1
|
Bianchi E, Ruggeri M, Vigani B, Aguzzi C, Rossi S, Sandri G. Synthesis and use of thermoplastic polymers for tissue engineering purposes. Int J Pharm X 2025; 9:100313. [PMID: 39807177 PMCID: PMC11729033 DOI: 10.1016/j.ijpx.2024.100313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/27/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025] Open
Abstract
Thermoplastic polymers provide a versatile platform to mimic various aspects of physiological extracellular matrix properties such as chemical composition, stiffness, and topography for use in cell and tissue engineering applications. In this review, we provide a brief overview of the most promising thermoplastic polymers, and in particular the thermoplastic polyesters, such as poly(lactic acid), poly(glycolic acid), and polycaprolactone, and the thermoplastic elastomers, such as polyurethanes, polyhydroxyalkanoates, and poly(butyl cyanoacrylate). A particular focus has been made on the synthesis processes, the processability and the biocompatibility. We also discuss how these materials can be applied in tissue engineering, mimicking tissues' structure and function, and stimulate mesenchymal stem cells differentiation and mechanotransduction.
Collapse
Affiliation(s)
- Eleonora Bianchi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Marco Ruggeri
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Barbara Vigani
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Carola Aguzzi
- Department of Pharmacy and Pharmaceutical Technology, University of Granada, Cartuja Campus, Granada 18071, Spain
| | - Silvia Rossi
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| | - Giuseppina Sandri
- Department of Drug Sciences, University of Pavia, Viale Taramelli 12, 27100 Pavia, Italy
| |
Collapse
|
2
|
Akagi S, Ando H, Matsuo CNA, Tajima K, Takata H, Matsushima T, Kusano T, Ishida T. A 3D Cell-Culture System That Uses Nano-Fibrillated Bacterial Cellulose to Prepare a Spherical Formulation of Culture Cells. Biol Pharm Bull 2025; 48:23-32. [PMID: 39864853 DOI: 10.1248/bpb.b24-00804] [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: 01/28/2025]
Abstract
A 3-dimensional (3D) cell culture is now being actively pursued to accomplish the in vivo-like cellular morphology and biological functions in cell culture. We recently obtained nano-fibrillated bacterial cellulose (NFBC). In this study, we developed a novel NFBC-based 3D cell-culture system, the OnGel method, and the Suspension method. HepG2 human liver cancer cells were cultured via these methods and formed spherical formulations in the optimized condition, 1.0% (w/v) of NFBC in the OnGel method, and 0.06-0.10% (w/v) of NFBC in the Suspension method. Non-cancerous cells such as human-induced pluripotent stem (iPS) cells and human mesenchymal stem cells (MSCs) also formed spherical formulations. It is noteworthy that both the size and cell viability of spheroids prepared via these methods were comparable to those cultured using commercially available 3D cell-culture systems. Both OnGel and Suspension methods are less complicated than the existing 3D cell-culture systems, which is an invaluable advantage for the preparation of cancer spheroids. The NFBC-based 3D cell-culture systems introduced here show great promise as a tool to prepare cultures for cell-derived spheroids for the progress of both in vitro and in vivo studies of the biological functioning of cells.
Collapse
Affiliation(s)
- Shunsuke Akagi
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
- Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Cristina Nana Amorim Matsuo
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Kenji Tajima
- Faculty of Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
- Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | | | | | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
- Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| |
Collapse
|
3
|
Sinha B, Biswas A, Kaushik S, Soni GV. Cellular and Nuclear Forces: An Overview. Methods Mol Biol 2025; 2881:3-39. [PMID: 39704936 DOI: 10.1007/978-1-0716-4280-1_1] [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: 12/21/2024]
Abstract
Biological cells sample their surrounding microenvironments using nanoscale force sensors on the cell surfaces. These surface-based force and stress sensors generate physical and chemical responses inside the cell. The inherently well-connected cytoskeleton and its physical contacts with the force elements on the nuclear membrane lead these physicochemical responses to cascade all the way inside the cell nucleus, physically altering the nuclear state. These physical alterations of the cell nucleus, through yet-unknown complex steps, elicit physical and functional responses from the chromatin in the form of altered gene expression profiles. This mechanism of force/stress sensing by the cell and then its nuclear response has been shown to play a vital role in maintaining robust cellular homeostasis, controlling gene expression profiles during developmental phases as well as cell differentiation. In the last few years, there has been appreciable progress toward the identification of the molecular players responsible for force sensing. However, the actual sensing mechanism of cell surface-bound force sensors and more importantly cascading of the signals, both physical (via cytosolic force sensing elements such as microtubule and actin framework) as well as chemical (cascade of biochemical signaling from cell surface to nuclear surface and further to the chromatin), inside the cell is poorly understood. In this chapter, we present a review of the currently known molecular players in cellular as well as nuclear force sensing repertoire and their possible mechanistic aspects. We also introduce various biophysical concepts and review some frequently used techniques that are used to describe the force/stress sensing and response of a cell. We hope that this will help in asking clearer questions and designing pointed experiments for better understanding of the force-dependent design principles of the cell surface, nuclear surface, and gene expression.
Collapse
Affiliation(s)
- Bidisha Sinha
- Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | - Arikta Biswas
- Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India
| | | | - Gautam V Soni
- Raman Research Institute, Bangalore, Karnataka, India.
| |
Collapse
|
4
|
Chang KC, Silvestri F, Oliphant MUJ, Martinez-Gakidis MA, Orgill DP, Garber JE, Dillon DD, Brugge JS. Breast organoid suspension cultures maintain long-term estrogen receptor expression and responsiveness. NPJ Breast Cancer 2024; 10:107. [PMID: 39702422 DOI: 10.1038/s41523-024-00714-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/27/2024] [Indexed: 12/21/2024] Open
Abstract
Organoid cultures offer a powerful technology to investigate many different aspects of development, physiology, and pathology of diverse tissues. Unlike standard tissue culture of primary breast epithelial cells, breast organoids preserve the epithelial lineages and architecture of the normal tissue. However, existing organoid culture methods are tedious, difficult to scale, and do not robustly retain estrogen receptor (ER) expression and responsiveness in long-term culture. Here, we describe a modified culture method to generate and maintain organoids as suspension cultures in reconstituted basement membrane (™Matrigel). This method improves organoid growth and uniformity compared to the conventional Matrigel dome embedding method, while maintaining the fidelity of the three major epithelial lineages. Using this adopted method, we are able to culture and passage purified hormone sensing (HS) cells that retain ER responsiveness upon estrogen stimulation in long-term culture. This culture system presents a valuable platform to study the events involved in initiation and evolution of ER-positive breast cancer.
Collapse
Affiliation(s)
- Kung-Chi Chang
- Department of Cell Biology and Ludwig Center at Harvard, Harvard Medical School, 240 Longwood Ave, Boston, MA, 02115, USA
| | - Francesca Silvestri
- Department of Cell Biology and Ludwig Center at Harvard, Harvard Medical School, 240 Longwood Ave, Boston, MA, 02115, USA
| | - Michael U J Oliphant
- Department of Cell Biology and Ludwig Center at Harvard, Harvard Medical School, 240 Longwood Ave, Boston, MA, 02115, USA
| | - M Angie Martinez-Gakidis
- Department of Cell Biology and Ludwig Center at Harvard, Harvard Medical School, 240 Longwood Ave, Boston, MA, 02115, USA
| | - Dennis P Orgill
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Brigham & Women's Hospital, 75 Francis St, Boston, MA, 02115, USA
| | - Judy E Garber
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Ave, Boston, MA, 02115, USA
| | - Deborah D Dillon
- Department of Pathology, Brigham & Women's Hospital, 75 Francis St, Boston, MA, 02115, USA
| | - Joan S Brugge
- Department of Cell Biology and Ludwig Center at Harvard, Harvard Medical School, 240 Longwood Ave, Boston, MA, 02115, USA.
| |
Collapse
|
5
|
Feng X, Cao F, Wu X, Xie W, Wang P, Jiang H. Targeting extracellular matrix stiffness for cancer therapy. Front Immunol 2024; 15:1467602. [PMID: 39697341 PMCID: PMC11653020 DOI: 10.3389/fimmu.2024.1467602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/06/2024] [Indexed: 12/20/2024] Open
Abstract
The physical characteristics of the tumor microenvironment (TME) include solid stress, interstitial fluid pressure, tissue stiffness and microarchitecture. Among them, abnormal changes in tissue stiffness hinder drug delivery, inhibit infiltration of immune killer cells to the tumor site, and contribute to tumor resistance to immunotherapy. Therefore, targeting tissue stiffness to increase the infiltration of drugs and immune cells can offer a powerful support and opportunities to improve the immunotherapy efficacy in solid tumors. In this review, we discuss the mechanical properties of tumors, the impact of a stiff TME on tumor cells and immune cells, and the strategies to modulate tumor mechanics.
Collapse
Affiliation(s)
- Xiuqin Feng
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fujun Cao
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiangji Wu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenyan Xie
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Jiang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Wang X, Komasa S, Tahara Y, Inui S, Matsumoto M, Maekawa K. Novel Injectable Collagen/Glycerol/Pullulan Gel Promotes Osteogenic Differentiation of Mesenchymal Stem Cells and the Repair of Rat Cranial Defects. Gels 2024; 10:775. [PMID: 39727533 DOI: 10.3390/gels10120775] [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: 10/06/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Bone tissue engineering is a technique that simulates the bone tissue microenvironment by utilizing cells, tissue scaffolds, and growth factors. The collagen hydrogel is a three-dimensional network bionic material that has properties and structures comparable to those of the extracellular matrix (ECM), making it an ideal scaffold and drug delivery system for tissue engineering. The clinical applications of this material are restricted due to its low mechanical strength. In this investigation, a collagen-based gel (atelocollagen/glycerol/pullulan [Col/Gly/Pul] gel) that is moldable and injectable with high adhesive qualities was created by employing a straightforward technique that involved the introduction of Gly and Pul. This study aimed to characterize the internal morphology and chemical composition of the Col/Gly/Pul gel, as well as to verify its osteogenic properties through in vivo and in vitro experiments. When compared to a standard pure Col hydrogel, this material is more adaptable to the complexity of the local environment of bone defects and the apposition of irregularly shaped flaws due to its greater mechanical strength, injectability, and moldability. Overall, the Col/Gly/Pul gel is an implant that shows great potential for the treatment of complex bone defects and the enhancement of bone regeneration.
