and is potentially safe to be used as a local pharmacological agent for the management of peri-implant inflammatory diseases.
Collapse
Affiliation(s)
- John Xiong
- College of Medicine and Dentistry, James Cook University, Smithfield, QLD 4878, Australia; (J.X.); (C.M.M.)
| | - Catherine M. Miller
- College of Medicine and Dentistry, James Cook University, Smithfield, QLD 4878, Australia; (J.X.); (C.M.M.)
| | - Dileep Sharma
- College of Medicine and Dentistry, James Cook University, Smithfield, QLD 4878, Australia; (J.X.); (C.M.M.)
- School of Health Sciences, College of Health, Medicine and Wellbeing, The University of Newcastle, Ourimbah, NSW 2258, Australia
| |
Collapse
|
85
|
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
|
86
|
M Galindo J, San-Millán I, Castillo-Sarmiento CA, Ballesteros-Yáñez I, Herrero MA, Merino S, Vázquez E. Mimicking the extracellular matrix by incorporating functionalized graphene into hybrid hydrogels. NANOSCALE 2023; 15:14238-14248. [PMID: 37599610 DOI: 10.1039/d3nr02689b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
The efficient functionalization of graphene with sulfonic groups using a sustainable approach facilitates the interaction of biomolecules with its surface. The inclusion of these graphene sheets inside a photopolymerized acrylamide-based hydrogel provides a 3D scaffold with viscoelastic behaviour closer to that found in natural tissues. Cell-culture experiments and differentiation assays with SH-SY5Y cells showed that these hybrid hydrogels are non-cytotoxic, thus making them potentially useful as scaffold materials mimicking the extracellular environment.
Collapse
Affiliation(s)
- Josué M Galindo
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain.
| | - Irene San-Millán
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain.
| | | | | | - M Antonia Herrero
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain.
| | - Sonia Merino
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain.
| | - Ester Vázquez
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071 Ciudad Real, Spain.
| |
Collapse
|
87
|
Nagendla NK, Muralidharan K, Raju M, Mohan H, Selvakumar P, Bhandi MM, Mudiam MKR, Ramalingam V. Comprehensive metabolomic analysis of Mangifera indica leaves using UPLC-ESI-Q-TOF-MS E for cell differentiation: An in vitro and in vivo study. Food Res Int 2023; 171:112993. [PMID: 37330843 DOI: 10.1016/j.foodres.2023.112993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 05/14/2023] [Accepted: 05/16/2023] [Indexed: 06/19/2023]
Abstract
The comprehensive metabolic profiling was performed in the leaf extracts of Mangifera indica and assessed for their significant therapeutic application in tissue engineering and regenerative medicine in both in vitro and in vivo studies. About 147 compounds were identified in the ethyl acetate and methanol extracts of M. indica using MS/MS fragmentation analysis and the selected compounds were quantified using LC-QqQ-MS analysis. The in vitro cytotoxic activity showed that the M. indica extracts enhance the proliferation of mouse myoblast cells in concentration-dependent manner. As well, the extracts of M. indica induce the myotube formation by generating oxidative stress in the C2C12 cells was confirmed. The western blot analysis clearly showed that the M. indica induce myogenic differentiation by upregulating the myogenic marker proteins such as PI3K, Akt, mTOR, MyoG, and MyoD. The in vivo studies showed that the extracts expedites the acute wound repair by formation of crust, wound closure and improves the blood perfusion towards the wound area. Together, the leaves of M. indica can be used as excellent therapeutic agent for tissue repair and wound healing applications.
Collapse
Affiliation(s)
- Narendra Kumar Nagendla
- Department of Analytical & Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Kathirvel Muralidharan
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India; Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Malothu Raju
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India; Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Harshavardhan Mohan
- Department of Chemistry, Research Institute of Physics and Chemistry, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Piramanayagam Selvakumar
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Murali Mohan Bhandi
- Department of Analytical & Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Mohana Krishna Reddy Mudiam
- Department of Analytical & Structural Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India.
| | - Vaikundamoorthy Ramalingam
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201 002, India; Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.
