1
|
Sonaye SY, Sikder P. Bioengineered Constructs as a Tissue Engineering-Based Therapy for Volumetric Muscle Loss. TISSUE ENGINEERING. PART B, REVIEWS 2025. [PMID: 40265282 DOI: 10.1089/ten.teb.2025.0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Severe skeletal muscle injuries involving substantial tissue loss can significantly impair muscle strength and functionality, reducing the quality of life for affected individuals. Such injuries, termed volumetric muscle loss, require extensive clinical intervention, as the body's innate healing mechanisms are insufficient to regenerate functional muscle. The current standard of care primarily involves autologous muscle tissue transfer, with some consideration of acellular synthetic constructs. However, both approaches have limited therapeutic efficacy, presenting challenges such as donor-site morbidity, infection risks, and suboptimal functional recovery. Over the past decade, skeletal muscle tissue engineering (SMTE) has emerged as a promising strategy for regenerating functional muscle through bioengineered constructs. Advanced biofabrication techniques, including bioprinting, have further enabled the development of synthetic constructs that closely mimic native muscle architecture. Given these advancements, a critical review of recent therapeutic strategies, their achievements, and limitations is necessary. This review examines the spectrum of bioengineered constructs developed from various biomaterials and evaluates their therapeutic potential. Special emphasis is placed on 3D bioprinting strategies and their role in creating physiologically relevant constructs for functional muscle restoration. In addition, the integration of machine learning in optimizing construct design, predicting cellular behavior, and enhancing tissue integration is discussed. The review indicates that despite significant progress in SMTE, key challenges remain, including replicating the complex structural organization of muscle tissue, minimizing fibrosis, and achieving vascularization and innervation to regenerate functional, strengthened muscle. Future research should address these barriers while prioritizing the development of translational, clinically relevant regenerative constructs. In addition, efforts should focus on advancing scalable, construct-based regenerative treatments that are readily available at the point of care and easily managed in surgical settings.
Collapse
Affiliation(s)
| | - Prabaha Sikder
- Department of Mechanical Engineering, Cleveland State University, Cleveland, Ohio, USA
| |
Collapse
|
2
|
Mobin A, Hashem Z, Corridon PR. Transforming meat waste into sustainable corneal keratoplasty models. Front Bioeng Biotechnol 2025; 13:1572127. [PMID: 40297283 PMCID: PMC12034655 DOI: 10.3389/fbioe.2025.1572127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
With a rapidly global population, there is a critical need to enhance food production and waste management. This necessity is driving opportunities for sustainable integrated food chains committed to biovalorization and circular bioeconomic practices. One approach that aligns with this vision relies on sustainable tissue engineering, which offers opportunities to leverage food systems in the search for natural biomaterials from agricultural waste. In this perspective, we propose utilizing common meat waste sources, often associated with a high environmental footprint, to develop tissue graft models. These models reduce agricultural waste, decrease the reliance on animal testing, and support both biovalorization and medical innovation. Specifically, we explore a unique approach to generate corneal transplantation models completely from discarded components of the meat food chain, using the eyes and bladders. This strategy involves creating keratoplasty models by reseeding the decellularized extracellular matrix (dECM), encompassing three major corneal regions: the epithelium, stroma, and endothelium. Interestingly, these scaffolds can be recellularized with cellular lineages derived from stem niches harvested from urine. This approach integrates waste management with regenerative medicine, fostering sustainable advancements in tissue engineering.
Collapse
Affiliation(s)
- Ayman Mobin
- Department of Biomedical Engineering and Biotechnology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Zayd Hashem
- Department of Biomedical Engineering and Biotechnology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Peter R. Corridon
- Department of Biomedical Engineering and Biotechnology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biomedical Engineering and Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
- Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
3
|
Dell AC, Maresca J, Davis BA, Isaji T, Dardik A, Geibel JP. Development and deployment of a functional 3D-bioprinted blood vessel. Sci Rep 2025; 15:11668. [PMID: 40188212 PMCID: PMC11972400 DOI: 10.1038/s41598-025-93276-y] [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: 11/11/2024] [Accepted: 03/05/2025] [Indexed: 04/07/2025] Open
Abstract
Creating vascular structures in vitro via bioprinting to replace damaged or missing vasculature has significant advantages over current surgical methods of vessel replacement. Using rat fibroblasts and smooth muscle cells, we have bioprinted a rat aorta using a rotating mandrel method to create the tubular replacement structure. Then, the 3D-bioprinted aortas were implanted into rats to determine their functionality long-term in vivo. The implanted vascular conduits were well-tolerated, well-incorporated into native vasculature, and showed the physiological behavior of a native vessel. The development and deployment of 3D-bioprinted vessels for repair of large vessels in an animal model paves the way for advancements in the treatment of vascular disease in humans.
Collapse
Affiliation(s)
- Annika C Dell
- The John B. Pierce Laboratory, Inc, New Haven, CT, 06519, USA.
- Fraunhofer IMTE, Fraunhofer Research Institution for Individualized and Cell-Based Medical Engineering, 23562, Lübeck, Germany.
| | - Jamie Maresca
- The John B. Pierce Laboratory, Inc, New Haven, CT, 06519, USA
| | - Bruce A Davis
- The John B. Pierce Laboratory, Inc, New Haven, CT, 06519, USA
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, CT, 06510, USA
| | - Toshihiko Isaji
- Division of Vascular Surgery, Department of Surgery, Yale School of Medicine, New Haven, Connecticut, CT, 06510, USA
| | - Alan Dardik
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, CT, 06510, USA
| | - John P Geibel
- The John B. Pierce Laboratory, Inc, New Haven, CT, 06519, USA.