Collapse
Affiliation(s)
- Xin Wang
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1, Kuzuhahanazono-cho, Hirakata-shi 573-1121, Osaka, Japan
| | - Satoshi Komasa
- Department of Oral Health Sciences, Osaka Dental University, 1-4-4, Makino-honmachi, Hirakata-shi 573-1144, Osaka, Japan
| | - Yoshiro Tahara
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe 610-0321, Kyoto, Japan
| | - Shihoko Inui
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1, Kuzuhahanazono-cho, Hirakata-shi 573-1121, Osaka, Japan
| | - Michiaki Matsumoto
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara-Miyakodani, Kyotanabe 610-0321, Kyoto, Japan
| | - Kenji Maekawa
- Department of Removable Prosthodontics and Occlusion, Osaka Dental University, 8-1, Kuzuhahanazono-cho, Hirakata-shi 573-1121, Osaka, Japan
| |
Collapse
|
7
|
Soliman BG, Chin IL, Li Y, Ishii M, Ho MH, Doan VK, Cox TR, Wang PY, Lindberg GCJ, Zhang YS, Woodfield TBF, Choi YS, Lim KS. Droplet-based microfluidics for engineering shape-controlled hydrogels with stiffness gradient. Biofabrication 2024; 16:045026. [PMID: 39121873 DOI: 10.1088/1758-5090/ad6d8e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
Current biofabrication strategies are limited in their ability to replicate native shape-to-function relationships, that are dependent on adequate biomimicry of macroscale shape as well as size and microscale spatial heterogeneity, within cell-laden hydrogels. In this study, a novel diffusion-based microfluidics platform is presented that meets these needs in a two-step process. In the first step, a hydrogel-precursor solution is dispersed into a continuous oil phase within the microfluidics tubing. By adjusting the dispersed and oil phase flow rates, the physical architecture of hydrogel-precursor phases can be adjusted to generate spherical and plug-like structures, as well as continuous meter-long hydrogel-precursor phases (up to 1.75 m). The second step involves the controlled introduction a small molecule-containing aqueous phase through a T-shaped tube connector to enable controlled small molecule diffusion across the interface of the aqueous phase and hydrogel-precursor. Application of this system is demonstrated by diffusing co-initiator sodium persulfate (SPS) into hydrogel-precursor solutions, where the controlled SPS diffusion into the hydrogel-precursor and subsequent photo-polymerization allows for the formation of unique radial stiffness patterns across the shape- and size-controlled hydrogels, as well as allowing the formation of hollow hydrogels with controllable internal architectures. Mesenchymal stromal cells are successfully encapsulated within hollow hydrogels and hydrogels containing radial stiffness gradient and found to respond to the heterogeneity in stiffness through the yes-associated protein mechano-regulator. Finally, breast cancer cells are found to phenotypically switch in response to stiffness gradients, causing a shift in their ability to aggregate, which may have implications for metastasis. The diffusion-based microfluidics thus finds application mimicking native shape-to-function relationship in the context of tissue engineering and provides a platform to further study the roles of micro- and macroscale architectural features that exist within native tissues.
Collapse
Affiliation(s)
- Bram G Soliman
- Light Activated Biomaterials (LAB) Group, University of Otago, Christchurch 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
- School of Material Science and Engineering, University of New South Wales, Sydney 2052, Australia
| | - Ian L Chin
- School of Human Sciences, The University of Western Australia, Perth 6009, Australia
| | - Yiwei Li
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| | - Melissa Ishii
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
| | - Minh Hieu Ho
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| | - Vinh Khanh Doan
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| | - Thomas R Cox
- The Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Peng Yuan Wang
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang 32500, People's Republic of China
| | - Gabriella C J Lindberg
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
- Phil and Penny Knight Campus for Accelerating Scientific Impact Department of Bioengineering, University of Oregon, Eugene, OR, United States of America
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States of America
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
| | - Yu Suk Choi
- School of Human Sciences, The University of Western Australia, Perth 6009, Australia
| | - Khoon S Lim
- Light Activated Biomaterials (LAB) Group, University of Otago, Christchurch 8011, New Zealand
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, University of Otago, Christchurch 8011, New Zealand
- School of Medical Sciences, Charles Perkins Centre, The University of Sydney, Sydney 2006, Australia
| |
Collapse
|
8
|
Oliverio R, Liberelle B, Patenaude V, Moreau V, Thomas E, Virgilio N, Banquy X, De Crescenzo G. Cofunctionalization of Macroporous Dextran Hydrogels with Adhesive Peptides and Growth Factors Enables Vascular Spheroid Sprouting. ACS Biomater Sci Eng 2024; 10:5080-5093. [PMID: 39038278 DOI: 10.1021/acsbiomaterials.4c00455] [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: 07/24/2024]
Abstract
Ensuring good definition of scaffolds used for 3D cell culture is a prominent challenge that hampers the development of tissue engineering platforms. Since dextran repels cell adhesion, using dextran-based materials biofunctionalized through a bottom-up approach allows for precise control over material definition. Here, we report the design of dextran hydrogels displaying a fully interconnected macropore network for the culture of vascular spheroids in vitro. We biofunctionalized the hydrogels with the RGD peptide sequence to promote cell adhesion. We used an affinity peptide pair, the E/K coiled coil, to load the gels with epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF). Dual functionalization with adhesive and proliferative cues allows vascular spheroids to colonize naturally cell-repellant dextran. In supplement-depleted medium, we report improved colonization of the macropores compared to that of unmodified dextran. Altogether, we propose a well-defined and highly versatile platform for tissue engineering and tissue vascularization applications.
Collapse
Affiliation(s)
- Romane Oliverio
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
- Faculty of Pharmacy, Axe Formulation et Analyse du Médicament (AFAM), Université de Montréal, Montréal H3T 1J4, Québec, Canada
| | - Benoît Liberelle
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| | - Victor Patenaude
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| | - Vaiana Moreau
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
- Department of Chemical Engineering, Centre de Recherche sur les Systèmes Polymères et Composites à Haute Performance (CREPEC), Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| | - Elian Thomas
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| | - Nick Virgilio
- Department of Chemical Engineering, Centre de Recherche sur les Systèmes Polymères et Composites à Haute Performance (CREPEC), Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| | - Xavier Banquy
- Faculty of Pharmacy, Axe Formulation et Analyse du Médicament (AFAM), Université de Montréal, Montréal H3T 1J4, Québec, Canada
| | - Gregory De Crescenzo
- Department of Chemical Engineering, Polytechnique Montréal, Montréal H3T 1J4, Québec, Canada
| |
Collapse
|
9
|
Gonçalves PP, da Silva CL, Bernardes N. Advancing cancer therapeutics: Integrating scalable 3D cancer models, extracellular vesicles, and omics for enhanced therapy efficacy. Adv Cancer Res 2024; 163:137-185. [PMID: 39271262 DOI: 10.1016/bs.acr.2024.07.001] [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: 09/15/2024]
Abstract
Cancer remains as one of the highest challenges to human health. However, anticancer drugs exhibit one of the highest attrition rates compared to other therapeutic interventions. In part, this can be attributed to a prevalent use of in vitro models with limited recapitulative potential of the in vivo settings. Three dimensional (3D) models, such as tumor spheroids and organoids, offer many research opportunities to address the urgent need in developing models capable to more accurately mimic cancer biology and drug resistance profiles. However, their wide adoption in high-throughput pre-clinical studies is dependent on scalable manufacturing to support large-scale therapeutic drug screenings and multi-omic approaches for their comprehensive cellular and molecular characterization. Extracellular vesicles (EVs), which have been emerging as promising drug delivery systems (DDS), stand to significantly benefit from such screenings conducted in realistic cancer models. Furthermore, the integration of these nanomedicines with 3D cancer models and omics profiling holds the potential to deepen our understanding of EV-mediated anticancer effects. In this chapter, we provide an overview of the existing 3D models used in cancer research, namely spheroids and organoids, the innovations in their scalable production and discuss how omics can facilitate the implementation of these models at different stages of drug testing. We also explore how EVs can advance drug delivery in cancer therapies and how the synergy between 3D cancer models and omics approaches can benefit in this process.
Collapse
Affiliation(s)
- Pedro P Gonçalves
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Nuno Bernardes
- Department of Bioengineering and iBB - Institute for Bioengineering and Biosciences at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
10
|
Brugge J, Chang KC, Silvestri F, Olipant M, Martinez-Gakidis MA, Orgill D, Garber J, Dillon D. Breast organoid suspension cultures maintain long-term estrogen receptor expression and responsiveness. RESEARCH SQUARE 2024:rs.3.rs-4463390. [PMID: 38947074 PMCID: PMC11213202 DOI: 10.21203/rs.3.rs-4463390/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Organoid cultures offer a powerful technology to investigate many different aspects of development, physiology, and pathology of diverse tissues. Unlike standard tissue culture of primary breast epithelial cells, breast organoids preserve the epithelial lineages and architecture of the normal tissue. However, existing organoid culture methods are tedious, difficult to scale, and do not robustly retain estrogen receptor (ER) expression and responsiveness in long-term culture. Here, we describe a modified culture method to generate and maintain organoids as suspension cultures in reconstituted basement membrane (™Matrigel). This method improves organoid growth and uniformity compared to the conventional Matrigel dome embedding method, while maintaining the fidelity of the three major epithelial lineages. Using this adopted method, we are able to culture and passage purified hormone sensing (HS) cells that retain ER responsiveness upon estrogen stimulation in long-term culture. This culture system presents a valuable platform to study the events involved in initiation and evolution of ER-positive breast cancer.
Collapse
|
11
|
Schuphan J, Stojanović N, Lin YY, Buhl EM, Aveic S, Commandeur U, Schillberg S, Fischer H. A Combination of Flexible Modified Plant Virus Nanoparticles Enables Additive Effects Resulting in Improved Osteogenesis. Adv Healthc Mater 2024; 13:e2304243. [PMID: 38417028 DOI: 10.1002/adhm.202304243] [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: 11/30/2023] [Revised: 02/16/2024] [Indexed: 03/01/2024]
Abstract
Plant virus nanoparticles (VNPs) genetically engineered to present osteogenic cues provide a promising method for biofunctionalizing hydrogels in bone tissue engineering. Flexible Potato virus X (PVX) nanoparticles substantially enhance the attachment and differentiation of human mesenchymal stem cells (hMSCs) by presenting the RGD motif, hydroxyapatite-binding peptide (HABP), or consecutive polyglutamates (E8) in a concentration-dependent manner. Therefore, it is hypothesized that Tobacco mosaic virus nanoparticles, which present 1.6 times more functional peptides than PVX, will meliorate such an impact. This study hypothesizes that cultivating hMSCs on a surface coated with a combination of two VNPs presenting peptides for either cell attachment or mineralization can achieve additionally enhancing effects on osteogenesis. Calcium minerals deposited by differentiating hMSCs increases two to threefold for this combination, while the Alkaline Phosphatase activity of hMSCs grown on the PVX-RGD/PVX-HABP-coated surface significantly surpasses any other VNP combination. Superior additive effects are observed for the first time by employing a combination of VNPs with varying functionalities. It is found that the flexible VNP geometry plays a more critical role than the concentration of functional peptides. In conclusion, various peptide-presenting plant VNPs exhibit an additive enhancing effect offering significant potential for effectively functionalizing cell-containing hydrogels in bone tissue engineering.
Collapse
Affiliation(s)
- Juliane Schuphan
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Natalija Stojanović
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ying-Ying Lin
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Sanja Aveic
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ulrich Commandeur
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Stefan Schillberg
- Institute for Molecular Biotechnology, RWTH Aachen University, Worringerweg 1, 52074, Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany
| |
Collapse
|
12
|
Wong CA, Fraticelli Guzmán NS, Read AT, Hedberg-Buenz A, Anderson MG, Feola AJ, Sulchek T, Ethier CR. A method for analyzing AFM force mapping data obtained from soft tissue cryosections. J Biomech 2024; 168:112113. [PMID: 38648717 PMCID: PMC11128031 DOI: 10.1016/j.jbiomech.2024.112113] [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: 11/16/2023] [Revised: 02/23/2024] [Accepted: 04/17/2024] [Indexed: 04/25/2024]
Abstract
Atomic force microscopy (AFM) is a valuable tool for assessing mechanical properties of biological samples, but interpretations of measurements on whole tissues can be difficult due to the tissue's highly heterogeneous nature. To overcome such difficulties and obtain more robust estimates of tissue mechanical properties, we describe an AFM force mapping and data analysis pipeline to characterize the mechanical properties of cryosectioned soft tissues. We assessed this approach on mouse optic nerve head and rat trabecular meshwork, cornea, and sclera. Our data show that the use of repeated measurements, outlier exclusion, and log-normal data transformation increases confidence in AFM mechanical measurements, and we propose that this methodology can be broadly applied to measuring soft tissue properties from cryosections.