| |
Collapse
|
88
|
Savva A, Saez J, Withers A, Barberio C, Stoeger V, Elias-Kirma S, Lu Z, Moysidou CM, Kallitsis K, Pitsalidis C, Owens RM. 3D organic bioelectronics for electrical monitoring of human adult stem cells. MATERIALS HORIZONS 2023; 10:3589-3600. [PMID: 37318042 PMCID: PMC10464098 DOI: 10.1039/d3mh00785e] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/05/2023] [Indexed: 06/16/2023]
Abstract
Three-dimensional in vitro stem cell models have enabled a fundamental understanding of cues that direct stem cell fate. While sophisticated 3D tissues can be generated, technology that can accurately monitor these complex models in a high-throughput and non-invasive manner is not well adapted. Here we show the development of 3D bioelectronic devices based on the electroactive polymer poly(3,4-ethylenedioxythiophene)-poly(styrenesulfonate)-(PEDOT:PSS) and their use for non-invasive, electrical monitoring of stem cell growth. We show that the electrical, mechanical and wetting properties as well as the pore size/architecture of 3D PEDOT:PSS scaffolds can be fine-tuned simply by changing the processing crosslinker additive. We present a comprehensive characterization of both 2D PEDOT:PSS thin films of controlled thicknesses, and 3D porous PEDOT:PSS structures made by the freeze-drying technique. By slicing the bulky scaffolds we generate homogeneous, porous 250 μm thick PEDOT:PSS slices, constituting biocompatible 3D constructs able to support stem cell cultures. These multifunctional slices are attached on indium-tin oxide substrates (ITO) with the help of an electrically active adhesion layer, enabling 3D bioelectronic devices with a characteristic and reproducible, frequency dependent impedance response. This response changes drastically when human adipose derived stem cells (hADSCs) grow within the porous PEDOT:PSS network as revealed by fluorescence microscopy. The increase of cell population within the PEDOT:PSS porous network impedes the charge flow at the interface between PEDOT:PSS and ITO, enabling the interface resistance (R1) to be used as a figure of merit to monitor the proliferation of stem cells. The non-invasive monitoring of stem cell growth allows for the subsequent differentiation 3D stem cell cultures into neuron like cells, as verified by immunofluorescence and RT-qPCR measurements. The strategy of controlling important properties of 3D PEDOT:PSS structures simply by altering processing parameters can be applied for development of a number of stem cell in vitro models as well as stem cell differentiation pathways. We believe the results presented here will advance 3D bioelectronic technology for both fundamental understanding of in vitro stem cell cultures as well as the development of personalized therapies.
Collapse
Affiliation(s)
- Achilleas Savva
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| | - Janire Saez
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
- Microfluidics Cluster UPV/EHU, BIOMICs Microfluidics Group, Lascaray Research Center, University of the Basque Country UPV/EHU, Avenida Miguel de Unamuno, 3, 01006, Vitoria-Gasteiz, Spain
- Basque Foundation for Science, IKERBASQUE, E-48011 Bilbao, Spain
- Bioaraba Health Research Institute, Microfluidics Cluster UPV/EHU, Vitoria-Gasteiz, Spain
| | - Aimee Withers
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| | - Chiara Barberio
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| | - Verena Stoeger
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| | - Shani Elias-Kirma
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| | - Zixuan Lu
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| | - Chrysanthi-Maria Moysidou
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| | - Konstantinos Kallitsis
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| | - Charalampos Pitsalidis
- Department of Physics, Khalifa University of Science and Technology, P. O. Box 127788, Abu Dhabi, United Arab Emirates
- Healthcare Engineering Innovation Center (HEIC), Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| | - Róisín M Owens
- Department of Chemical Engineering and Biotechnology, University of Cambridge, CB3 0AS Cambridge, UK.