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut, CT, 06510, USA.
| |
Collapse
|
4
|
Kopinski-Grünwald O, Schandl S, Gusev J, Chamalaki OE, Ovsianikov A. Surface functionalization of microscaffolds produced by high-resolution 3D printing: A new layer of freedom. Mater Today Bio 2025; 31:101452. [PMID: 39896295 PMCID: PMC11783114 DOI: 10.1016/j.mtbio.2025.101452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/25/2024] [Accepted: 01/03/2025] [Indexed: 02/04/2025] Open
Abstract
Scaffolded-spheroids represent novel building blocks for bottom-up tissue assembly, allowing to produce constructs with high initial cell density. Previously, we demonstrated the successful differentiation of such building blocks, produced from immortalized human adipose-derived stem cells, towards different phenotypes, and the possibility of creating macro-sized tissue-like constructs in vitro. The culture of cells in vitro depends on the supply of various nutrients and biomolecules, such as growth factors, usually supplemented in the culture medium. Another means for growth factor delivery (in vitro and in vivo) is the release from the scaffold to alter the biological response of surrounding cells (e.g. by release of VEGF).1 As a proof of concept for this approach, we sought to biofunctionalize the surface of the microscaffolds with heparin as a "universal linker" that would allow binding a variety of growth factors/biomolecules. An aminolysis step in an organic solvent made it possible to generate a hydrophilic and charged surface. The backbone of the amine, as well as reaction conditions, led to an adjustable surface modification. The amount of heparin on the surface was increased with an ethylene glycol-based diamine backbone and varied between 8 and 40 ng per microscaffold. Choosing a suitable linker allows easy adjustment of the loading of VEGF and other heparin-binding proteins. Initial results indicated that up to 5 ng VEGF could be loaded per microscaffold, generating a steady VEGF release for 16 days. We report an easy-to-perform, scalable surface modification approach of polyester-based resin that leads to adjustable surface concentrations of heparin. The successful surface aminolysis opens the route to various modifications and broadens the spectrum of biomolecules which can be delivered.
Collapse
Affiliation(s)
| | | | - Jegor Gusev
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien (Technische Universität Wien), Getreidemarkt 9/308, 1060, Vienna, Austria
| | - Ourania Evangelia Chamalaki
- Research Group 3D Printing and Biofabrication, Institute of Materials Science and Technology, TU Wien (Technische Universität Wien), Getreidemarkt 9/308, 1060, Vienna, Austria
| | | |
Collapse
|
5
|
Rasekh M, Arshad MS, Ahmad Z. Advances in Drug Delivery Integrated with Regenerative Medicine: Innovations, Challenges, and Future Frontiers. Pharmaceutics 2025; 17:456. [PMID: 40284451 PMCID: PMC12030587 DOI: 10.3390/pharmaceutics17040456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 03/19/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
Advances in drug delivery systems adapted with regenerative medicine have transformed healthcare by introducing innovative strategies to treat (and repair in many instances) disease-impacted regions of the human body. This review provides a comprehensive analysis of the latest developments and challenges in integrating drug delivery technologies with regenerative medicine. Recent advances in drug delivery technologies, including the design of biomaterials, localized delivery techniques, and controlled release systems guided by mathematical models, are explored to illustrate their role in enhancing therapeutic precision and efficacy. Additionally, regenerative medicine approaches are analyzed, with a focus on extracellular matrix components, stem cell-based therapies, and emerging strategies for organ regeneration in both soft and hard tissue and in vitro model engineering. In particular, the review also discusses the applications of cellular components, including stem cells, immune cells, endothelial cells, and specialized cells such as chondrocytes and osteoblasts, and highlights advancements in cell delivery methods and cell-cell interaction modulation. In addition, future directions and pivotal trends emphasizing the importance of interdisciplinary collaboration and cutting-edge innovations are provided to address successful therapeutic outcomes in regenerative medicine.
Collapse
Affiliation(s)
- Manoochehr Rasekh
- College of Engineering, Design and Physical Sciences, Brunel University of London, Uxbridge UB8 3PH, UK
| | | | - Zeeshan Ahmad
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, UK
| |
Collapse
|
6
|
Sleem B, Nassereldine R, Ghazi V, Eid K, Hemdanieh M, Nassereddine M. From Bone To Blood Flow: Tissue Engineering In Orthopedics - A Narrative Review. Orthop Rev (Pavia) 2025; 17:132223. [PMID: 40176925 PMCID: PMC11964394 DOI: 10.52965/001c.132223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 02/04/2025] [Indexed: 04/05/2025] Open
Abstract
Musculoskeletal injuries and degenerative conditions necessitate advanced regenerative solutions. Tissue engineering has emerged as a pivotal field in orthopedic care, particularly in vascularized bone and cartilage regeneration. This narrative review examines the latest advancements in vascular tissue engineering, including scaffold design, cell-based techniques, and growth factor delivery. A comprehensive literature search was conducted using PubMed, ScienceDirect, and Google Scholar, focusing on innovations and challenges in the field. Vascularized bone grafts (VBGs) outperform non-vascularized counterparts in promoting healing and integration. Advances in scaffold materials, such as smart scaffolds and hybrid biomaterials, enhance osteogenesis and angiogenesis. Cellular therapies, utilizing mesenchymal stem cells and induced pluripotent stem cells, synergistically improve vascularization and bone regeneration. Growth factors like VEGF and bone morphogenic protein (BMP-2), integrated with innovative delivery systems, enable sustained angiogenic stimulation and scaffold integration. While significant strides have been made, challenges persist in achieving full vascular integration and replicating native tissue architecture. Innovations in scaffold technology and vascular surgery techniques hold promise for transforming orthopedic tissue engineering and improving patient outcomes.
Collapse
Affiliation(s)
- Bshara Sleem
- Faculty of MedicineAmerican University of Beirut Medical Center
| | - Rakan Nassereldine
- Department of Vascular SurgeryAmerican University of Beirut Medical Center
| | - Victor Ghazi
- Faculty of MedicineAmerican University of Beirut Medical Center
| | - Karine Eid
- Faculty of MedicineAmerican University of Beirut Medical Center
| | - Maya Hemdanieh
- Division of Orthopedic SurgeryAmerican University of Beirut Medical Center
| | | |
Collapse
|
7
|
Winkler P, Mao Y. Dual Delivery of Cells and Bioactive Molecules for Wound Healing Applications. Molecules 2025; 30:1577. [PMID: 40286165 PMCID: PMC11990229 DOI: 10.3390/molecules30071577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/21/2025] [Accepted: 03/30/2025] [Indexed: 04/29/2025] Open
Abstract
Chronic wounds not only cause significant patient morbidity but also impose a substantial economic burden on the healthcare system. The primary barriers to wound healing include a deficiency of key modulatory factors needed to progress beyond the stalled inflammatory phase and an increased susceptibility to infections. While antimicrobial agents have traditionally been used to treat infections, stem cells have recently emerged as a promising therapy due to their regenerative properties, including the secretion of cytokines and immunomodulators that support wound healing. This study aims to develop an advanced dual-delivery system integrating stem cells and antibiotics. Stem cells have previously been delivered by encapsulation in gelatin methacrylate (GelMA) hydrogels. To explore a more effective delivery method, GelMA was processed into microparticles (MP). Compared to a bulk GelMA hydrogel (HG) encapsulation, GelMA MP supported greater cell growth and enhanced in vitro wound healing activity of human mesenchymal stem cells (hMSCs), likely due to a larger surface area for cell attachment and improved nutrient exchange. To incorporate antimicrobial properties, the broad-spectrum antibiotics penicillin/streptomycin (PS) were loaded into a bulk GelMA hydrogel, which was then cryo-milled into MPs to serve as carriers for hMSCs. To achieve a more sustained antibiotic release, gelatin nanoparticles (NP) were used as carriers for PS. PS was either incorporated during NP synthesis (NP+PS(S)) or absorbed into NP after synthesis (NP+PS(A)). MPs containing PS, NP+PS(S), or NP+PS(A) were tested for their cell carrier functions and antibacterial activities. The incorporation of PS did not compromise the cell-carrying function of MP configurations. The anti-S. aureus activity was detected in conditioned media from MPs for up to eight days-four days longer than from bulk HG containing PS. Notably, the presence of hMSCs prolonged the antimicrobial activity of MPs, suggesting a synergistic effect between stem cells and antibiotics. PS loaded via synthesis (NP+PS(S)) exhibited a delayed initial release, whereas PS loaded via absorption (NP+PS(A)) provided a more immediate release, with potential for sustained delivery. This study demonstrates the feasibility of a dual-delivery system integrating thera.