Collapse
Affiliation(s)
- Cydney A Wong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | | | - A Thomas Read
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Adam Hedberg-Buenz
- Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA
| | - Michael G Anderson
- Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA
| | - Andrew J Feola
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Department of Ophthalmology, Emory University, Atlanta, GA; Center for Visual & Neurocognitive Rehabilitation, Atlanta Veterans Affairs Medical Center, Decatur, GA
| | - Todd Sulchek
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - C Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA; Department of Ophthalmology, Emory University, Atlanta, GA.
| |
Collapse
|
13
|
Cheng X, Xu B, Lei B, Wang S. Opposite Mechanical Preference of Bone/Nerve Regeneration in 3D-Printed Bioelastomeric Scaffolds/Conduits Consistently Correlated with YAP-Mediated Stem Cell Osteo/Neuro-Genesis. Adv Healthc Mater 2024; 13:e2301158. [PMID: 38211963 DOI: 10.1002/adhm.202301158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 12/31/2023] [Indexed: 01/13/2024]
Abstract
To systematically unveil how substrate stiffness, a critical factor in directing cell fate through mechanotransduction, correlates with tissue regeneration, novel biodegradable and photo-curable poly(trimethylene carbonate) fumarates (PTMCFs) for fabricating elastomeric 2D substrates and 3D bone scaffolds/nerve conduits, are presented. These substrates and structures with adjustable stiffness serve as a unique platform to evaluate how this mechanical cue affects the fate of human umbilical cord mesenchymal stem cells (hMSCs) and hard/soft tissue regeneration in rat femur bone defect and sciatic nerve transection models; whilst, decoupling from topographical and chemical cues. In addition to a positive relationship between substrate stiffness (tensile modulus: 90-990 kPa) and hMSC adhesion, spreading, and proliferation mediated through Yes-associated protein (YAP), opposite mechanical preference is revealed in the osteogenesis and neurogenesis of hMSCs as they are significantly enhanced on the stiff and compliant substrates, respectively. In vivo tissue regeneration demonstrates the same trend: bone regeneration prefers the stiffer scaffolds; while, nerve regeneration prefers the more compliant conduits. Whole-transcriptome analysis further shows that upregulation of Rho GTPase activity and the downstream genes in the compliant group promote nerve repair, providing critical insight into the design strategies of biomaterials for stem cell regulation and hard/soft tissue regeneration through mechanotransduction.
Collapse
Affiliation(s)
- Xiaopeng Cheng
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Bowen Xu
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Bingxi Lei
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510006, China
| | - Shanfeng Wang
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| |
Collapse
|
14
|
Yoon J, Han H, Jang J. Nanomaterials-incorporated hydrogels for 3D bioprinting technology. NANO CONVERGENCE 2023; 10:52. [PMID: 37968379 PMCID: PMC10651626 DOI: 10.1186/s40580-023-00402-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
In the field of tissue engineering and regenerative medicine, various hydrogels derived from the extracellular matrix have been utilized for creating engineered tissues and implantable scaffolds. While these hydrogels hold immense promise in the healthcare landscape, conventional bioinks based on ECM hydrogels face several challenges, particularly in terms of lacking the necessary mechanical properties required for 3D bioprinting process. To address these limitations, researchers are actively exploring novel nanomaterial-reinforced ECM hydrogels for both mechanical and functional aspects. In this review, we focused on discussing recent advancements in the fabrication of engineered tissues and monitoring systems using nanobioinks and nanomaterials via 3D bioprinting technology. We highlighted the synergistic benefits of combining numerous nanomaterials into ECM hydrogels and imposing geometrical effects by 3D bioprinting technology. Furthermore, we also elaborated on critical issues remaining at the moment, such as the inhomogeneous dispersion of nanomaterials and consequent technical and practical issues, in the fabrication of complex 3D structures with nanobioinks and nanomaterials. Finally, we elaborated on plausible outlooks for facilitating the use of nanomaterials in biofabrication and advancing the function of engineered tissues.
Collapse
Affiliation(s)
- Jungbin Yoon
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Hohyeon Han
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- Institute of Convergence Science, Yonsei University, Seoul, South Korea.
| |
Collapse
|
15
|
Wong CA, Fraticelli Guzmán NS, Read AT, Hedberg-Buenz A, Anderson MG, Feola AJ, Sulchek T, Ethier CR. A Method for Analyzing AFM Force Mapping Data Obtained from Soft Tissue Cryosections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566263. [PMID: 38014311 PMCID: PMC10680563 DOI: 10.1101/2023.11.08.566263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Atomic force microscopy (AFM) is a valuable tool for assessing mechanical properties of biological samples, but interpretations of measurements on whole tissues can be difficult due to the tissue's highly heterogeneous nature. To overcome such difficulties and obtain more robust estimates of tissue mechanical properties, we describe an AFM force mapping and data analysis pipeline to characterize the mechanical properties of cryosectioned soft tissues. We assessed this approach on mouse optic nerve head and rat trabecular meshwork, cornea, and sclera. Our data show that the use of repeated measurements, outlier exclusion, and log-normal data transformation increases confidence in AFM mechanical measurements, and we propose that this methodology can be broadly applied to measuring soft tissue properties from cryosections.
Collapse
Affiliation(s)
- Cydney A Wong
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | | | - A Thomas Read
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
| | - Adam Hedberg-Buenz
- Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA
| | - Michael G Anderson
- Department of Molecular Physiology & Biophysics, University of Iowa, Iowa City, IA
| | - Andrew J Feola
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Department of Ophthalmology, Emory University, Atlanta, GA
- Center for Visual & Neurocognitive Rehabilitation, Atlanta VA Medical Center, Atlanta GA
| | - Todd Sulchek
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - C Ross Ethier
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Ophthalmology, Emory University, Atlanta, GA
| |
Collapse
|
16
|
Abalymov A, Pinchasik BE, Akasov RA, Lomova M, Parakhonskiy BV. Strategies for Anisotropic Fibrillar Hydrogels: Design, Cell Alignment, and Applications in Tissue Engineering. Biomacromolecules 2023; 24:4532-4552. [PMID: 37812143 DOI: 10.1021/acs.biomac.3c00503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Efficient cellular alignment in biomaterials presents a considerable challenge, demanding the refinement of appropriate material morphologies, while ensuring effective cell-surface interactions. To address this, biomaterials are continuously researched with diverse coatings, hydrogels, and polymeric surfaces. In this context, we investigate the influence of physicochemical parameters on the architecture of fibrillar hydrogels that significantly orient the topography of flexible hydrogel substrates, thereby fostering cellular adhesion and spatial organization. Our Review comprehensively assesses various techniques for aligning polymer fibrils within hydrogels, specifically interventions applied during and after the cross-linking process. These methodologies include mechanical strains, precise temperature modulation, controlled fluidic dynamics, and chemical modulators, as well as the use of magnetic and electric fields. We highlight the intrinsic appeal of these methodologies in fabricating cell-aligning interfaces and discuss their potential implications within the fields of biomaterials and tissue engineering, particularly concerning the pursuit of optimal cellular alignment.
Collapse
Affiliation(s)
- Anatolii Abalymov
- Science Medical Center, Saratov State University, 410012 Saratov, Russia
| | - Bat-El Pinchasik
- School of Mechanical Engineering, Faculty of Engineering, Tel-Aviv University, 69978 Tel-Aviv, Israel
| | - Roman A Akasov
- Sechenov University and Federal Scientific Research Centre "Crystallography and Photonics" of Russian Academy of Sciences, 101000 Moscow, Russia
| | - Maria Lomova
- Science Medical Center, Saratov State University, 410012 Saratov, Russia
| | - Bogdan V Parakhonskiy
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
17
|
Shi X, Janmey PA. Large Polyacrylamide Hydrogels for Large-Batch Cell Culture and Mechanobiological Studies. Macromol Biosci 2023; 23:e2300042. [PMID: 37128976 PMCID: PMC10524403 DOI: 10.1002/mabi.202300042] [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: 02/16/2023] [Revised: 03/24/2023] [Indexed: 05/03/2023]
Abstract
The rigidity of a cell's substrate or extracellular matrix plays a vital role in regulating cell and tissue functions. Polyacrylamide (PAAm) hydrogels are one of the most widely used cell culture substrates that provide a physiologically relevant range of stiffness. However, it is still arduous and time-consuming to prepare PAAm substrates in large batches for high-yield or multiscale cell cultures. In this communication, a simple method to prepare PAAm hydrogels with less time cost and easily accessible materials is presented. The hydrogel is mechanically uniform and supports cell culture in a large batch. It is further shown that the stiffness of the hydrogel covers a large range of Young's modulus and is sensed by cells, regulating various cell features including changes in cell morphology, proliferation, and contractility. This method improves the reproducibility of mechanobiology studies and can be easily applied for mechanobiology research requiring large numbers of cells or experimental groups.
Collapse
Affiliation(s)
- Xuechen Shi
- Institute for Medicine and Engineering and Department of Physiology, University of Pennsylvania, Philadelphia, 19104, USA
| | - Paul A Janmey
- Institute for Medicine and Engineering and Department of Physiology, University of Pennsylvania, Philadelphia, 19104, USA
| |
Collapse
|
18
|
Cortes Ruiz MF, Brusentsev Y, Lindström SB, Xu C, Wågberg L. Shape-recovering nanocellulose networks: Preparation, characterization and modeling. Carbohydr Polym 2023; 315:120950. [PMID: 37230608 DOI: 10.1016/j.carbpol.2023.120950] [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: 01/31/2023] [Revised: 03/20/2023] [Accepted: 04/22/2023] [Indexed: 05/27/2023]
Abstract
Development of strong cellulose nanofibril (CNF) networks for advanced applications, such as in the biomedical field, is of high importance owing to the biocompatible nature and plant-based origin of cellulose nanofibrils. Nevertheless, lack of mechanical strength and complex synthesis methods hinder the application of these materials in areas where both toughness and manufacturing simplicity are required. In this work, we introduce a facile method for the synthesis of a low solid content (< 2 wt%), covalently crosslinked CNF hydrogel where Poly (N-isopropylacrylamide) (NIPAM) chains are utilized as crosslinks between the nanofibrils. The resulting networks have the capability to fully recover the shape in which they were formed after various drying and rewetting cycles. Characterization of the hydrogel and its constitutive components was performed using X-ray scattering, rheological investigations and uniaxial testing in compression. Influence of covalent crosslinks was compared with networks crosslinked by the addition of CaCl2. Among other things the results show that the mechanical properties of the hydrogels can be tuned by controlling the ionic strength of the surrounding medium. Finally, a mathematical model was developed based on the experimental results, which describes and predicts to a decent degree the large-deformation, elastoplastic behavior, and fracture of these networks.
Collapse
Affiliation(s)
- Maria F Cortes Ruiz
- Fiber Technology Division, Fiber and Polymer Technology Department, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden; Wallenberg Wood Science Center, Fiber and Polymer Technology Department, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden
| | - Yury Brusentsev
- Laboratory of Natural Materials Technology, Åbo Akademi University, 20500 Åbo, Finland
| | | | - Chunlin Xu
- Laboratory of Natural Materials Technology, Åbo Akademi University, 20500 Åbo, Finland
| | - Lars Wågberg
- Fiber Technology Division, Fiber and Polymer Technology Department, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden; Wallenberg Wood Science Center, Fiber and Polymer Technology Department, KTH Royal Institute of Technology, 114 28 Stockholm, Sweden.
| |
Collapse
|
19
|
Naim G, Bruchiel-Spanier N, Betsis S, Eliaz N, Mandler D. Vat Polymerization by Three-Dimensional Printing and Curing of Antibacterial Zinc Oxide Nanoparticles Embedded in Poly(ethylene glycol) Diacrylate for Biomedical Applications. Polymers (Basel) 2023; 15:3586. [PMID: 37688212 PMCID: PMC10490083 DOI: 10.3390/polym15173586] [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: 08/14/2023] [Revised: 08/24/2023] [Accepted: 08/27/2023] [Indexed: 09/10/2023] Open
Abstract
Digital light processing (DLP) is a vat photopolymerization 3D printing technique with increasingly broad application prospects, particularly in personalized medicine, such as the creation of medical devices. Different resins and printing parameters affect the functionality of these devices. One of the many problems that biomedical implants encounter is inflammation and bacteria growth. For this reason, many studies turn to the addition of antibacterial agents to either the bulk material or as a coating. Zinc oxide nanoparticles (ZnO NPs) have shown desirable properties, including antibacterial activity with negligible toxicity to the human body, allowing their use in a wide range of applications. In this project, we developed a resin of poly(ethylene glycol) diacrylate (PEGDA), a cross-linker known for its excellent mechanical properties and high biocompatibility in a 4:1 weight ratio of monomers to water. The material's mechanical properties (Young's modulus, maximum elongation, and ultimate tensile strength) were found similar to those of human cartilage. Furthermore, the ZnO NPs embedding matrix showed strong antibacterial activity against Escherichia coli (E. coli) and Staphylococcus aureus (S.A.). As the ZnO NPs ratio was changed, only a minor effect on the mechanical properties of the material was observed, whereas strong antibacterial properties against both bacteria were achieved in the case of 1.5 wt.% NPs.