| |
Collapse
|
89
|
Jaime-Rodríguez M, Cadena-Hernández AL, Rosales-Valencia LD, Padilla-Sánchez JM, Chavez-Santoscoy RA. Are genetic drift and stem cell adherence in laboratory culture issues for cultivated meat production? Front Nutr 2023; 10:1189664. [PMID: 37701376 PMCID: PMC10493286 DOI: 10.3389/fnut.2023.1189664] [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: 03/19/2023] [Accepted: 08/11/2023] [Indexed: 09/14/2023] Open
Abstract
Mesenchymal stem cell-based cultivated meat is a promising solution to the ecological and ethical problems posed by traditional meat production, since it exhibits a protein content and composition that is more comparable to original meat proteins than any other source of cultivated meat products, including plants, bacteria, and fungi. Nonetheless, the nature and laboratory behavior of mesenchymal stem cells pose two significant challenges for large-scale production: genetic drift and adherent growth in culture. Culture conditions used in the laboratory expose the cells to a selective pressure that causes genetic drift, which may give rise to oncogene activation and the loss of "stemness." This is why genetic and functional analysis of the cells during culture is required to determine the maximum number of passages within the laboratory where no significant mutations or loss of function are detected. Moreover, the adherent growth of mesenchymal stem cells can be an obstacle for their large-scale production since volume to surface ratio is limited for high volume containers. Multi-tray systems, roller bottles, and microcarriers have been proposed as potential solutions to scale-up the production of adherent cells required for cultivated meat. The most promising solutions for the safety problems and large-scale obstacles for cultivated meat production are the determination of a limit number of passages based on a genetic analysis and the use of microcarriers from edible materials to maximize the volume to surface proportion and decrease the downstream operations needed for cultivated meat production.
Collapse
|
90
|
Malektaj H, Nour S, Imani R, Siadati MH. Angiogenesis induction as a key step in cardiac tissue Regeneration: From angiogenic agents to biomaterials. Int J Pharm 2023; 643:123233. [PMID: 37460050 DOI: 10.1016/j.ijpharm.2023.123233] [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: 01/25/2023] [Revised: 07/02/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. After myocardial infarction, the vascular supply of the heart is damaged or blocked, leading to the formation of scar tissue, followed by several cardiac dysfunctions or even death. In this regard, induction of angiogenesis is considered as a vital process for supplying nutrients and oxygen to the cells in cardiac tissue engineering. The current review aims to summarize different approaches of angiogenesis induction for effective cardiac tissue repair. Accordingly, a comprehensive classification of induction of pro-angiogenic signaling pathways through using engineered biomaterials, drugs, angiogenic factors, as well as combinatorial approaches is introduced as a potential platform for cardiac regeneration application. The angiogenic induction for cardiac repair can enhance patient treatment outcomes and generate economic prospects for the biomedical industry. The development and commercialization of angiogenesis methods often involves collaboration between academic institutions, research organizations, and biomedical companies.
Collapse
Affiliation(s)
- Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, Aalborg 9220, Denmark
| | - Shirin Nour
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, VIC 3010, Australia; Department of Chemical Engineering, The University of Melbourne, VIC 3010, Australia
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran.
| | - Mohammad H Siadati
- Materials Science and Engineering Faculty, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
91
|
Zhang X, Karagöz Z, Swapnasrita S, Habibovic P, Carlier A, van Rijt S. Development of Mesoporous Silica Nanoparticle-Based Films with Tunable Arginine-Glycine-Aspartate Peptide Global Density and Clustering Levels to Study Stem Cell Adhesion and Differentiation. ACS APPLIED MATERIALS & INTERFACES 2023; 15:38171-38184. [PMID: 37527490 PMCID: PMC10436245 DOI: 10.1021/acsami.3c04249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/20/2023] [Indexed: 08/03/2023]
Abstract
Stem cell adhesion is mediated via the binding of integrin receptors to adhesion motifs present in the extracellular matrix (ECM). The spatial organization of adhesion ligands plays an important role in stem cell integrin-mediated adhesion. In this study, we developed a series of biointerfaces using arginine-glycine-aspartate (RGD)-functionalized mesoporous silica nanoparticles (MSN-RGD) to study the effect of RGD adhesion ligand global density (ligand coverage over the surface), spacing, and RGD clustering levels on stem cell adhesion and differentiation. To prepare the biointerface, MSNs were chemically functionalized with RGD peptides via an antifouling poly(ethylene glycol) (PEG) linker. The RGD surface functionalization ratio could be controlled to create MSNs with high and low RGD ligand clustering levels. MSN films with varying RGD global densities could be created by blending different ratios of MSN-RGD and non-RGD-functionalized MSNs together. A computational simulation study was performed to analyze nanoparticle distribution and RGD spacing on the resulting surfaces to determine experimental conditions. Enhanced cell adhesion and spreading were observed when RGD global density increased from 1.06 to 5.32 nmol cm-2 using highly clustered RGD-MSN-based films. Higher RGD ligand clustering levels led to larger cell spreading and increased formation of focal adhesions. Moreover, a higher RGD ligand clustering level promoted the expression of alkaline phosphatase in hMSCs. Overall, these findings indicate that both RGD global density and clustering levels are crucial variables in regulating stem cell behaviors. This study provides important information about ligand-integrin interactions, which could be implemented into biomaterial design to achieve optimal performance of adhesive functional peptides.