Collapse
Affiliation(s)
| | - Yong Mao
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA;
| |
Collapse
|
8
|
Lahouty M, Soleymanzadeh A, Kazemi S, Saadati-Maleki H, Masoudi S, Ghasemi A, Kazemi T, Mehranfar S, Fadaee M. Cell-based immunotherapy in oesophageal cancer. J Drug Target 2025:1-11. [PMID: 40063049 DOI: 10.1080/1061186x.2025.2477077] [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: 01/21/2025] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/18/2025]
Abstract
Oesophageal cancer (EC) is among the most common illnesses globally, and its prognosis is unfavourable. Surgery, radiotherapy and chemotherapy are the primary therapy options for EC. Despite the occasional efficacy of these traditional treatment modalities, individuals with EC remain at a significant risk for local recurrence and metastasis. Consequently, innovative and efficacious medicines are required. In recent decades, clinical practice has effectively implemented cell therapy, which includes both stem cell and non-stem cell-based approaches, as an innovative tumour treatment, offering renewed hope to patients with oesophageal squamous cell carcinoma (ESCC). This paper examines the theoretical framework and contemporary advancements in cell treatment for individuals with EC. We further described current clinical studies and summarised essential data related to survival and safety assessments.
Collapse
Affiliation(s)
- Masoud Lahouty
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Sama Kazemi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Haniyeh Saadati-Maleki
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Sanaz Masoudi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arash Ghasemi
- Faculty of Medicine, Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Mehranfar
- Department of Genetics and Immunology, Urmia University of Medical Sciences, Urmia, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
9
|
Samiei M, Harmsen MC, Abdolahinia ED, Barar J, Petridis X. Scaffold-Free Strategies in Dental Pulp/Dentine Tissue Engineering: Current Status and Implications for Regenerative Biological Processes. Bioengineering (Basel) 2025; 12:198. [PMID: 40001717 PMCID: PMC11851408 DOI: 10.3390/bioengineering12020198] [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: 01/31/2025] [Revised: 02/16/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Conventionally, root canal treatment is performed when the dental pulp is severely damaged or lost due to dental trauma or bacterial endodontic infections. This treatment involves removing the compromised or infected pulp tissue, disinfecting the root canal system, and sealing it with inert, non-degradable materials. However, contemporary endodontic treatment has shifted from merely obturating the root canal system with inert materials to guiding endodontic tissue regeneration through biological approaches. The ultimate goal of regenerative endodontics is to restore dental pulp tissue with structural organization and functional characteristics akin to the native pulp, leveraging advancements in tissue engineering and biomaterial sciences. Dental pulp tissue engineering commonly employs scaffold-based strategies, utilizing biomaterials as initial platforms for cell and growth factor delivery, which subsequently act as scaffolds for cell proliferation, differentiation and maturation. However, cells possess an intrinsic capacity for self-organization into spheroids and can generate their own extracellular matrix, eliminating the need for external scaffolds. This self-assembling property presents a promising alternative for scaffold-free dental pulp engineering, addressing limitations associated with biomaterial-based approaches. This review provides a comprehensive overview of cell-based, self-assembling and scaffold-free approaches in dental pulp tissue engineering, highlighting their potential advantages and challenges in advancing regenerative endodontics.
Collapse
Affiliation(s)
- Mohammad Samiei
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.S.); (M.C.H.)
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran
| | - Martin Conrad Harmsen
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (M.S.); (M.C.H.)
| | - Elaheh Dalir Abdolahinia
- Department of Oral Science and Translation Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33314, USA;
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| | - Xenos Petridis
- Department of Endodontics, Section of Dental Pathology & Therapeutics, School of Dentistry, National and Kapodistrian University of Athens, 115 27 Athens, Greece
- Department of Endodontology, Section of Fundamental Dentistry, Center for Dentistry and Oral Hygiene, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands
| |
Collapse
|
10
|
Salgado García MG, Díaz NF, García López G, Álvarez Maya I, Hernández Jimenez C, Roman Maldonado Y, Mendoza Aguayo DJ, Díaz Martínez NE. Evaluation methods for decellularized tissues: A focus on human amniotic membrane. J Biosci Bioeng 2025; 139:85-94. [PMID: 39592317 DOI: 10.1016/j.jbiosc.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024]
Abstract
Tissue engineering, a multidisciplinary research field aiming to revolutionize regenerative medicine, relies on scaffolds for optimal cell cultures and organ development. Decellularized tissue extracellular matrices (dECM) scaffolds, particularly from human amniotic membrane (hAM), show promise in clinical applications. This review discusses the significance of scaffolds, emphasizing dECM-based hAM scaffolds, delving into ECM complexities, decellularization processes, and evaluation methods. Raman spectroscopy emerges as a non-destructive tool for evaluating ECM preservation, presenting potential for quantifying ECM components in hAM before and after decellularization. The review explores the role of hAM as a biomaterial, detailing its composition and characteristics and emphasizes the importance of evaluating ultrastructural components and suggests Raman spectroscopy as a valuable technique for this purpose.