Collapse
Affiliation(s)
- Guy Naim
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (G.N.); (N.B.-S.); (S.B.)
| | - Netta Bruchiel-Spanier
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (G.N.); (N.B.-S.); (S.B.)
| | - Shelly Betsis
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (G.N.); (N.B.-S.); (S.B.)
| | - Noam Eliaz
- Department of Materials Science and Engineering, Tel-Aviv University, Tel Aviv 6997801, Israel;
| | - Daniel Mandler
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel; (G.N.); (N.B.-S.); (S.B.)
| |
Collapse
|
20
|
Cell–scaffold interactions in tissue engineering for oral and craniofacial reconstruction. Bioact Mater 2023; 23:16-44. [DOI: 10.1016/j.bioactmat.2022.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/22/2022] [Accepted: 10/30/2022] [Indexed: 11/09/2022] Open
|
21
|
Serafin A, Culebras M, Collins MN. Synthesis and evaluation of alginate, gelatin, and hyaluronic acid hybrid hydrogels for tissue engineering applications. Int J Biol Macromol 2023; 233:123438. [PMID: 36709805 DOI: 10.1016/j.ijbiomac.2023.123438] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/16/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
Tissue engineering (TE) has been proposed extensively as a potential solution to the worldwide shortages of donor organs needed for transplantation. Over the years, numerous hydrogel formulations have been studied for various TE endeavours, including bone, cardiac or neural TE treatment strategies. Amongst the materials used, organic and biocompatible materials which aim to mimic the natural extracellular matrix of the native tissue have been investigated to create biomimicry regenerative environments. As such, the comparison between studies using the same materials is often difficult to accomplish due to varying material concentrations, preparation strategies, and laboratory settings, and as such these variables have a huge impact on the physio-chemical properties of the hydrogel systems. The purpose of the current study is to investigate popular biomaterials such as alginate, hyaluronic acid and gelatin in a variety of concentrations and hydrogel formulations. This aims to provide a clear and comprehensive understanding of their behaviours and provide a rational approach as to the appropriate selection of natural polysaccharides in specific targeted TE strategies.
Collapse
Affiliation(s)
- Aleksandra Serafin
- School of Engineering, Bernal Institute, University of Limerick, Limerick, Ireland
| | - Mario Culebras
- Materials Science Institute (ICMUV), Universitat de València, c/ Catedrático José Beltrán 2, 46980 Paterna, Valencia, Spain
| | - Maurice N Collins
- School of Engineering, Bernal Institute, University of Limerick, Limerick, Ireland; Health Research Institute and AMBER University of Limerick, Limerick V94 T9PX, Ireland.
| |
Collapse
|
22
|
Sarker S, Colton A, Wen Z, Xu X, Erdi M, Jones A, Kofinas P, Tubaldi E, Walczak P, Janowski M, Liang Y, Sochol RD. 3D-Printed Microinjection Needle Arrays via a Hybrid DLP-Direct Laser Writing Strategy. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201641. [PMID: 37064271 PMCID: PMC10104452 DOI: 10.1002/admt.202201641] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Indexed: 06/19/2023]
Abstract
Microinjection protocols are ubiquitous throughout biomedical fields, with hollow microneedle arrays (MNAs) offering distinctive benefits in both research and clinical settings. Unfortunately, manufacturing-associated barriers remain a critical impediment to emerging applications that demand high-density arrays of hollow, high-aspect-ratio microneedles. To address such challenges, here, a hybrid additive manufacturing approach that combines digital light processing (DLP) 3D printing with "ex situ direct laser writing (esDLW)" is presented to enable new classes of MNAs for fluidic microinjections. Experimental results for esDLW-based 3D printing of arrays of high-aspect-ratio microneedles-with 30 μm inner diameters, 50 μm outer diameters, and 550 μm heights, and arrayed with 100 μm needle-to-needle spacing-directly onto DLP-printed capillaries reveal uncompromised fluidic integrity at the MNA-capillary interface during microfluidic cyclic burst-pressure testing for input pressures in excess of 250 kPa (n = 100 cycles). Ex vivo experiments perform using excised mouse brains reveal that the MNAs not only physically withstand penetration into and retraction from brain tissue but also yield effective and distributed microinjection of surrogate fluids and nanoparticle suspensions directly into the brains. In combination, the results suggest that the presented strategy for fabricating high-aspect-ratio, high-density, hollow MNAs could hold unique promise for biomedical microinjection applications.
Collapse
Affiliation(s)
- Sunandita Sarker
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA; Maryland Robotics Center, University of Maryland, College Park, MD 20742, USA; Institute for Systems Research, University of Maryland, College Park, MD 20742, USA
| | - Adira Colton
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA; Maryland Robotics Center, University of Maryland, College Park, MD 20742, USA; Institute for Systems Research, University of Maryland, College Park, MD 20742, USA
| | - Ziteng Wen
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA
| | - Xin Xu
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA
| | - Metecan Erdi
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA
| | - Anthony Jones
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA; Maryland Robotics Center, University of Maryland, College Park, MD 20742, USA; Institute for Systems Research, University of Maryland, College Park, MD 20742, USA
| | - Peter Kofinas
- Department of Chemical and Biomolecular Engineering, University of Maryland, College Park, MD 20742, USA
| | - Eleonora Tubaldi
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA; Maryland Robotics Center, University of Maryland, College Park, MD 20742, USA; Institute for Systems Research, University of Maryland, College Park, MD 20742, USA
| | - Piotr Walczak
- Program in Image Guided Neurointerventions, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Miroslaw Janowski
- Program in Image Guided Neurointerventions, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Yajie Liang
- Program in Image Guided Neurointerventions, Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ryan D Sochol
- Department of Mechanical Engineering, University of Maryland, College Park, MD 20742, USA; Maryland Robotics Center, University of Maryland, College Park, MD 20742, USA; Institute for Systems Research, University of Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA; Robert E. Fischell Institute for Biomedical Devices, University of Maryland, College Park, MD 20742, USA
| |
Collapse
|
23
|
Kerch G. Severe COVID-19-A Review of Suggested Mechanisms Based on the Role of Extracellular Matrix Stiffness. Int J Mol Sci 2023; 24:1187. [PMID: 36674700 PMCID: PMC9861790 DOI: 10.3390/ijms24021187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
The severity of COVID-19 commonly depends on age-related tissue stiffness. The aim was to review publications that explain the effect of microenvironmental extracellular matrix stiffness on cellular processes. Platelets and endothelial cells are mechanosensitive. Increased tissue stiffness can trigger cytokine storm with the upregulated expression of pro-inflammatory cytokines, such as tumor necrosis factor alpha and interleukin IL-6, and tissue integrity disruption, leading to enhanced virus entry and disease severity. Increased tissue stiffness in critically ill COVID-19 patients triggers platelet activation and initiates plague formation and thrombosis development. Cholesterol content in cell membrane increases with aging and further enhances tissue stiffness. Membrane cholesterol depletion decreases virus entry to host cells. Membrane cholesterol lowering drugs, such as statins or novel chitosan derivatives, have to be further developed for application in COVID-19 treatment. Statins are also known to decrease arterial stiffness mitigating cardiovascular diseases. Sulfated chitosan derivatives can be further developed for potential use in future as anticoagulants in prevention of severe COVID-19. Anti-TNF-α therapies as well as destiffening therapies have been suggested to combat severe COVID-19. The inhibition of the nuclear factor kappa-light-chain-enhancer of activated B cells pathway must be considered as a therapeutic target in the treatment of severe COVID-19 patients. The activation of mechanosensitive platelets by higher matrix stiffness increases their adhesion and the risk of thrombus formation, thus enhancing the severity of COVID-19.
Collapse
Affiliation(s)
- Garry Kerch
- Faculty of Materials Science and Applied Chemistry, Riga Technical University, 1048 Riga, Latvia
| |
Collapse
|
24
|
Katz RR, West JL. Tunable PEG Hydrogels for Discerning Differential Tumor Cell Response to Biomechanical Cues. Adv Biol (Weinh) 2022; 6:e2200084. [PMID: 35996804 DOI: 10.1002/adbi.202200084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 08/01/2022] [Indexed: 01/28/2023]
Abstract
Increased extracellular matrix (ECM) density in the tumor microenvironment has been shown to influence aspects of tumor progression such as proliferation and invasion. Increased matrix density means cells experience not only increased mechanical properties, but also a higher density of bioactive sites. Traditional in vitro ECM models like Matrigel and collagen do not allow these properties to be investigated independently. In this work, a poly(ethylene glycol)-based scaffold is used which modifies with integrin-binding sites for cell attachment and matrix metalloproteinase 2 and 9 sensitive sites for enzyme-mediated degradation. The polymer backbone density and binding site concentration are independently tuned and the effect each of these properties and their interaction have on the proliferation, invasion, and focal complex formation of two different tumor cell lines is evaluated. It is seen that the cell line of epithelial origin (Hs 578T, triple negative breast cancer) proliferates more, invades less, and forms more mature focal complexes in response to an increase in matrix adhesion sites. Conversely, the cell line of mesenchymal origin (HT1080, fibrosarcoma) proliferates more in 2D culture but less in 3D culture, invades less, and forms more mature focal complexes in response to an increase in matrix stiffness.
Collapse
Affiliation(s)
- Rachel R Katz
- Department of Biomedical Engineering, Duke University, Fitzpatrick Center (FCIEMAS), Room 1427, 101 Science Drive, Campus Box 90281, Durham, NC, 27708-0281, USA
| | - Jennifer L West
- Department of Biomedical Engineering, Duke University, Fitzpatrick Center (FCIEMAS), Room 1427, 101 Science Drive, Campus Box 90281, Durham, NC, 27708-0281, USA.,Department of Biomedical Engineering, University of Virginia, 351 McCormick Rd, Charlottesville, VA, 22904, USA
| |
Collapse
|
25
|
Di X, Jin X, Ai J, Xiang L, Gao X, Xiao K, Li H, Luo D, Wang K. YAP/Smad3 promotes pathological extracellular matrix microenviroment-induced bladder smooth muscle proliferation in bladder fibrosis progression. MedComm (Beijing) 2022; 3:e169. [PMID: 36176734 PMCID: PMC9477793 DOI: 10.1002/mco2.169] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/17/2022] [Accepted: 07/22/2022] [Indexed: 11/06/2022] Open
Abstract
Fibrosis is a chronic inflammation process with excess extracellular matrix (ECM) deposition that cannot be reversed. Patients suffer from bladder dysfunction caused by bladder fibrosis. Moreover, the interactive mechanisms between ECM and bladder fibrosis are still obscure. Hence, we assessed the pivotal effect of Yes-associated protein (YAP) on the proliferation of bladder smooth muscle in fibrosis process. We identified that stiff ECM increased the expression and translocation of YAP in the nucleus of human bladder smooth muscle cell (hBdSMC). Sequencings and proteomics revealed that YAP bound to Smad3 and promoted the proliferation of hBdSMC via MAPK/ERK signaling pathway in stiff ECM. Moreover, CUT and TAG sequencing and dual-luciferase assays demonstrated that Smad3 inhibited the transcription of JUN. The YAP inhibitor CA3 was used in a partial bladder outlet obstruction (pBOO) rat model. The results showed that CA3 attenuated bladder smooth muscle proliferation. Collectively, YAP binding with Smad3 in the nucleus inhibited the transcription of JUN, and promoted the proliferation of bladder smooth muscle through the MAPK/ERK signaling pathway. The current study identified a novel mechanism of mechanical force induced bladder fibrosis that provided insights in YAP-associated organ fibrosis.