Collapse
Affiliation(s)
- Xingzhen Zhang
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Zeynep Karagöz
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Sangita Swapnasrita
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Pamela Habibovic
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Aurélie Carlier
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Sabine van Rijt
- Department of Instructive
Biomaterials Engineering MERLN Institute for Technology-Inspired Regenerative
Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
92
|
Chang CC, Jiang SS, Tsai FY, Hsu PJ, Hsieh CC, Wang LT, Yen ML, Yen BL. Targeting Conserved Pathways in 3D Spheroid Formation of Diverse Cell Types for Translational Application: Enhanced Functional and Antioxidant Capacity. Cells 2023; 12:2050. [PMID: 37626861 PMCID: PMC10453086 DOI: 10.3390/cells12162050] [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: 07/08/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Three-dimensional (3D) in vitro spheroid/organoid culture increasingly appears to better mimic physiological states than standard 2D systems. The biological consequence of 3D spheroids, however, differs for different cell types: for pluripotent embryonic stem cells (ESCs), differentiation and loss of stemness occur, while the converse is true for somatic and cancer cells. Despite such diverse consequences, there are likely conserved mechanisms governing 3D spheroid formation across cell types that are unknown but could be efficiently targeted for translational application. To elucidate such processes, we performed transcriptome analysis with functional validation on 2D- and 3D-cultured mouse ESCs, mesenchymal stromal/stem cells (MSCs), and cancer cells. At both the transcriptomic and functional levels, 3D spheroid formation resulted in commitment towards known cell-specific functional outcomes. Surprisingly in all cell types, downregulation of the cholesterol synthesis pathway was found during 3D spheroid formation, with modulation concomitantly affecting 3D spheroid formation and cell-specific consequences; similar results were seen with human cell types. Furthermore, improved antioxidant capacity after 3D spheroid formation across cell types was further enhanced with modulation of the pathway. These findings demonstrate the profound cell-specific consequences and the translational value of understanding conserved mechanisms across diverse cell types after 3D spheroid formation.
Collapse
Affiliation(s)
- Chia-Chi Chang
- Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), Taipei 114, Taiwan
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 350, Taiwan
| | | | - Fang-Yu Tsai
- National Institute of Cancer Research, NHRI, Zhunan 350, Taiwan
| | - Pei-Ju Hsu
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 350, Taiwan
| | - Chen-Chan Hsieh
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 350, Taiwan
| | - Li-Tzu Wang
- Department of Obstetrics/Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, Taipei 100, Taiwan
| | - Men-Luh Yen
- Department of Obstetrics/Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, Taipei 100, Taiwan
| | - B. Linju Yen
- Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), Taipei 114, Taiwan
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 350, Taiwan
| |
Collapse
|
93
|
Luo L, Liu L, Ding Y, Dong Y, Ma M. Advances in biomimetic hydrogels for organoid culture. Chem Commun (Camb) 2023; 59:9675-9686. [PMID: 37455615 DOI: 10.1039/d3cc01274c] [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: 07/18/2023]
Abstract
An organoid is a 3-dimensional (3D) cell culture system that mimics the structural and functional characteristics of organs, and it has promising applications in regenerative medicine, precision drug screening and personalised therapy. However, current culture techniques of organoids usually use mouse tumour-derived scaffolds (Matrigel) or other animal-derived decellularised extracellular matrices as culture systems with poorly defined components and undefined chemical and physical properties, which limit the growth of organoids and the reproducibility of culture conditions. In contrast, some synthetic culture materials have emerged in recent years with well-defined compositions, and flexible adjustment and optimisation of physical and chemical properties, which can effectively support organoid growth and development and prolong survival time of organoid in vitro. In this review, we will introduce the challenge of animal-derived decellularised extracellular matrices in organoid culture, and summarise the categories of biomimetic hydrogels currently used for organoid culture, and then discuss the future opportunities and perspectives in the development of advanced hydrogels in organoids. We hope that this review can promote academic communication in the field of organoid research and provide some assistance in advancing the development of organoid cultivation technology.