Collapse
Affiliation(s)
- Miriam Guadalupe Salgado García
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico; Instituto Tecnológico y de Estudios Superiores de Occidente (ITESO), Tlaquepaque, Mexico
| | | | | | - Ikuri Álvarez Maya
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico
| | | | - Yvonne Roman Maldonado
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico; Instituto Tecnológico y de Estudios Superiores de Occidente (ITESO), Tlaquepaque, Mexico
| | | | - Néstor Emmanuel Díaz Martínez
- Laboratorio de Reprogramación Celular y Bioingeniería de Tejidos, Biotecnología Médica y Farmacéutica, Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco (CIATEJ), Guadalajara, Mexico.
| |
Collapse
|
11
|
Sousa AC, Alvites R, Lopes B, Sousa P, Moreira A, Coelho A, Santos JD, Atayde L, Alves N, Maurício AC. Three-Dimensional Printing/Bioprinting and Cellular Therapies for Regenerative Medicine: Current Advances. J Funct Biomater 2025; 16:28. [PMID: 39852584 PMCID: PMC11765675 DOI: 10.3390/jfb16010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 01/26/2025] Open
Abstract
The application of three-dimensional (3D) printing/bioprinting technologies and cell therapies has garnered significant attention due to their potential in the field of regenerative medicine. This paper aims to provide a comprehensive overview of 3D printing/bioprinting technology and cell therapies, highlighting their results in diverse medical applications, while also discussing the capabilities and limitations of their combined use. The synergistic combination of 3D printing and cellular therapies has been recognised as a promising and innovative approach, and it is expected that these technologies will progressively assume a crucial role in the treatment of various diseases and conditions in the foreseeable future. This review concludes with a forward-looking perspective on the future impact of these technologies, highlighting their potential to revolutionize regenerative medicine through enhanced tissue repair and organ replacement strategies.
Collapse
Affiliation(s)
- Ana Catarina Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Rui Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
- Instituto Universitário de Ciências da Saúde (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, Gandra, 4585-116 Paredes, Portugal
| | - Bruna Lopes
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Patrícia Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Alícia Moreira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - André Coelho
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - José Domingos Santos
- REQUIMTE-LAQV, Departamento de Engenharia Metalúrgica e Materiais, Faculdade de Engenharia, UP, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal;
| | - Luís Atayde
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| | - Nuno Alves
- Centre for Rapid and Sustainable Product Development (CDRSP), Polytechnic Institute of Leiria, Rua de Portugal—Zona Industrial, 2430-028 Marinha Grande, Portugal;
| | - Ana Colette Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, nº 228, 4050-313 Porto, Portugal; (A.C.S.); (R.A.); (B.L.); (P.S.); (A.M.); (A.C.); (L.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Av. Universidade Técnica, 1300-477 Lisboa, Portugal
| |
Collapse
|
12
|
Gurdal M, Zeugolis DI. Macromolecular crowding agent dependent extracellular matrix deposition and growth factor retention in human corneal fibroblast cultures. Exp Eye Res 2025; 250:110162. [PMID: 39571777 DOI: 10.1016/j.exer.2024.110162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/04/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
The major obstacle in the commercialisation and clinical translation of tissue engineered medicines is the required for the development of implantable tissue surrogates prolonged in vitro culture. Macromolecular crowding (MMC) enhances and accelerates extracellular matrix (ECM) deposition, thus offering an opportunity to bridge the gap between research and development in tissue engineered substitutes. However, the optimal MMC agent is still elusive. Herein, we first assessed the biophysical properties of the most widely used MMC agents [κλ carrageenan (κλ CR), λ carrageenan (λ CR) and Ficoll™ cocktail (FC)] and then assessed their effect in basic cell function, ECM deposition and growth factor retention in human corneal fibroblast (hCF) cultures. Dynamic light scattering analysis revealed that both CR macromolecules had significantly lower and higher zeta potential and hydrodynamic radius, respectively, than the FC. None of the MMC agents affected hCF morphology and all induced similar hCF viability, proliferation and metabolic activity. Electrophoresis and immunofluorescence analyses made apparent that at day 10 (longest time point assessed), the FC brought about the highest fibronectin and collagen types I, III, IV, V and VI deposition. Deposited ECM pattern analysis showed that at day 10, the FC induced the lowest lacunarity and normalised end points and the highest fractal dimension and % high density matrix. Further immunofluorescence analysis revealed no significant differences between the groups in vimentin, aldehyde dehydrogenase 3 family member A1, keratocan, paired box protein 6 and α-smooth muscle actin. Importantly, at day 10, the FC resulted in the highest growth factor retention (20 molecules). Our data clearly illustrate a MMC agent dependent cell response, with the FC having the highest positive effect in hCF cultures.
Collapse
Affiliation(s)
- Mehmet Gurdal
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland.
| |
Collapse
|
13
|
Dave R, Pandey K, Patel R, Gour N, Bhatia D. Biological Scaffolds in 3D Cell Models: Driving Innovation in Drug Discovery. Stem Cell Rev Rep 2025; 21:147-166. [PMID: 39388081 DOI: 10.1007/s12015-024-10800-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 10/15/2024]
Abstract
The discipline of 3D cell modeling is currently undergoing a surge of captivating developments that are enhancing the realism and utility of tissue simulations. Using bioinks which represent cells, scaffolds, and growth factors scientists can construct intricate tissue architectures layer by layer using innovations like 3D bioprinting. Drug testing can be accelerated and organ functions more precisely replicated owing to the precise control that microfluidic technologies and organ-on-chip devices offer over the cellular environment. Tissue engineering is becoming more dynamic with materials that can modify their surroundings with the advent of hydrogels and smart biomaterials. Advances in spheroids and organoids are not only bringing us towards more effective and customized therapies, but they are also improving their ability to resemble actual human tissues. Confocal and two-photon microscopy are examples of advanced imaging methods that provide precise images of the functioning and interaction of cells. Artificial Intelligence models have applications for enhanced scaffold designs and for predicting the response of tissues to medications. Furthermore, via strengthening predictive models, optimizing data analysis, and simplifying 3D cell culture design, artificial intelligence is revolutionizing this field. When combined, these technologies are improving our ability to conduct research and moving us toward more individualized and effective medical interventions.