Collapse
Affiliation(s)
- Xing‐Peng Di
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology)West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xi Jin
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology)West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Jian‐Zhong Ai
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology)West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Li‐Yuan Xiang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology)West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Xiao‐Shuai Gao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology)West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Kai‐Wen Xiao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology)West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Hong Li
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology)West China Hospital, Sichuan UniversityChengduSichuanChina
| | - De‐Yi Luo
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology)West China Hospital, Sichuan UniversityChengduSichuanChina
| | - Kun‐Jie Wang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology)West China Hospital, Sichuan UniversityChengduSichuanChina
| |
Collapse
|
26
|
Diamantides N, Slyker L, Martin S, Rodriguez MR, Bonassar LJ. Pre-glycation impairs gelation of high concentration collagen solutions. J Biomed Mater Res A 2022; 110:1953-1963. [PMID: 36183358 PMCID: PMC9648490 DOI: 10.1002/jbm.a.37431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 06/30/2022] [Accepted: 07/21/2022] [Indexed: 09/29/2023]
Abstract
There remains a need for stiffer collagen hydrogels for tissue engineering and disease modeling applications. Pre-glycation, or glycation of collagen in solution prior to gelation, has been shown to increase the mechanics of collagen hydrogels while maintaining high viability of encapsulated cells. The stiffness of glycated collagen gels can be increased by increasing the collagen concentration, sugar concentration, and glycation time. However, previous studies on pre-glycation of collagen have used low collagen concentrations and/or low sugar concentrations and have not investigated the effect of glycation time. Therefore, the objective of this study was to determine the effects of pre-glycation with high sugar concentrations (up to 500 mM) and extended glycation times (up to 21 days) on high concentration collagen (8 mg/ml). The addition of sugar to the collagen and the formation of advanced glycation end products (AGEs) were quantified. The ability to gel successfully and rheological properties were determined and correlated with biochemical characterizations. Successful collagen gelation and rheological properties of pre-glycated collagen were found to be strongly dependent on the ratio of added sugars to added AGEs with high ratios impairing gelation and low ratios resulting in optimal storage moduli. There is likely a competing effect during pre-glycation of the formation of AGEs resulting in crosslinking of collagen and the formation of Amadori intermediates acting to increase collagen solubility. Overall, this study shows that collagen glycation can be optimized by increasing the formation of AGEs while maintaining a low ratio of added sugar to added AGEs.
Collapse
Affiliation(s)
| | - Leigh Slyker
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Sara Martin
- Department of Materials Science and Engineering, Cornell University, Ithaca, NY
| | | | - Lawrence J. Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY
| |
Collapse
|
27
|
Effects of sodium hypochlorite and ethylenediaminetetraacetic acid on proliferation, osteogenic/odontogenic differentiation, and mechanosensitive gene expression of human dental pulp stem cells. Tissue Cell 2022; 79:101955. [DOI: 10.1016/j.tice.2022.101955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 10/06/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022]
|
28
|
Designer injectable matrices of photocrosslinkable carboxymethyl cellulose methacrylate based hydrogels as cell carriers for gel type autologous chondrocyte implantation (GACI). Int J Biol Macromol 2022; 224:465-482. [DOI: 10.1016/j.ijbiomac.2022.10.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 10/10/2022] [Accepted: 10/15/2022] [Indexed: 11/05/2022]
|
29
|
Oliverio R, Patenaude V, Liberelle B, Virgilio N, Banquy X, De Crescenzo G. Macroporous dextran hydrogels for controlled growth factor capture and delivery using coiled-coil interactions. Acta Biomater 2022; 153:190-203. [PMID: 36113720 DOI: 10.1016/j.actbio.2022.09.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/24/2022] [Accepted: 09/08/2022] [Indexed: 11/01/2022]
Abstract
Macroporous hydrogels possess a vast potential for various applications in the biomedical field. However, due to their large pore size allowing for unrestricted diffusion in the macropore network, macroporous hydrogels alone are not able to efficiently capture and release biomolecules in a controlled manner. There is thus a need for biofunctionalized, affinity-based gels that can efficiently load and release biomolecules in a sustained and controlled manner. For this purpose, we report here the use of a E/K coiled-coil affinity pair for the controlled capture and delivery of growth factors from highly interconnected, macroporous dextran hydrogels. By conjugating the Kcoil peptide to the dextran backbone, we achieved controlled loading and release of Ecoil-tagged Epidermal and Vascular Endothelial Growth Factors. To finely tune the behavior of the gels, we propose four control parameters: (i) macropore size, (ii) Kcoil grafting density, (iii) Ecoil valency and (iv) E/K affinity. We demonstrate that Kcoil grafting can produce a 20-fold increase in passive growth factor capture by macroporous dextran gels. Furthermore, we demonstrate that our gels can release as little as 20% of the loaded growth factors over one week, while retaining bioactivity. Altogether, we propose a versatile, highly tunable platform for the controlled delivery of growth factors in biomedical applications. STATEMENT OF SIGNIFICANCE: This work presents a highly tunable platform for growth factor capture and sustained delivery using affinity peptides in macroporous, fully interconnected dextran hydrogels. It addresses several ongoing challenges by presenting: (i) a versatile platform for the delivery of a wide range of stable, bioactive molecules, (ii) a passive, affinity-based loading of growth factors in the platform, paving the way for in situ (re)loading of the device and (iii) four different control parameters to finely tune growth factor capture and release. Altogether, our macroporous dextran hydrogels have a vast potential for applications in controlled delivery, tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Romane Oliverio
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Québec H3T 1J4, Canada; Faculty of Pharmacy, Axe Formulation et Analyse du Médicament (AFAM), Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Victor Patenaude
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Québec H3T 1J4, Canada
| | - Benoît Liberelle
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Québec H3T 1J4, Canada
| | - Nick Virgilio
- Department of Chemical Engineering, Centre de Recherche sur les Systèmes Polymères et Composites à Haute Performance (CREPEC), Polytechnique Montréal, Montréal, Québec H3T 1J4, Canada
| | - Xavier Banquy
- Faculty of Pharmacy, Axe Formulation et Analyse du Médicament (AFAM), Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | - Gregory De Crescenzo
- Department of Chemical Engineering, Polytechnique Montréal, Montréal, Québec H3T 1J4, Canada.
| |
Collapse
|
30
|
Jubelin C, Muñoz-Garcia J, Griscom L, Cochonneau D, Ollivier E, Heymann MF, Vallette FM, Oliver L, Heymann D. Three-dimensional in vitro culture models in oncology research. Cell Biosci 2022; 12:155. [PMID: 36089610 PMCID: PMC9465969 DOI: 10.1186/s13578-022-00887-3] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 08/18/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractCancer is a multifactorial disease that is responsible for 10 million deaths per year. The intra- and inter-heterogeneity of malignant tumors make it difficult to develop single targeted approaches. Similarly, their diversity requires various models to investigate the mechanisms involved in cancer initiation, progression, drug resistance and recurrence. Of the in vitro cell-based models, monolayer adherent (also known as 2D culture) cell cultures have been used for the longest time. However, it appears that they are often less appropriate than the three-dimensional (3D) cell culture approach for mimicking the biological behavior of tumor cells, in particular the mechanisms leading to therapeutic escape and drug resistance. Multicellular tumor spheroids are widely used to study cancers in 3D, and can be generated by a multiplicity of techniques, such as liquid-based and scaffold-based 3D cultures, microfluidics and bioprinting. Organoids are more complex 3D models than multicellular tumor spheroids because they are generated from stem cells isolated from patients and are considered as powerful tools to reproduce the disease development in vitro. The present review provides an overview of the various 3D culture models that have been set up to study cancer development and drug response. The advantages of 3D models compared to 2D cell cultures, the limitations, and the fields of application of these models and their techniques of production are also discussed.
Collapse
|
31
|
Su T, Xu M, Lu F, Chang Q. Adipogenesis or osteogenesis: destiny decision made by mechanical properties of biomaterials. RSC Adv 2022; 12:24501-24510. [PMID: 36128379 PMCID: PMC9425444 DOI: 10.1039/d2ra02841g] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022] Open
Abstract
Regenerative medicine affords an effective approach for restoring defect-associated diseases, and biomaterials play a pivotal role as cell niches to support the cell behavior and decide the destiny of cell differentiation. Except for chemical inducers, mechanical properties such as stiffness, pore size and topography of biomaterials play a crucial role in the regulation of cell behaviors and functions. Stiffness may determine the adipogenesis or osteogenesis of mesenchymal stem cells (MSCs) via the translocation of yes-associated protein (YAP) and the transcriptional coactivator with a PDZ-binding motif (TAZ). External forces transmit through cytoskeleton reorientation to assist nuclear deformation and molecule transport, meanwhile, signal pathways including the Hippo, FAK/RhoA/ROCK, and Wnt/β-catenin have been evidenced to participate in the mechanotransduction. Different pore sizes not only tailor the scaffold stiffness but also conform to the requirements of cell migration and vessels in-growth. Topography guides cell geometry along with mobility and determines the cell fate ascribed to micro/nano-scale contact. Herein, we highlight the recent progress in exploring the regulation mechanism by the physical properties of biomaterials, which might lead to more innovative regenerative strategies for adipose or bone tissue repair.
Collapse
Affiliation(s)
- Ting Su
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University 510515 China
| | - Mimi Xu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University 510515 China
| | - Feng Lu
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University 510515 China
| | - Qiang Chang
- Department of Plastic and Cosmetic Surgery, Nanfang Hospital, Southern Medical University 510515 China
| |
Collapse
|
32
|
Wu D, Hou Y, Chu Z, Wei Q, Hong W, Lin Y. Ligand Mobility-Mediated Cell Adhesion and Spreading. ACS APPLIED MATERIALS & INTERFACES 2022; 14:12976-12983. [PMID: 35282676 DOI: 10.1021/acsami.1c22603] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Cells live in a highly dynamic environment where their physical connection and communication with the outside are achieved through receptor-ligands binding. Therefore, a precise knowledge of the interaction between receptors and ligands is critical for our understanding of how cells execute different biological duties. Interestingly, recent evidence has shown that the mobility of ligands at the cell-extracellular matrix (ECM) interface significantly affects the adhesion and spreading of cells, while the underlying mechanism remains unclear. Here, we present a modeling investigation to address this critical issue. Specifically, by adopting the Langevin dynamics, the random movement of ligands was captured by assigning a stochastic force along with a viscous drag on them. After that, the evolution of adhesion and subsequent spreading of cells were analyzed by considering the force-regulated binding/breakage of individual molecular bonds connecting polymerizing actin bundles inside the cell to the ECM. Interestingly, a biphasic relationship between adhesion and ligand diffusivity was predicted, resulting in maximized cell spreading at intermediate mobility of ligand molecules. In addition, this peak position was found to be dictated by the aggregation of ligands, effectively reducing their diffusivity, and how fast bond association/dissociation can occur. These predictions are in excellent agreement with our experimental observations where distinct ligand mobility was introduced by tuning the interactions between the self-assembly polymer coating and the surface.