Collapse
Affiliation(s)
- Lili Luo
- Department of Nutrition and Health, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian, Beijing, 100193, P. R. China.
| | - Libing Liu
- Department of Nutrition and Health, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian, Beijing, 100193, P. R. China.
| | - Yuxuan Ding
- Department of Nutrition and Health, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian, Beijing, 100193, P. R. China.
| | - Yixuan Dong
- Department of Nutrition and Health, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian, Beijing, 100193, P. R. China.
| | - Min Ma
- Department of Nutrition and Health, China Agricultural University, No. 2 Yuanmingyuan West Road, Haidian, Beijing, 100193, P. R. China.
| |
Collapse
|
94
|
Ferraz MDAMM, Ferronato GDA. Opportunities involving microfluidics and 3D culture systems to the in vitro embryo production. Anim Reprod 2023; 20:e20230058. [PMID: 37638255 PMCID: PMC10449241 DOI: 10.1590/1984-3143-ar2023-0058] [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: 04/26/2022] [Accepted: 06/29/2023] [Indexed: 08/29/2023] Open
Abstract
Traditional methods of gamete handling, fertilization, and embryo culture often face limitations in efficiency, consistency, and the ability to closely mimic in vivo conditions. This review explores the opportunities presented by microfluidic and 3D culture systems in overcoming these challenges and enhancing in vitro embryo production. We discuss the basic principles of microfluidics, emphasizing their inherent advantages such as precise control of fluid flow, reduced reagent consumption, and high-throughput capabilities. Furthermore, we delve into microfluidic devices designed for gamete manipulation, in vitro fertilization, and embryo culture, highlighting innovations such as droplet-based microfluidics and on-chip monitoring. Next, we explore the integration of 3D culture systems, including the use of biomimetic scaffolds and organ-on-a-chip platforms, with a particular focus on the oviduct-on-a-chip. Finally, we discuss the potential of these advanced systems to improve embryo production outcomes and advance our understanding of early embryo development. By leveraging the unique capabilities of microfluidics and 3D culture systems, we foresee significant advancements in the efficiency, effectiveness, and clinical success of in vitro embryo production.
Collapse
Affiliation(s)
- Marcia de Almeida Monteiro Melo Ferraz
- Faculty of Veterinary Medicine, Ludwig-Maximilians University of Munich, Oberschleißheim, Germany
- Gene Center, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Giuliana de Avila Ferronato
- Faculty of Veterinary Medicine, Ludwig-Maximilians University of Munich, Oberschleißheim, Germany
- Gene Center, Ludwig-Maximilians University of Munich, Munich, Germany
| |
Collapse
|
95
|
Kutluk H, Bastounis EE, Constantinou I. Integration of Extracellular Matrices into Organ-on-Chip Systems. Adv Healthc Mater 2023; 12:e2203256. [PMID: 37018430 PMCID: PMC11468608 DOI: 10.1002/adhm.202203256] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/20/2023] [Indexed: 04/07/2023]
Abstract
The extracellular matrix (ECM) is a complex, dynamic network present within all tissues and organs that not only acts as a mechanical support and anchorage point but can also direct fundamental cell behavior, function, and characteristics. Although the importance of the ECM is well established, the integration of well-controlled ECMs into Organ-on-Chip (OoC) platforms remains challenging and the methods to modulate and assess ECM properties on OoCs remain underdeveloped. In this review, current state-of-the-art design and assessment of in vitro ECM environments is discussed with a focus on their integration into OoCs. Among other things, synthetic and natural hydrogels, as well as polydimethylsiloxane (PDMS) used as substrates, coatings, or cell culture membranes are reviewed in terms of their ability to mimic the native ECM and their accessibility for characterization. The intricate interplay among materials, OoC architecture, and ECM characterization is critically discussed as it significantly complicates the design of ECM-related studies, comparability between works, and reproducibility that can be achieved across research laboratories. Improving the biomimetic nature of OoCs by integrating properly considered ECMs would contribute to their further adoption as replacements for animal models, and precisely tailored ECM properties would promote the use of OoCs in mechanobiology.