Collapse
Affiliation(s)
- Raj Dave
- Department of Chemistry, Indrashil University, Kadi, Mehsana, Gujarat, India
| | - Kshipra Pandey
- Department of Biosciences, Indrashil University, Kadi, Mehsana, Gujarat, India
| | - Ritu Patel
- Department of Biosciences, Indrashil University, Kadi, Mehsana, Gujarat, India
| | - Nidhi Gour
- Department of Chemistry, Indrashil University, Kadi, Mehsana, Gujarat, India.
| | - Dhiraj Bhatia
- Department of Biological Engineering Discipline, Indian Institute of Technology, Palaj, 382355, Gujarat, India.
| |
Collapse
|
14
|
Reutter S, Kern J, Jakob Y, Rotter N, Gvaramia D. Small spheroids for head and neck cartilage tissue engineering. Sci Rep 2024; 14:32114. [PMID: 39738737 PMCID: PMC11686322 DOI: 10.1038/s41598-024-83847-w] [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: 02/20/2024] [Accepted: 12/17/2024] [Indexed: 01/02/2025] Open
Abstract
The demand for cartilage reconstruction in the head and neck region arises frequently due to trauma, malignancies, and hereditary diseases. Traditional tissue engineering produces cartilage from a small biopsy by combining biomaterials and expanded cells. However, this top-down approach is associated with several limitations, including the non-uniform distribution of cells, lack of physiological cell-cell and cell-matrix interactions, and compromised mechanical properties and tissue architecture. The capacity of cells to aggregate into microtissues enables an alternative bottom-up approach to producing cartilage with or without further scaffolding support. Here we explored the optimal conditions for obtaining small spheroids from head and neck cartilage tissues. We used chondrocytes (CCs) and chondroprogenitors (CPCs) isolated from auricular and nasoseptal cartilage to prepare spheroids using ultra-low attachment (ULA) plates or micromass cultures. Different cell densities were tested to estimate the minimal cell number required for optimal spheroid formation. Furthermore, we evaluated the influence of key chondrogenic cytokines, such as transforming growth factor (TGF)-β, connective tissue growth factor (CTGF), and insulin-like growth factor (IGF)-1, on spheroid morphology and the production of cartilage extracellular matrix (ECM) components. Spheroids expressing cartilage markers were formed with 2.5 × 104 cells in a commercially available chondrogenic differentiation medium on ULA plates but not in conventional micromass cultures. Differences were seen in auricular and nasal spheroids with respect to growth patterns and response to cytokine composition. Auricular spheroids were larger and showed size increase in culture, whereas nasal aggregates tended to shrink. Cytokines differentially influenced spheroid growth, and ECM structure and composition. Under all tested conditions, both spheroid types generated one or more cartilage ECM components, including elastin, which was also found in nasal spheroids despite their hyaline origin. Our results suggest that spheroid cultures can offer a viable approach to generating mature cartilage tissue without a biomaterial scaffold. Furthermore, nasal CCs and CPCs can be used to generate elastic cartilage. The findings of the study provide technical insights toward the goal of obtaining cartilage microtissues that can be potentially used for reconstructive procedures of HNC cartilage defects.
Collapse
Affiliation(s)
- Sven Reutter
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Clinic Mannheim, University of Heidelberg, Mannheim, Germany
| | - Johann Kern
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Clinic Mannheim, University of Heidelberg, Mannheim, Germany
| | - Yvonne Jakob
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Clinic Mannheim, University of Heidelberg, Mannheim, Germany
| | - Nicole Rotter
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Department of Otorhinolaryngology, Head and Neck Surgery, University Clinic Mannheim, University of Heidelberg, Mannheim, Germany
| | - David Gvaramia
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
- Department of Otorhinolaryngology, Head and Neck Surgery, University Clinic Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
15
|
Rendon-Romero LM, Rojas-Martinez A. Advances in the Development of Auricular Cartilage Bioimplants. TISSUE ENGINEERING. PART B, REVIEWS 2024. [PMID: 39723986 DOI: 10.1089/ten.teb.2024.0227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Conditions such as congenital abnormalities, cancer, infections, and trauma can severely impact the integrity of the auricular cartilage, resulting in the need for a replacement structure. Current implants, carved from the patient's rib, involve multiple surgeries and carry risks of adverse events such as contamination, rejection, and reabsorption. Tissue engineering aims to develop lifelong auricular bioimplants using different methods, different cell types, growth factors and maintenance media formulations, and scaffolding materials compatible with the host. This review aims to examine the progress in auricular bioengineering, focusing on improvements derived from in vivo models and clinical trials, as well as the author's suggestions to enhance the methods. For this scope review, 30 articles were retrieved through Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, plus 6 manually selected articles. The methods reported in the articles were categorized into four levels according to the development phases: source of cells, cell media supplementation, scaffold, or scaffold-free methods, and experimental in vivo or clinical approaches. Many methods have demonstrated potential for the development of bioimplants; four clinical trials reported a structure like the external ear that could be maintained after overcoming post-transplant inflammation. However, several challenges must be solved, such as obtaining a structure that accurately replicates the shape and size of the patient's healthy contralateral auricle and improvements to avoid immunological rejection and resorption of the bioimplant.
Collapse
|
16
|
Martin R, Joung D. The Promise and Challenges of Bioprinting in Tissue Engineering. MICROMACHINES 2024; 15:1529. [PMID: 39770282 PMCID: PMC11727761 DOI: 10.3390/mi15121529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 12/21/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025]
Abstract
Organ transplantation, biomimetic organ models, and the restoration of damaged or eviscerated tissues have been key goals in surgical and medical research since their inception [...].
Collapse
Affiliation(s)
- Ryan Martin
- Department of Physics, Virginia Commonwealth University, Richmond, VA 23284, USA;
| | - Daeha Joung
- Department of Physics, Virginia Commonwealth University, Richmond, VA 23284, USA;
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
17
|
García-Sobrino R, Martínez-Campos E, Marcos-Ríos D, Zepeda-Rodríguez Z, Valentín JL, Sanz-Horta R, León-Calero M, Reinecke H, Elvira C, Gallardo A, Rodríguez-Hernández J. Development of Thermosensitive Hydrogels with Tailor-Made Geometries to Modulate Cell Harvesting of Non-Flat Cell Cultures. Gels 2024; 10:802. [PMID: 39727560 DOI: 10.3390/gels10120802] [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: 10/13/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024] Open
Abstract
Considering the complexity in terms of design that characterizes the different tissues of the human body, it is necessary to study and develop more precise therapies. In this sense, this article presents the possibility of fabricating photocurable thermosensitive hydrogels with free geometry and based on N-Vinyl Caprolactam (VCL) with the aim of modulating the adhesion of non-planar cell cultures. The fabrication process is based on the use as a mold of two-layer thick water-soluble polyvinyl alcohol (PVA) previously printed by Extrusion Material (MatEx). From this technology it has been possible to obtain hydrogels with different 3D geometries and different crosslinking percentages (2, 4 and 6 mol%). Studies have shown that networks reduce their thermosensitivity not only when the percentage of crosslinking in the formulation increases, but also when the thickness of the hydrogel obtained increases. Based on this reduction in thermosensitivity, the less crosslinked (2 mol%) hydrogels have been evaluated to carry out a novel direct application in which hydrogels with curved geometry have allowed cell adhesion and proliferation at 37 °C with the endothelial cell line C166-GFP; likewise, non-aggressive cell detachment was observed when the hydrogel temperature was reduced to values of 20 °C. Therefore, the present manuscript shows a novel application for the synthesis of free-form thermosensitive hydrogels that allows modulation of non-planar cell cultures.