Collapse
Affiliation(s)
- Di Wu
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong 000000, China
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yong Hou
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong 000000, China
| | - Zhiqin Chu
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Hong Kong 000000, China
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu 610065, China
| | - Wei Hong
- Department of Mechanics and Aerospace Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Yuan Lin
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong 000000, China
- HKU-Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, Guangdong 518057, China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong 000000, China
| |
Collapse
|
33
|
Luo T, Tan B, Zhu L, Wang Y, Liao J. A Review on the Design of Hydrogels With Different Stiffness and Their Effects on Tissue Repair. Front Bioeng Biotechnol 2022; 10:817391. [PMID: 35145958 PMCID: PMC8822157 DOI: 10.3389/fbioe.2022.817391] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/07/2022] [Indexed: 12/20/2022] Open
Abstract
Tissue repair after trauma and infection has always been a difficult problem in regenerative medicine. Hydrogels have become one of the most important scaffolds for tissue engineering due to their biocompatibility, biodegradability and water solubility. Especially, the stiffness of hydrogels is a key factor, which influence the morphology of mesenchymal stem cells (MSCs) and their differentiation. The researches on this point are meaningful to the field of tissue engineering. Herein, this review focus on the design of hydrogels with different stiffness and their effects on the behavior of MSCs. In addition, the effect of hydrogel stiffness on the phenotype of macrophages is introduced, and then the relationship between the phenotype changes of macrophages on inflammatory response and tissue repair is discussed. Finally, the future application of hydrogels with a certain stiffness in regenerative medicine and tissue engineering has been prospected.
Collapse
Affiliation(s)
- Tianyi Luo
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Bowen Tan
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lengjing Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yating Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jinfeng Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Centre for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Jinfeng Liao,
| |
Collapse
|
34
|
Wang Q, Xu W, Koppolu R, van Bochove B, Seppälä J, Hupa L, Willför S, Xu C, Wang X. Injectable thiol-ene hydrogel of galactoglucomannan and cellulose nanocrystals in delivery of therapeutic inorganic ions with embedded bioactive glass nanoparticles. Carbohydr Polym 2022; 276:118780. [PMID: 34823793 DOI: 10.1016/j.carbpol.2021.118780] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/24/2021] [Accepted: 10/13/2021] [Indexed: 01/31/2023]
Abstract
We propose an injectable nanocomposite hydrogel that is photo-curable via light-induced thiol-ene addition between methacrylate modified O-acetyl-galactoglucomannan (GGMMA) and thiolated cellulose nanocrystal (CNC-SH). Compared to free-radical chain polymerization, the orthogonal step-growth of thiol-ene addition allows a less heterogeneous hydrogel network and more rapid crosslinking kinetics. CNC-SH reinforced the GGMMA hydrogel as both a nanofiller and a crosslinker to GGMMA resulting in an interpenetrating network via thiol-ene addition. Importantly, the mechanical stiffness of the GGMMA/CNC-SH hydrogel is mainly determined by the stoichiometric ratio between the thiol groups on CNC-SH and the methacrylate groups in GGMMA. Meanwhile, the bioactive glass nanoparticle (BaGNP)-laden hydrogels of GGMMA/CNC-SH showed a sustained release of therapeutic ions in simulated body fluid in vitro, which extended the bioactive function of hydrogel matrix. Furthermore, the suitability of the GGMMA/CNC-SH formulation as biomaterial resin to fabricate digitally designed hydrogel constructs via digital light processing (DLP) lithography printing was evaluated.
Collapse
Affiliation(s)
- Qingbo Wang
- Laboratory of Natural Materials Technology, Åbo Akademi University, Henrikinkatu 2, Turku FI-20500, Finland
| | - Wenyang Xu
- Laboratory of Natural Materials Technology, Åbo Akademi University, Henrikinkatu 2, Turku FI-20500, Finland
| | - Rajesh Koppolu
- Laboratory of Natural Materials Technology, Åbo Akademi University, Henrikinkatu 2, Turku FI-20500, Finland
| | - Bas van Bochove
- Polymer Technology, School of Chemical Engineering, Aalto University, Kemistintie 1D, Espoo FI-02150, Finland
| | - Jukka Seppälä
- Polymer Technology, School of Chemical Engineering, Aalto University, Kemistintie 1D, Espoo FI-02150, Finland
| | - Leena Hupa
- Laboratory of Molecular Science and Technology, Åbo Akademi University, Henrikinkatu 2, Turku FI-20500, Finland
| | - Stefan Willför
- Laboratory of Natural Materials Technology, Åbo Akademi University, Henrikinkatu 2, Turku FI-20500, Finland
| | - Chunlin Xu
- Laboratory of Natural Materials Technology, Åbo Akademi University, Henrikinkatu 2, Turku FI-20500, Finland
| | - Xiaoju Wang
- Laboratory of Natural Materials Technology, Åbo Akademi University, Henrikinkatu 2, Turku FI-20500, Finland; Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Tykistökatu 6A, Turku FI-20520, Finland.
| |
Collapse
|
35
|
Narita T, Kondo M, Oishi Y. Macroscopic Banding Pattern of Collagen Gel Formed by a Diffusion-Reaction Process. ACS OMEGA 2022; 7:1014-1020. [PMID: 35036765 PMCID: PMC8756805 DOI: 10.1021/acsomega.1c05601] [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: 10/07/2021] [Accepted: 11/30/2021] [Indexed: 06/14/2023]
Abstract
Shapes and patterns observed in internal organs and tissues are reproducibly and robustly produced over a long distance (up to millimeters in length). The most fundamental remaining question is how these long geometries of shape and pattern form arise from the genetic message. Recent studies have demonstrated that extracellular matrix (ECM) critically participates as a structural foundation on which cells can organize and communicate. ECMs may be a key to understanding the underlying mechanisms of long-distance patterning and morphogenesis. However, previous studies in this field mainly focused on the complexes and interaction of cells and ECM. This paper pays particular attention to ECM and demonstrates that collagen, a major ECM component, natively possesses the reproducible and definite patterning ability reaching centimeter-scale length. The macroscopic pattern consists of striped transparent layers. The observation under crossed Nicols demonstrates that the layers consist of alternately arranged polarized and unpolarized parts. Confocal fluorescence microscopy studies revealed that the polarized and unpolarized segments include collagen-rich and -poor regions, respectively. The patterning process was proposed based on the Liesegang banding formation, which are mineral precipitation bands formed in hydrogel matrixes. These findings will give hints to the questions about long-distance cell alignment and provide new clues to artificially control cell placement over micron size in the field of regenerative medicine.
Collapse
|
36
|
The Role of Substrate Topography and Stiffness on MSC Cells Functions: Key Material Properties for Biomimetic Bone Tissue Engineering. Biomimetics (Basel) 2021; 7:biomimetics7010007. [PMID: 35076475 PMCID: PMC8788532 DOI: 10.3390/biomimetics7010007] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 11/16/2022] Open
Abstract
The hypothesis of the present research is that by altering the substrate topography and/or stiffness to make it biomimetic, we can modulate cells behavior. Substrates with similar surface chemistry and varying stiffnesses and topographies were prepared. Bulk PCL and CNTs-reinforced PCL composites were manufactured by solvent casting method and electrospinning and further processed to obtain tunable moduli of elasticity in the range of few MPa. To ensure the same chemical profile for the substrates, a protein coating was added. Substrate topography and properties were investigated. Further on, the feedback of Wharton’s Jelly Umbilical Cord Mesenchymal Stem Cells to substrates characteristics was investigated. Solvent casting scaffolds displayed superior mechanical properties compared to the corresponding electrospun films. However, the biomimetic fibrous texture of the electrospun substrates induced improved feedback of the cells with respect to their viability and proliferation. Cells’ adhesion and differentiation was remarkably pronounced on solvent casting substrates compared to the electrospun substrates. Soft substates improved cells multiplication and migration, while stiff substrates induced differentiation into bone cells. Aspects related to the key factors and the ideal properties of substrates and microenvironments were clarified, aiming towards the deep understanding of the required optimum biomimetic features of biomaterials.
Collapse
|
37
|
Abstract
Organoids-cellular aggregates derived from stem or progenitor cells that recapitulate organ function in miniature-are of growing interest in developmental biology and medicine. Organoids have been developed for organs and tissues such as the liver, gut, brain, and pancreas; they are used as organ surrogates to study a wide range of questions in basic and developmental biology, genetic disorders, and therapies. However, many organoids reported to date have been cultured in Matrigel, which is prepared from the secretion of Engelbreth-Holm-Swarm mouse sarcoma cells; Matrigel is complex and poorly defined. This complexity makes it difficult to elucidate Matrigel-specific factors governing organoid development. In this review, we discuss promising Matrigel-free methods for the generation and maintenance of organoids that use decellularized extracellular matrix (ECM), synthetic hydrogels, or gel-forming recombinant proteins.
Collapse
Affiliation(s)
- Mark T Kozlowski
- DEVCOM US Army Research Laboratory, Weapons and Materials Research Directorate, Science of Extreme Materials Division, Polymers Branch, 6300 Rodman Rd. Building 4600, Aberdeen Proving Ground, Aberdeen, MD, 21005, USA.
| | - Christiana J Crook
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Hsun Teresa Ku
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
- Irell and Manella Graduate School of Biological Sciences, Beckman Research Institute of City of Hope, 1500 Duarte Rd., Duarte, CA, 91010, USA
| |
Collapse
|
38
|
Sharifi S, Sharifi H, Akbari A, Chodosh J. Systematic optimization of visible light-induced crosslinking conditions of gelatin methacryloyl (GelMA). Sci Rep 2021; 11:23276. [PMID: 34857867 PMCID: PMC8640009 DOI: 10.1038/s41598-021-02830-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 11/18/2021] [Indexed: 11/09/2022] Open
Abstract
Gelatin methacryloyl (GelMA) is one of the most widely used photo-crosslinkable biopolymers in tissue engineering. In in presence of an appropriate photoinitiator, the light activation triggers the crosslinking process, which provides shape fidelity and stability at physiological temperature. Although ultraviolet (UV) has been extensively explored for photo-crosslinking, its application has been linked to numerous biosafety concerns, originated from UV phototoxicity. Eosin Y, in combination with TEOA and VC, is a biosafe photoinitiation system that can be activated via visible light instead of UV and bypasses those biosafety concerns; however, the crosslinking system needs fine-tuning and optimization. In order to systematically optimize the crosslinking conditions, we herein independently varied the concentrations of Eosin Y [(EY)], triethanolamine (TEOA), vinyl caprolactam (VC), GelMA precursor, and crosslinking times and assessed the effect of those parameters on the properties the hydrogel. Our data showed that except EY, which exhibited an optimal concentration (~ 0.05 mM), increasing [TEOA], [VA], [GelMA], or crosslinking time improved mechanical (tensile strength/modulus and compressive modulus), adhesion (lap shear strength), swelling, biodegradation properties of the hydrogel. However, increasing the concentrations of crosslinking reagents ([TEOA], [VA], [GelMA]) reduced cell viability in 3-dimensional (3D) cell culture. This study enabled us to optimize the crosslinking conditions to improve the properties of the GelMA hydrogel and to generate a library of hydrogels with defined properties essential for different biomedical applications.