Collapse
Affiliation(s)
- Hazal Kutluk
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| | - Effie E. Bastounis
- Institute of Microbiology and Infection Medicine (IMIT)Eberhard Karls University of TübingenAuf der Morgenstelle 28, E872076TübingenGermany
- Cluster of Excellence “Controlling Microbes to Fight Infections” EXC 2124Eberhard Karls University of TübingenAuf der Morgenstelle 2872076TübingenGermany
| | - Iordania Constantinou
- Institute of Microtechnology (IMT)Technical University of BraunschweigAlte Salzdahlumer Str. 20338124BraunschweigGermany
- Center of Pharmaceutical Engineering (PVZ)Technical University of BraunschweigFranz‐Liszt‐Str. 35a38106BraunschweigGermany
| |
Collapse
|
96
|
Fusi G, Constantinides M, Fissoun C, Pichard L, Pers YM, Ferreira-Lopez R, Pantesco V, Poulet C, Malaise O, De Seny D, Lemaitre JM, Jorgensen C, Brondello JM. Senescence-Driven Inflammatory and Trophic Microenvironment Imprints Mesenchymal Stromal/Stem Cells in Osteoarthritic Patients. Biomedicines 2023; 11:1994. [PMID: 37509633 PMCID: PMC10377055 DOI: 10.3390/biomedicines11071994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Senescent cells promote progressive tissue degeneration through the establishment of a combined inflammatory and trophic microenvironment. The cellular senescence state has therefore emerged as a central driving mechanism of numerous age-related diseases, including osteoarthritis (OA), the most common rheumatic disease. Senescence hallmarks are detectable in chondrocytes, synoviocytes and sub-chondral bone cells. This study investigates how the senescence-driven microenvironment could impact the cell fate of resident osteoarticular mesenchymal stromal/stem cells (MSCs) that are hence contributing to OA disease progression. For that purpose, we performed a comparative gene expression analysis of MSCs isolated from healthy donors that were in vitro chronically exposed either to interferon-gamma (IFN-γ) or Transforming Growth Factor beta 1 (TGFβ1), two archetypical factors produced by senescent cells. Both treatments reduced MSC self-renewal capacities by upregulating different senescence-driven cycle-dependent kinase inhibitors. Furthermore, a common set of differentially expressed genes was identified in both treated MSCs that was also found enriched in MSCs isolated from OA patients. These findings highlight an imprinting of OA MSCs by the senescent joint microenvironment that changes their matrisome gene expression. Altogether, this research gives new insights into OA etiology and points to new innovative therapeutic opportunities to treat OA patients.
Collapse
Affiliation(s)
- Giuseppe Fusi
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
| | | | | | - Lydiane Pichard
- SAFE-iPSC Facility INGESTEM, Montpellier University Hospital, 34298 Montpellier, France
| | - Yves-Marie Pers
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Montpellier University Hospital, 34298 Montpellier, France
| | - Rosanna Ferreira-Lopez
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Montpellier University Hospital, 34298 Montpellier, France
| | | | - Christophe Poulet
- Laboratory and Service of Rheumatology, GIGA-I3, Université de Liège, 4000 Liege, Belgium
| | - Olivier Malaise
- Laboratory and Service of Rheumatology, GIGA-I3, Université de Liège, 4000 Liege, Belgium
| | - Dominique De Seny
- Laboratory and Service of Rheumatology, GIGA-I3, Université de Liège, 4000 Liege, Belgium
| | - Jean-Marc Lemaitre
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- SAFE-iPSC Facility INGESTEM, Montpellier University Hospital, 34298 Montpellier, France
| | - Christian Jorgensen
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- Clinical Immunology and Osteoarticular Diseases Therapeutic Unit, Montpellier University Hospital, 34298 Montpellier, France
| | | |
Collapse
|
97
|
Xu KL, Mauck RL, Burdick JA. Modeling development using hydrogels. Development 2023; 150:dev201527. [PMID: 37387575 PMCID: PMC10323241 DOI: 10.1242/dev.201527] [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] [Indexed: 07/01/2023]
Abstract
The development of multicellular complex organisms relies on coordinated signaling from the microenvironment, including both biochemical and mechanical interactions. To better understand developmental biology, increasingly sophisticated in vitro systems are needed to mimic these complex extracellular features. In this Primer, we explore how engineered hydrogels can serve as in vitro culture platforms to present such signals in a controlled manner and include examples of how they have been used to advance our understanding of developmental biology.