Collapse
Affiliation(s)
- Rubén García-Sobrino
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
- Group of Organic Synthesis and Bioevaluation, Instituto Pluridisciplinar, Universidad Complutense de Madrid (UCM), Associated Unit to the ICTP-IQM-CSIC, Paseo Juan XXIII, n° 1, 28040 Madrid, Spain
- Department of Applied Mathematics, Materials Science and Engineering and Electronic Technology, Universidad Rey Juan Carlos, Calle Tulipán s/n, 28933 Móstoles, Spain
| | - Enrique Martínez-Campos
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
- Group of Organic Synthesis and Bioevaluation, Instituto Pluridisciplinar, Universidad Complutense de Madrid (UCM), Associated Unit to the ICTP-IQM-CSIC, Paseo Juan XXIII, n° 1, 28040 Madrid, Spain
| | - Daniel Marcos-Ríos
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
- Group of Organic Synthesis and Bioevaluation, Instituto Pluridisciplinar, Universidad Complutense de Madrid (UCM), Associated Unit to the ICTP-IQM-CSIC, Paseo Juan XXIII, n° 1, 28040 Madrid, Spain
| | - Zenen Zepeda-Rodríguez
- Elastomers Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Juan L Valentín
- Elastomers Group, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Raúl Sanz-Horta
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Marina León-Calero
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Helmut Reinecke
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Carlos Elvira
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Alberto Gallardo
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| | - Juan Rodríguez-Hernández
- Polymer Functionalization Group, Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros-Consejo Superior de Investigaciones Científicas (ICTP-CSIC), Calle Juan de la Cierva, n° 3, 28006 Madrid, Spain
| |
Collapse
|
18
|
Son KH, Kim DH, Park S, Kim HJ, Park M, Kim SJ, Lee SJ, Ahn K, Lee JW. Spherical Shell Bioprinting to Produce Uniform Spheroids with Controlled Sizes. J Funct Biomater 2024; 15:350. [PMID: 39590553 PMCID: PMC11595458 DOI: 10.3390/jfb15110350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/14/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
Conventional cell spheroid production methods are largely manual, leading to variations in size and shape that compromise consistency and reliability for use in cell-based therapeutic applications. To enhance spheroid production, a spherical shell bioprinting system was implemented, enabling the high-throughput generation of uniform cell spheroids with precisely controlled sizes. The system encapsulates cells within thin alginate hydrogel shells formed through bioprinting and ion crosslinking reactions. Alginate-calcium ion crosslinking created alginate shells that contained gelatin-based bioinks with embedded cells, facilitating spontaneous cell aggregation within the shells and eliminating the need for plastic wells. By adjusting cell concentrations in the alginate-gelatin bioink, we achieved precise control over spheroid size, maintaining a sphericity above 0.94 and size deviations within ±10 µm. This method has been successfully applied to various cell types including cancer cells, fibroblasts, chondrocytes, and epithelial cells, demonstrating its versatility. This scalable approach enhances the reliability of cell therapy and drug screening, offering a robust platform for future biomedical applications.
Collapse
Affiliation(s)
- Kuk Hui Son
- Department of Thoracic and Cardiovascular Surgery, Gil Medical Center, College of Medicine, Gachon University, 21, Namdong-daero 774 Beon-gil, Namdong-gu, Incheon 21565, Republic of Korea;
| | - Dong-Ha Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Seunghye Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea;
| | - Hyun Jae Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Mira Park
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Seung-Jin Kim
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Medical Center Boulevard, Winston-Salem, NC 27157, USA;
| | - Keunsun Ahn
- Research Institute, Sphebio Co., Ltd., 501-ho, 3, Achasan-ro 11ga-gil, Seongdong-gu, Seoul 04796, Republic of Korea; (D.-H.K.); (H.J.K.); (M.P.); (S.-J.K.)
| | - Jin Woo Lee
- Department of Health Sciences and Technology, GAIHST, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea;
- Department of Molecular Medicine, College of Medicine, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea
| |
Collapse
|
19
|
Luo X, Pang Z, Li J, Anh M, Kim BS, Gao G. Bioengineered human arterial equivalent and its applications from vascular graft to in vitro disease modeling. iScience 2024; 27:111215. [PMID: 39555400 PMCID: PMC11565542 DOI: 10.1016/j.isci.2024.111215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2024] Open
Abstract
Arterial disorders such as atherosclerosis, thrombosis, and aneurysm pose significant health risks, necessitating advanced interventions. Despite progress in artificial blood vessels and animal models aimed at understanding pathogenesis and developing therapies, limitations in graft functionality and species discrepancies restrict their clinical and research utility. Addressing these issues, bioengineered arterial equivalents (AEs) with enhanced vascular functions have been developed, incorporating innovative technologies that improve clinical outcomes and enhance disease progression modeling. This review offers a comprehensive overview of recent advancements in bioengineered AEs, systematically summarizing the bioengineered technologies used to construct these AEs, and discussing their implications for clinical application and pathogenesis understanding. Highlighting current breakthroughs and future perspectives, this review aims to inform and inspire ongoing research in the field, potentially transforming vascular medicine and offering new avenues for preclinical and clinical advances.