Collapse
Affiliation(s)
- Sina Sharifi
- Disruptive Technology Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, MA, USA.
| | - Hannah Sharifi
- Disruptive Technology Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, MA, USA
| | - Ali Akbari
- Solid Tumor Research Center, Research Institute for Cellular and Molecular Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - James Chodosh
- Disruptive Technology Laboratory, Department of Ophthalmology, Harvard Medical School, Massachusetts Eye and Ear and Schepens Eye Research Institute, Boston, MA, USA
| |
Collapse
|
39
|
Modulation of stem cell response using biodegradable polyester films with different stiffness. BIOMEDICAL ENGINEERING ADVANCES 2021. [DOI: 10.1016/j.bea.2021.100007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
40
|
Goswami R, Arya RK, Sharma S, Dutta B, Stamov DR, Zhu X, Rahaman SO. Mechanosensing by TRPV4 mediates stiffness-induced foreign body response and giant cell formation. Sci Signal 2021; 14:eabd4077. [PMID: 34726952 PMCID: PMC9976933 DOI: 10.1126/scisignal.abd4077] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Implantation of biomaterials or devices into soft tissue often leads to the development of the foreign body response (FBR), an inflammatory condition that can cause implant failure, tissue injury, and death of the patient. Macrophages accumulate and fuse to generate destructive foreign body giant cells (FBGCs) at the tissue-implant interface, leading to the development of fibrous scar tissue around the implant that is generated by myofibroblasts. We previously showed that the FBR in vivo and FBGC formation in vitro require transient receptor potential vanilloid 4 (TRPV4), a mechanosensitive ion channel. Here, we report that TRPV4 was required specifically for the FBR induced by implant stiffness independently of biochemical cues and for intracellular stiffening that promotes FBGC formation in vitro. TRPV4 deficiency reduced collagen deposition and the accumulation of macrophages, FBGCs, and myofibroblasts at stiff, but not soft, implants in vivo and inhibited macrophage-induced differentiation of wild-type fibroblasts into myofibroblasts in vitro. Atomic force microscopy demonstrated that TRPV4 was required for implant-adjacent tissue stiffening in vivo and for cytoskeletal remodeling and intracellular stiffening induced by fusogenic cytokines in vitro. Together, these data suggest a mechanism whereby a reciprocal functional interaction between TRPV4 and substrate stiffness leads to cytoskeletal remodeling and cellular force generation to promote FBGC formation during the FBR.
Collapse
Affiliation(s)
- Rishov Goswami
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Rakesh K. Arya
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Shweta Sharma
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Bidisha Dutta
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - Dimitar R. Stamov
- JPK BioAFM Business, Nano Surfaces Division, Bruker Nano GmbH, Am Studio 2D, 12489 Berlin, Germany
| | - Xiaoping Zhu
- Department of Veterinary Medicine, University of Maryland, College Park, MD 20742, USA
| | - Shaik O. Rahaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA.,Corresponding author.:
| |
Collapse
|
41
|
Stoycheva D, Simsek H, Weber W, Hauser AE, Klotzsch E. External cues to drive B cell function towards immunotherapy. Acta Biomater 2021; 133:222-230. [PMID: 33636402 DOI: 10.1016/j.actbio.2021.02.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/31/2022]
Abstract
Immunotherapy stands out as a powerful and promising therapeutic strategy in the treatment of cancer, infections, and autoimmune diseases. Adoptive immune therapies are usually centered on modified T cells and their specific expansion towards antigen-specific T cells against cancer and other diseases. However, despite their unmatched features, the potential of B cells in immunotherapy is just beginning to be explored. The main role of B cells in the immune response is to secrete antigen-specific antibodies and provide long-term protection against foreign pathogens. They further function as antigen-presenting cells (APCs) and secrete pro- and anti-inflammatory cytokines and thus exert positive and negative regulatory stimuli on other cells involved in the immune response such as T cells. Therefore, while hyperactivation of B cells can cause autoimmunity, their dysfunctions lead to severe immunodeficiencies. Only suitably activated B cells can play an active role in the treatment of cancers, infections, and autoimmune diseases. As a result, studies have focused on B cell-targeted immunotherapies in recent years. For this, the development, functions, interactions with the microenvironment, and clinical importance of B cells should be well understood. In this review, we summarize the main events during B cell activation. From the viewpoint of mechanobiology we discuss the translation of external cues such as surface topology, substrate stiffness, and biochemical signaling into B cell functions. We further dive into current B cell-targeted therapy strategies and their clinical applications. STATEMENT OF SIGNIFICANCE: B cells are proving as a promising tool in the field of immunotherapy. B cells exhibit various functions such as antibody production, antigen presentation or secretion of immune-regulatory factors which can be utilized in the fight against oncological or immunological disorders. In this review we discuss the importance of external mechanobiological cues such as surface topology, substrate stiffness, and biochemical signaling on B cell function. We further summarize B cell-targeted therapy strategies and their clinical applications, as in the context of anti-tumor responses and autoimmune diseases.
Collapse
|
42
|
Pasqua M, Di Gesù R, Chinnici CM, Conaldi PG, Francipane MG. Generation of Hepatobiliary Cell Lineages from Human Induced Pluripotent Stem Cells: Applications in Disease Modeling and Drug Screening. Int J Mol Sci 2021; 22:8227. [PMID: 34360991 PMCID: PMC8348238 DOI: 10.3390/ijms22158227] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/13/2022] Open
Abstract
The possibility to reproduce key tissue functions in vitro from induced pluripotent stem cells (iPSCs) is offering an incredible opportunity to gain better insight into biological mechanisms underlying development and disease, and a tool for the rapid screening of drug candidates. This review attempts to summarize recent strategies for specification of iPSCs towards hepatobiliary lineages -hepatocytes and cholangiocytes-and their use as platforms for disease modeling and drug testing. The application of different tissue-engineering methods to promote accurate and reliable readouts is discussed. Space is given to open questions, including to what extent these novel systems can be informative. Potential pathways for improvement are finally suggested.
Collapse
Affiliation(s)
- Mattia Pasqua
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
| | - Roberto Di Gesù
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
| | - Cinzia Maria Chinnici
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
- Dipartimento della Ricerca, IRCCS ISMETT, 90127 Palermo, Italy;
| | | | - Maria Giovanna Francipane
- Fondazione Ri.MED, 90133 Palermo, Italy; (M.P.); (R.D.G.); (C.M.C.)
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
43
|
Velasco-Mallorquí F, Rodríguez-Comas J, Ramón-Azcón J. Cellulose-based scaffolds enhance pseudoislets formation and functionality. Biofabrication 2021; 13. [PMID: 34075893 DOI: 10.1088/1758-5090/ac00c3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/13/2021] [Indexed: 12/17/2022]
Abstract
In vitroresearch for the study of type 2 diabetes (T2D) is frequently limited by the availability of a functional model for islets of Langerhans. To overcome the limitations of obtaining pancreatic islets from different sources, such as animal models or human donors, immortalized cell lines as the insulin-producing INS1Eβ-cells have appeared as a valid alternative to model insulin-related diseases. However, immortalized cell lines are mainly used in flat surfaces or monolayer distributions, not resembling the spheroid-like architecture of the pancreatic islets. To generate islet-like structures, the use of scaffolds appeared as a valid tool to promote cell aggregations. Traditionally-used hydrogel encapsulation methods do not accomplish all the requisites for pancreatic tissue engineering, as its poor nutrient and oxygen diffusion induces cell death. Here, we use cryogelation technology to develop a more resemblance scaffold with the mechanical and physical properties needed to engineer pancreatic tissue. This study shows that carboxymethyl cellulose (CMC) cryogels prompted cells to generateβ-cell clusters in comparison to gelatin-based scaffolds, that did not induce this cell organization. Moreover, the high porosity achieved with CMC cryogels allowed us to create specific range pseudoislets. Pseudoislets formed within CMC-scaffolds showed cell viability for up to 7 d and a better response to glucose over conventional monolayer cultures. Overall, our results demonstrate that CMC-scaffolds can be used to control the organization and function of insulin-producingβ-cells, representing a suitable technique to generateβ-cell clusters to study pancreatic islet function.
Collapse
Affiliation(s)
- Ferran Velasco-Mallorquí
- Biosensors for Bioengineering, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, Barcelona 08028, Spain
| | - Júlia Rodríguez-Comas
- Biosensors for Bioengineering, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, Barcelona 08028, Spain
| | - Javier Ramón-Azcón
- Biosensors for Bioengineering, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, Barcelona 08028, Spain.,ICREA-Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| |
Collapse
|
44
|
Lei R, Akins EA, Wong KCY, Repina NA, Wolf KJ, Dempsey GE, Schaffer DV, Stahl A, Kumar S. Multiwell Combinatorial Hydrogel Array for High-Throughput Analysis of Cell-ECM Interactions. ACS Biomater Sci Eng 2021; 7:2453-2465. [PMID: 34028263 DOI: 10.1021/acsbiomaterials.1c00065] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Biophysical cues in the extracellular matrix (ECM) regulate cell behavior in a complex, nonlinear, and interdependent manner. To quantify these important regulatory relationships and gain a comprehensive understanding of mechanotransduction, there is a need for high-throughput matrix platforms that enable parallel culture and analysis of cells in various matrix conditions. Here we describe a multiwell hyaluronic acid (HA) platform in which cells are cultured on combinatorial arrays of hydrogels spanning a range of elasticities and adhesivities. Our strategy utilizes orthogonal photopatterning of stiffness and adhesivity gradients, with the stiffness gradient implemented by a programmable light illumination system. The resulting platform allows individual treatment and analysis of each matrix environment while eliminating contributions of haptotaxis and durotaxis. In human mesenchymal stem cells, our platform recapitulates expected relationships between matrix stiffness, adhesivity, and cell mechanosensing. We further applied the platform to show that as integrin ligand density falls, cell adhesion and migration depend more strongly on CD44-mediated interactions with the HA backbone. We anticipate that our system could bear great value for mechanistic discovery and screening where matrix mechanics and adhesivity are expected to influence phenotype.
Collapse
Affiliation(s)
- Ruoxing Lei
- Department of Chemistry, Latimer Hall, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Erin A Akins
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States.,University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States
| | - Kelly C Y Wong
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Nicole A Repina
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States.,University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States
| | - Kayla J Wolf
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States.,University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States
| | - Garrett E Dempsey
- Department of Nutritional Sciences and Toxicology, Morgan Hall, University of California, Berkeley, California 94720, United States
| | - David V Schaffer
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States.,University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States.,Department of Molecular and Cell Biology, Life Sciences Addition, University of California, Berkeley, California 94720, United States.,Department of Chemical and Biomolecular Engineering, Gilman Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Andreas Stahl
- University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States.,Department of Nutritional Sciences and Toxicology, Morgan Hall, University of California, Berkeley, California 94720, United States
| | - Sanjay Kumar
- Department of Bioengineering, Stanley Hall, University of California, Berkeley, Berkeley, California 94720, United States.,University of California, Berkeley - University of California, San Francisco Graduate Program in Bioengineering, Stanley Hall, Berkeley, California 94720, United States.,Department of Chemical and Biomolecular Engineering, Gilman Hall, University of California, Berkeley, Berkeley, California 94720, United States.,Department of Bioengineering and Therapeutic Sciences, Byers Hall, University of California, San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
45
|
Alshehri S, Susapto HH, Hauser CAE. Scaffolds from Self-Assembling Tetrapeptides Support 3D Spreading, Osteogenic Differentiation, and Angiogenesis of Mesenchymal Stem Cells. Biomacromolecules 2021; 22:2094-2106. [PMID: 33908763 PMCID: PMC8382244 DOI: 10.1021/acs.biomac.1c00205] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/15/2021] [Indexed: 01/01/2023]
Abstract
The apparent rise of bone disorders demands advanced treatment protocols involving tissue engineering. Here, we describe self-assembling tetrapeptide scaffolds for the growth and osteogenic differentiation of human mesenchymal stem cells (hMSCs). The rationally designed peptides are synthetic amphiphilic self-assembling peptides composed of four amino acids that are nontoxic. These tetrapeptides can quickly solidify to nanofibrous hydrogels that resemble the extracellular matrix and provide a three-dimensional (3D) environment for cells with suitable mechanical properties. Furthermore, we can easily tune the stiffness of these peptide hydrogels by just increasing the peptide concentration, thus providing a wide range of peptide hydrogels with different stiffnesses for 3D cell culture applications. Since successful bone regeneration requires both osteogenesis and vascularization, our scaffold was found to be able to promote angiogenesis of human umbilical vein endothelial cells (HUVECs) in vitro. The results presented suggest that ultrashort peptide hydrogels are promising candidates for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Salwa Alshehri
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Hepi H. Susapto
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Charlotte A. E. Hauser
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
46
|
Garnica-Galvez S, Korntner SH, Skoufos I, Tzora A, Diakakis N, Prassinos N, Zeugolis DI. Hyaluronic Acid as Macromolecular Crowder in Equine Adipose-Derived Stem Cell Cultures. Cells 2021; 10:859. [PMID: 33918830 PMCID: PMC8070604 DOI: 10.3390/cells10040859] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 01/10/2023] Open
Abstract
The use of macromolecular crowding in the development of extracellular matrix-rich cell-assembled tissue equivalents is continuously gaining pace in regenerative engineering. Despite the significant advancements in the field, the optimal macromolecular crowder still remains elusive. Herein, the physicochemical properties of different concentrations of different molecular weights hyaluronic acid (HA) and their influence on equine adipose-derived stem cell cultures were assessed. Within the different concentrations and molecular weight HAs, the 10 mg/mL 100 kDa and 500 kDa HAs exhibited the highest negative charge and hydrodynamic radius, and the 10 mg/mL 100 kDa HA exhibited the lowest polydispersity index and the highest % fraction volume occupancy. Although HA had the potential to act as a macromolecular crowding agent, it did not outperform carrageenan and Ficoll®, the most widely used macromolecular crowding molecules, in enhanced and accelerated collagen I, collagen III and collagen IV deposition.