Collapse
Affiliation(s)
- Karen L. Xu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Robert L. Mauck
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80303, USA
| |
Collapse
|
98
|
Schwab A, Wesdorp MA, Xu J, Abinzano F, Loebel C, Falandt M, Levato R, Eglin D, Narcisi R, Stoddart MJ, Malda J, Burdick JA, D'Este M, van Osch GJ. Modulating design parameters to drive cell invasion into hydrogels for osteochondral tissue formation. J Orthop Translat 2023; 41:42-53. [PMID: 37691639 PMCID: PMC10485598 DOI: 10.1016/j.jot.2023.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/08/2023] [Accepted: 07/03/2023] [Indexed: 09/12/2023] Open
Abstract
Background The use of acellular hydrogels to repair osteochondral defects requires cells to first invade the biomaterial and then to deposit extracellular matrix for tissue regeneration. Due to the diverse physicochemical properties of engineered hydrogels, the specific properties that allow or even improve the behaviour of cells are not yet clear. The aim of this study was to investigate the influence of various physicochemical properties of hydrogels on cell migration and related tissue formation using in vitro, ex vivo and in vivo models. Methods Three hydrogel platforms were used in the study: Gelatine methacryloyl (GelMA) (5% wt), norbornene hyaluronic acid (norHA) (2% wt) and tyramine functionalised hyaluronic acid (THA) (2.5% wt). GelMA was modified to vary the degree of functionalisation (DoF 50% and 80%), norHA was used with varied degradability via a matrix metalloproteinase (MMP) degradable crosslinker and THA was used with the addition of collagen fibrils. The migration of human mesenchymal stromal cells (hMSC) in hydrogels was studied in vitro using a 3D spheroid migration assay over 48h. In addition, chondrocyte migration within and around hydrogels was investigated in an ex vivo bovine cartilage ring model (three weeks). Finally, tissue repair within osteochondral defects was studied in a semi-orthotopic in vivo mouse model (six weeks). Results A lower DoF of GelMA did not affect cell migration in vitro (p = 0.390) and led to a higher migration score ex vivo (p < 0.001). The introduction of a MMP degradable crosslinker in norHA hydrogels did not improve cell infiltration in vitro or in vivo. The addition of collagen to THA resulted in greater hMSC migration in vitro (p = 0.031) and ex vivo (p < 0.001). Hydrogels that exhibited more cell migration in vitro or ex vivo also showed more tissue formation in the osteochondral defects in vivo, except for the norHA group. Whereas norHA with a degradable crosslinker did not improve cell migration in vitro or ex vivo, it did significantly increase tissue formation in vivo compared to the non-degradable crosslinker (p < 0.001). Conclusion The modification of hydrogels by adapting DoF, use of a degradable crosslinker or including fibrillar collagen can control and improve cell migration and tissue formation for osteochondral defect repair. This study also emphasizes the importance of performing both in vitro and in vivo testing of biomaterials, as, depending on the material, the results might be affected by the model used.The translational potential of this article: This article highlights the potential of using acellular hydrogels to repair osteochondral defects, which are common injuries in orthopaedics. The study provides a deeper understanding of how to modify the properties of hydrogels to control cell migration and tissue formation for osteochondral defect repair. The results of this article also highlight that the choice of the used laboratory model can affect the outcome. Testing hydrogels in different models is thus advised for successful translation of laboratory results to the clinical application.