Collapse
Affiliation(s)
- Xi Luo
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Zherui Pang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology, Zhuhai 519088, China
| | - Minjun Anh
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
20
|
Kim M, Wang X, Li Y, Lin Z, Collins CP, Liu Y, Ahn Y, Tsal HM, Song JW, Duan C, Zhu Y, Sun C, He TC, Luo Y, Reid RR, Ameer GA. Personalized composite scaffolds for accelerated cell- and growth factor-free craniofacial bone regeneration. Bioact Mater 2024; 41:427-439. [PMID: 39188380 PMCID: PMC11345904 DOI: 10.1016/j.bioactmat.2024.07.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 08/28/2024] Open
Abstract
Approaches to regenerating bone often rely on integrating biomaterials and biological signals in the form of cells or cytokines. However, from a translational point of view, these approaches are challenging due to the sourcing and quality of the biologic, unpredictable immune responses, complex regulatory paths, and high costs. We describe a simple manufacturing process and a material-centric 3D-printed composite scaffold system (CSS) that offers distinct advantages for clinical translation. The CSS comprises a 3D-printed porous polydiolcitrate-hydroxyapatite composite elastomer infused with a polydiolcitrate-graphene oxide hydrogel composite. Using a micro-continuous liquid interface production 3D printer, we fabricate a precise porous ceramic scaffold with 60 wt% hydroxyapatite resembling natural bone. The resulting scaffold integrates with a thermoresponsive hydrogel composite in situ to fit the defect, which is expected to enhance surface contact with surrounding tissue and facilitate biointegration. The antioxidative properties of citrate polymers prevent long-term inflammatory responses. The CSS stimulates osteogenesis in vitro and in vivo. Within 4 weeks in a calvarial critical-sized bone defect model, the CSS accelerated ECM deposition (8-fold) and mineralized osteoid (69-fold) compared to the untreated. Through spatial transcriptomics, we demonstrated the comprehensive biological processes of CSS for prompt osseointegration. Our material-centric approach delivers impressive osteogenic properties and streamlined manufacturing advantages, potentially expediting clinical application for bone reconstruction surgeries.
Collapse
Affiliation(s)
- Mirae Kim
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Xinlong Wang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yiming Li
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Zitong Lin
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Caralyn P. Collins
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208 USA
| | - Yugang Liu
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yujin Ahn
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61820, USA
| | - Hsiu-Ming Tsal
- Department of Radiology, The University of Chicago, Chicago, IL, 60637, USA
| | - Joseph W. Song
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Chongwen Duan
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Cheng Sun
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208 USA
| | - Tong-Chuan He
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Clinical and Translational Sciences Institute, Northwestern University, Chicago, IL, 60611, USA
- Center for Collaborative AI in Healthcare, Institute for AI in Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Russell R. Reid
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Guillermo A. Ameer
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
- Center for Advanced Regenerative Engineering, Northwestern University, Evanston, IL, 60208, USA
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Chemistry of Life Process Institute, Northwestern University, Chicago, IL, 60208, USA
- International Institute for Nanotechnology, Northwestern University, Evanston, IL, 60208, USA
| |
Collapse
|
21
|
Panchuk I, Smirnikhina S. Toolbox for creating three-dimensional liver models. Biochem Biophys Res Commun 2024; 731:150375. [PMID: 39018971 DOI: 10.1016/j.bbrc.2024.150375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/15/2024] [Accepted: 07/08/2024] [Indexed: 07/19/2024]
Abstract
Research within the hepato-biliary system and hepatic function is currently experiencing heightened interest, this is due to the high frequency of relapse rates observed in chronic conditions, as well as the imperative for the development of innovative therapeutic strategies to address both inherited and acquired diseases within this domain. The most commonly used sources for studying hepatocytes include primary human hepatocytes, human hepatic cancer cell lines, and hepatic-like cells derived from induced pluripotent stem cells. However, a significant challenge in primary hepatic cell culture is the rapid decline in their phenotypic characteristics, dedifferentiation and short cultivation time. This limitation creates various problems, including the inability to maintain long-term cell cultures, which can lead to failed experiments in drug development and the creation of relevant disease models for researchers' purposes. To address these issues, the creation of a powerful 3D cell model could play a pivotal role as a personalized disease model and help reduce the use of animal models during certain stages of research. Such a cell model could be used for disease modelling, genome editing, and drug discovery purposes. This review provides an overview of the main methods of 3D-culturing liver cells, including a discussion of their characteristics, advantages, and disadvantages.
Collapse
Affiliation(s)
- Irina Panchuk
- Research Centre for Medical Genetics, Moscow, Russian Federation.
| | | |
Collapse
|
22
|
Dong Y, Zhou X, Ding Y, Luo Y, Zhao H. Advances in tumor microenvironment: Applications and challenges of 3D bioprinting. Biochem Biophys Res Commun 2024; 730:150339. [PMID: 39032359 DOI: 10.1016/j.bbrc.2024.150339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024]
Abstract
The tumor microenvironment (TME) assumes a pivotal role in the treatment of oncological diseases, given its intricate interplay of diverse cellular components and extracellular matrices. This dynamic ecosystem poses a serious challenge to traditional research methods in many ways, such as high research costs, inefficient translation, poor reproducibility, and low modeling success rates. These challenges require the search for more suitable research methods to accurately model the TME, and the emergence of 3D bioprinting technology is transformative and an important complement to these traditional methods to precisely control the distribution of cells, biomolecules, and matrix scaffolds within the TME. Leveraging digital design, the technology enables personalized studies with high precision, providing essential experimental flexibility. Serving as a critical bridge between in vitro and in vivo studies, 3D bioprinting facilitates the realistic 3D culturing of cancer cells. This comprehensive article delves into cutting-edge developments in 3D bioprinting, encompassing diverse methodologies, biomaterial choices, and various 3D tumor models. Exploration of current challenges, including limited biomaterial options, printing accuracy constraints, low reproducibility, and ethical considerations, contributes to a nuanced understanding. Despite these challenges, the technology holds immense potential for simulating tumor tissues, propelling personalized medicine, and constructing high-resolution organ models, marking a transformative trajectory in oncological research.
Collapse
Affiliation(s)
- Yingying Dong
- The First School of Climical Medicine of Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xue Zhou
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China; State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China.
| | - Yunyi Ding
- Department of Emergency Medicine, The Second Affiliated Hospital of Zhejiang University, School, Hangzhou, 310009, China.
| | - Yichen Luo
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China; State Key Laboratory of Fluid Power & Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China.
| | - Hong Zhao
- The First School of Climical Medicine of Zhejiang Chinese Medical University, Hangzhou, 310053, China; Department of Breast Surgery, The First Affiliated Hospital of Zhejiang University of Traditional Chinese Medicine, (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, 310060, China.