Collapse
Affiliation(s)
- Sergio Garnica-Galvez
- Laboratory of Animal Science, Nutrition and Biotechnology, Department of Agriculture, University of Ioannina, 47100 Arta, Greece; (S.G.-G.); (I.S.); (A.T.)
- School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (N.D.); (N.P.)
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), H92 W2TY Galway, Ireland;
| | - Stefanie H. Korntner
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), H92 W2TY Galway, Ireland;
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), H92 W2TY Galway, Ireland
| | - Ioannis Skoufos
- Laboratory of Animal Science, Nutrition and Biotechnology, Department of Agriculture, University of Ioannina, 47100 Arta, Greece; (S.G.-G.); (I.S.); (A.T.)
| | - Athina Tzora
- Laboratory of Animal Science, Nutrition and Biotechnology, Department of Agriculture, University of Ioannina, 47100 Arta, Greece; (S.G.-G.); (I.S.); (A.T.)
| | - Nikolaos Diakakis
- School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (N.D.); (N.P.)
| | - Nikitas Prassinos
- School of Veterinary Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (N.D.); (N.P.)
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), H92 W2TY Galway, Ireland;
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), H92 W2TY Galway, Ireland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Faculty of Biomedical Sciences, Università della Svizzera Italiana (USI), 6904 Lugano, Switzerland
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), School of Mechanical and Materials Engineering, University College Dublin (UCD), D04 V1W8 Dublin, Ireland
| |
Collapse
|
47
|
Wang C, Sinha S, Jiang X, Murphy L, Fitch S, Wilson C, Grant G, Yang F. Matrix Stiffness Modulates Patient-Derived Glioblastoma Cell Fates in Three-Dimensional Hydrogels. Tissue Eng Part A 2021; 27:390-401. [PMID: 32731804 PMCID: PMC7984937 DOI: 10.1089/ten.tea.2020.0110] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/17/2020] [Indexed: 01/13/2023] Open
Abstract
Cancer progression is known to be accompanied by changes in tissue stiffness. Previous studies have primarily employed immortalized cell lines and 2D hydrogel substrates, which do not recapitulate the 3D tumor niche. How matrix stiffness affects patient-derived cancer cell fate in 3D remains unclear. In this study, we report a matrix metalloproteinase-degradable poly(ethylene-glycol)-based hydrogel platform with brain-mimicking biochemical cues and tunable stiffness (40-26,600 Pa) for 3D culture of patient-derived glioblastoma xenograft (PDTX GBM) cells. Our results demonstrate that decreasing hydrogel stiffness enhanced PDTX GBM cell proliferation, and hydrogels with stiffness 240 Pa and below supported robust PDTX GBM cell spreading in 3D. PDTX GBM cells encapsulated in hydrogels demonstrated higher drug resistance than 2D control, and increasing hydrogel stiffness further enhanced drug resistance. Such 3D hydrogel platforms may provide a valuable tool for mechanistic studies of the role of niche cues in modulating cancer progression for different cancer types.
Collapse
Affiliation(s)
- Christine Wang
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, California, USA
| | - Sauradeep Sinha
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, California, USA
| | - Xinyi Jiang
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Luke Murphy
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, California, USA
| | - Sergio Fitch
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| | - Christy Wilson
- Department of Neurosurgery, Stanford University, School of Medicine, Stanford, California, USA
| | - Gerald Grant
- Department of Neurosurgery, Stanford University, School of Medicine, Stanford, California, USA
| | - Fan Yang
- Department of Bioengineering, Schools of Engineering and Medicine, Stanford University, Stanford, California, USA
- Department of Orthopaedic Surgery, Stanford University, Stanford, California, USA
| |
Collapse
|
48
|
Lei K, Kurum A, Tang L. Mechanical Immunoengineering of T cells for Therapeutic Applications. Acc Chem Res 2020; 53:2777-2790. [PMID: 33258577 DOI: 10.1021/acs.accounts.0c00486] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
T cells, a key component in adaptive immunity, are central to many immunotherapeutic modalities aimed at treating various diseases including cancer, infectious diseases, and autoimmune disorders. The past decade has witnessed tremendous progress in immunotherapy, which aims at activation or suppression of the immune responses for disease treatments. Most strikingly, cancer immunotherapy has led to curative responses in a fraction of patients with relapsed or refractory cancers. However, extending those clinical benefits to a majority of cancer patients remains challenging. In order to improve both efficacy and safety of T cell-based immunotherapies, significant effort has been devoted to modulating biochemical signals to enhance T cell proliferation, effector functions, and longevity. Such strategies include discovery of new immune checkpoints, design of armored chimeric antigen receptor (CAR) T cells, and targeted delivery of stimulatory cytokines and so on.Despite the intense global research effort in developing novel cancer immunotherapies, a major dimension of the interactions between cancer and the immune system, its biomechanical aspect, has been largely underappreciated. Throughout their lifecycle, T cells constantly survey a multitude of organs and tissues and experience diverse biomechanical environments, such as shear force in the blood flow and a broad range of tissue stiffness. Furthermore, biomechanical properties of tissues or cells may be altered in disease and inflammation. Biomechanical cues, including both passive mechanical cues and active mechanical forces, have been shown to govern T cell development, activation, migration, differentiation, and effector functions. In other words, T cells can sense, respond to, and adapt to both passive mechanical cues and active mechanical forces.Biomechanical cues have been intensively studied at a fundamental level but are yet to be extensively incorporated in the design of immunotherapies. Nonetheless, the growing knowledge of T cell mechanobiology has formed the basis for the development of novel engineering strategies to mechanically modulate T cell immunity, a nascent field that we termed "mechanical immunoengineering". Mechanical immunoengineering exploits biomechanical cues (e.g., stiffness and external forces) to modulate T cell differentiation, proliferation, effector functions, etc., for diagnostic or therapeutic applications. It provides an additional dimension, complementary to traditional modulation of biochemical cues (e.g., antigen density and co-stimulatory signals), to tailor T cell immune responses and enhance therapeutic outcomes. For example, stiff antigen-presenting matrices have been shown to enhance T cell proliferation independently of the intensity of biochemical stimulatory signals. Current strategies of mechanical immunoengineering of T cells can be categorized into two major fields including passive mechanical cue-oriented and active force-oriented strategies. In this Account, we first present a brief overview of T cell mechanobiology. Next, we summarize recent advances in mechanical immunoengineering, discuss the roles of chemistry and material science in the development of these engineering strategies, and highlight potential therapeutic applications. Finally, we present our perspective on the future directions in mechanical immunoengineering and critical steps to translate mechanical immunoengineering strategies into therapeutic applications in the clinic.
Collapse
|
49
|
Gaukås NH, Huynh QS, Pratap AA, Einarsrud MA, Grande T, Holsinger RMD, Glaum J. In Vitro Biocompatibility of Piezoelectric K0.5Na0.5NbO3 Thin Films on Platinized Silicon Substrates. ACS APPLIED BIO MATERIALS 2020; 3:8714-8721. [DOI: 10.1021/acsabm.0c01111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Nikolai Helth Gaukås
- Department of Materials Science and Engineering, NTNU Norwegian University of Science and Technology, Sem Sælands vei 12, Trondheim, Norway
- Laboratory of Molecular Neuroscience and Dementia, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett St., Camperdown, NSW 2050, Australia
| | - Quy-Susan Huynh
- Laboratory of Molecular Neuroscience and Dementia, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett St., Camperdown, NSW 2050, Australia
- Discipline of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Anishchal A. Pratap
- Laboratory of Molecular Neuroscience and Dementia, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett St., Camperdown, NSW 2050, Australia
- Discipline of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Mari-Ann Einarsrud
- Department of Materials Science and Engineering, NTNU Norwegian University of Science and Technology, Sem Sælands vei 12, Trondheim, Norway
| | - Tor Grande
- Department of Materials Science and Engineering, NTNU Norwegian University of Science and Technology, Sem Sælands vei 12, Trondheim, Norway
| | - R. M. Damian Holsinger
- Laboratory of Molecular Neuroscience and Dementia, Brain and Mind Centre, Faculty of Medicine and Health, The University of Sydney, 94 Mallett St., Camperdown, NSW 2050, Australia
- Discipline of Pathology, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Julia Glaum
- Department of Materials Science and Engineering, NTNU Norwegian University of Science and Technology, Sem Sælands vei 12, Trondheim, Norway
| |
Collapse
|
50
|
Ma J, Cai H, Long X, Cheng K, Xu X, Zhang D, Li J. Hyaluronic acid bioinspired polymers for the regulation of cell chondrogenic and osteogenic differentiation. Int J Biol Macromol 2020; 161:1011-1020. [PMID: 32531368 DOI: 10.1016/j.ijbiomac.2020.06.064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 06/01/2020] [Accepted: 06/08/2020] [Indexed: 12/28/2022]
Abstract
As the simplest glycosaminoglycan (GAG) in extracellular matrix, hyaluronic acid (HA) takes part in several important biological processes, such as regulating cell proliferation, differentiation, and migration. In this work, a series of HA-inspired polymers with different saccharide and carboxylate units (HA-analogue polymers) are synthesized by free radical polymerization, and characterized using Fourier transform infrared spectroscopy (FT-IR), gel permeation chromatography (GPC) and nuclear magnetic resonance spectrometer (NMR), Moreover, cell experiments demonstrate that HA-analogue polymers with a certain proportion of saccharide and carboxylate (PM1G1) units shows a positive effect on the proliferation and differentiation of bone marrow mesenchymal stem cells (BMSCs). Furthermore, HA-analogue polymers have prominent cartilage inductive capacity in chondrogenic induction medium (CIM) and brilliant bone inductive capacity in osteogenic induction medium (OIM) toward BMSCs. Therefore, it is confirmed that the HA-analogue polymers can effectively mimic the functions of HA and have broad potential application prospects in the biomedical and clinical fields.
Collapse
Affiliation(s)
- Jiayun Ma
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Huijuan Cai
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Xiaoling Long
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Kai Cheng
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Xinyuan Xu
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Dongyue Zhang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Jianshu Li
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China; State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|