Collapse
Affiliation(s)
- Andrea Schwab
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
- AO Research Institute Davos, AO Foundation, Davos Platz, Switzerland
| | - Marinus A. Wesdorp
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Jietao Xu
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - Florencia Abinzano
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Claudia Loebel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Marc Falandt
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Utrecht University, Utrecht, the Netherlands
| | - Riccardo Levato
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Utrecht University, Utrecht, the Netherlands
| | - David Eglin
- Mines Saint-Etienne, University Jean Monnet, INSERM, UMR 1059, Saint-Etienne, France
- Advanced Organ Bioengineering and Therapeutics, Faculty of Science and Technology, TechMed Center, University of Twente, Enschede, the Netherlands
| | - Roberto Narcisi
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | | | - Jos Malda
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, the Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Sciences, Utrecht University, Utrecht, the Netherlands
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Matteo D'Este
- AO Research Institute Davos, AO Foundation, Davos Platz, Switzerland
| | - Gerjo J.V.M. van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC, University Medical Center Rotterdam, the Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology, Delft, the Netherlands
| |
Collapse
|
99
|
Wiktorska M, Sacewicz-Hofman I, Niewiarowska J. The endothelial-to-mesenchymal transition changes the focal adhesion site proteins levels and the SLRP-lumican level in HMEC-1 cell line. Exp Cell Res 2023:113692. [PMID: 37392962 DOI: 10.1016/j.yexcr.2023.113692] [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: 01/25/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
Scleroderma, the chronic autoimmune disease is a consequence of inflammation in the connective tissue. Prolonged duration affects formation of compact connective tissue strands (scarring) within the target organ. Endothelial cells undergoing endothelial-to-mesenchymal transition (EndMT) are the source of fibroblast phenotype-resembling cells. EndMT contributes to reorganization of the focal adhesion proteins (FA), including integrins, and intensive extracellular matrix (ECM) remodelling. However, in endothelial cells, the relationship between EndMT and the interaction of integrin receptors with lumican - a component of ECM, is still unclear. Our findings indicate that at the early stages of EndMT caused by Snail-1 transcription factor overexpression, the level of the β1 integrin subunit and its phosphorylation are elevated. Simultaneously, the changes in the level of proteins that build FAs and promote activation of integrin receptors as well as a decrease in lumican quantity were observed. These modulations contributed to increased migration of human microvascular endothelial cells, HMEC-1. Our findings were achieved by WB, ELISA and wound healing assay. Taken altogether, transfection of HMEC-1 cells with Snail-1 plasmids inducing the early stages of EndMT results in the increase of total FAK and integrin β1 phosphorylation as well as cell migration: phenomena which are modulated by interaction with lumican.
Collapse
Affiliation(s)
- Magdalena Wiktorska
- Department of Molecular Cell Mechanisms, Medical University of Lodz, 92-215, Lodz, Poland
| | | | - Jolanta Niewiarowska
- Department of Molecular Cell Mechanisms, Medical University of Lodz, 92-215, Lodz, Poland.
| |
Collapse
|
100
|
Castro VIB, Araújo AR, Duarte F, Sousa-Franco A, Reis RL, Pashkuleva I, Pires RA. Glycopeptide-Based Supramolecular Hydrogels Induce Differentiation of Adipose Stem Cells into Neural Lineages. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37327399 DOI: 10.1021/acsami.3c05309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
We applied a bottom-up approach to develop biofunctional supramolecular hydrogels from an aromatic glycodipeptide. The self-assembly of the glycopeptide was induced by either temperature manipulation (heating-cooling cycle) or solvent (DMSO to water) switch. The sol-gel transition was salt-triggered in cell culture media and resulted in gels with the same chemical compositions but different mechanical properties. Human adipose derived stem cells (hASCs) cultured on these gels under basal conditions (i.e., without differentiation factors) overexpressed neural markers, such as GFAP, Nestin, MAP2, and βIII-tubulin, confirming the differentiation into neural lineages. The mechanical properties of the gels influenced the number and distribution of the adhered cells. A comparison with gels obtained from the nonglycosylated peptide showed that glycosylation is crucial for the biofunctionality of the hydrogels by capturing and preserving essential growth factors, e.g., FGF-2.
Collapse
Affiliation(s)
- Vânia I B Castro
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal
- ICVS/3B's─PT Government Associated Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Ana R Araújo
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal
- ICVS/3B's─PT Government Associated Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Filipa Duarte
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal
- ICVS/3B's─PT Government Associated Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - António Sousa-Franco
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal
- ICVS/3B's─PT Government Associated Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal
- ICVS/3B's─PT Government Associated Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Iva Pashkuleva
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal
- ICVS/3B's─PT Government Associated Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Ricardo A Pires
- 3B's Research Group, I3Bs─Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, 4805-017 Guimarães, Portugal
- ICVS/3B's─PT Government Associated Laboratory, 4805-017 Braga/Guimarães, Portugal
| |
Collapse
|