| |
Collapse
|
23
|
Wu KY, Belaiche M, Wen Y, Choulakian MY, Tran SD. Advancements in Polymer Biomaterials as Scaffolds for Corneal Endothelium Tissue Engineering. Polymers (Basel) 2024; 16:2882. [PMID: 39458711 PMCID: PMC11511139 DOI: 10.3390/polym16202882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Corneal endothelial dysfunction is a leading cause of vision loss globally, frequently requiring corneal transplantation. However, the limited availability of donor tissues, particularly in developing countries, has spurred on the exploration of tissue engineering strategies, with a focus on polymer biomaterials as scaffolds for corneal endotlhelium regeneration. This review provides a comprehensive overview of the advancements in polymer biomaterials, focusing on their role in supporting the growth, differentiation, and functional maintenance of human corneal endothelial cells (CECs). Key properties of scaffold materials, including optical clarity, biocompatibility, biodegradability, mechanical stability, permeability, and surface wettability, are discussed in detail. The review also explores the latest innovations in micro- and nano-topological morphologies, fabrication techniques such as electrospinning and 3D/4D bioprinting, and the integration of drug delivery systems into scaffolds. Despite significant progress, challenges remain in translating these technologies to clinical applications. Future directions for research are highlighted, including the need for improved biomaterial combinations, a deeper understanding of CEC biology, and the development of scalable manufacturing processes. This review aims to serve as a resource for researchers and clinician-scientists seeking to advance the field of corneal endothelium tissue engineering.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Myriam Belaiche
- Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Ying Wen
- Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Mazen Y. Choulakian
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
24
|
Yuan Z, Zhang Y, He X, Wang X, Wang X, Ren S, Su J, Shen J, Li X, Xiao Z. Engineering mesenchymal stem cells for premature ovarian failure: overcoming challenges and innovating therapeutic strategies. Theranostics 2024; 14:6487-6515. [PMID: 39479455 PMCID: PMC11519806 DOI: 10.7150/thno.102641] [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: 08/21/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024] Open
Abstract
Premature ovarian failure (POF) is a leading cause of infertility in women, causing significant psychological and physical distress. Current therapeutic options are limited, necessitating the exploration of new treatments. Mesenchymal stem cells (MSCs), known for their remarkable homing and regenerative properties, have emerged as a promising intervention for POF. However, their clinical efficacy has been inconsistent. This paper aims to address these challenges by examining the cellular heterogeneity within MSC populations, which is crucial for identifying and selecting specific functional subpopulations for clinical applications. Understanding this heterogeneity can enhance therapeutic efficacy and ensure treatment stability. Additionally, this review comprehensively examines the literature on the effectiveness, safety, and ethical considerations of MSCs for ovarian regeneration, with a focus on preclinical and clinical trials. We also discuss potential strategies involving genetically and tissue-engineered MSCs. By integrating insights from these studies, we propose new directions for the design of targeted MSC treatments for POF and related disorders, potentially improving outcomes, addressing safety concerns, and expanding therapeutic options while ensuring ethical compliance.
Collapse
Affiliation(s)
- Zijun Yuan
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yinping Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xinyu He
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiang Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xingyue Wang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Siqi Ren
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiahong Su
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Xiang Li
- Sichuan College of Traditional Chinese Medicine, Sichuan Mianyang 621000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
- Department of Pharmacology, School of Pharmacy, Sichuan College of Traditional Chinese Medicine, Sichuan Mianyang 621000, China
- Luzhou People's Hospital, Luzhou, Sichuan, China
| |
Collapse
|
25
|
Patil SJ, Thorat VM, Koparde AA, Bhosale RR, Bhinge SD, Chavan DD, Tiwari DD. Theranostic Applications of Scaffolds in Current Biomedical Research. Cureus 2024; 16:e71694. [PMID: 39559663 PMCID: PMC11571282 DOI: 10.7759/cureus.71694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 10/17/2024] [Indexed: 11/20/2024] Open
Abstract
Theranostics, a remarkable combination of diagnostics and therapeutics, has given rise to tissue/organ-format theranostic scaffolds that integrate targeted therapy and real-time disease monitoring. The scaffold is a 3D structuring template for cell or tissue attachment and growth. These scaffolds offer unprecedented opportunities for personalized medicine and hold great potential for revolutionizing healthcare. Recent advancements in fabrication techniques have enabled the creation of highly intricate and precisely engineered scaffolds with controllable physical and chemical properties, enhancing their therapeutic potential for tissue engineering and regenerative medicine. This paper proposes a new categorization method for scaffolds in tissue engineering based on the relativity of scaffold design-independent parameters. Five types of scaffolds are defined at different levels, highlighting the importance of understanding and analyzing scaffold types. It possesses the ability to seamlessly integrate diagnostics and therapeutics within a single platform, enhancing the efficacy and precision of personalized medicine. Natural scaffolds derived from biomaterials and synthetic scaffolds fabricated by human intervention are discussed, with synthetic scaffolds offering advantages such as tunable mechanical properties and controlled drug delivery, while natural scaffolds provide inherent biocompatibility and bioactivity, making them ideal for promoting cellular responses. The use of synthetic scaffolds shows great promise in advancing regenerative medicine and improving patient outcomes. The transfer of new technologies and changes in society have accelerated the evolution of health monitoring into the era of personal health monitoring. Using emerging health data, cost-effective analytics, wireless sensor networks, mobile smartphones, and easy internet access, the combination of these technologies is expected to accelerate the transition to personal health monitoring outside of traditional healthcare settings. The main objective of this review article is to provide a comprehensive overview of the theranostic applications of scaffolds in current biomedical research, highlighting their dual role in therapy and diagnostics. The review aims to explore the latest advancements in scaffold design, fabrication, and functionalization, emphasizing how these innovations contribute to improved therapeutic efficacy, targeted drug delivery, and the real-time monitoring of disease progression across various medical fields.
Collapse
Affiliation(s)
- Sarika J Patil
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Vandana M Thorat
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Akshada A Koparde
- Department of Pharmaceutical Chemistry, Krishna Institute of Pharmacy, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Rohit R Bhosale
- Department of Pharmaceutics, Krishna Foundation's Jaywant Institute of Pharmacy, Karad, IND
| | - Somnath D Bhinge
- Department of Pharmaceutical Chemistry, Rajarambapu College of Pharmacy, Kasegaon, IND
| | - Dhanashri D Chavan
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| | - Devkumar D Tiwari
- Department of Pharmacology, Krishna Institute of Medical Sciences, Krishna Vishwa Vidyapeeth (Deemed to be University), Karad, IND
| |
Collapse
|
26
|
Rampin A, Rossoni A, Chaniotaki L, Gkiatas IS, Tzora A, Skoufos I, Diakakis N, Prassinos N, Zeugolis DI. Xenogeneic versus allogeneic serum and macromolecular crowding in human tenocyte cultures. Eur J Cell Biol 2024; 103:151445. [PMID: 39024989 DOI: 10.1016/j.ejcb.2024.151445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024] Open
Abstract
Allogeneic serum and tissue-specific extracellular matrix have been shown to maintain permanently differentiated cell phenotype in culture. This is of particular importance for human tenocytes, a cell population that readily loses its function during ex vivo culture. With these in mind, herein we extracted human tenocytes using either foetal bovine serum or human serum, cultured them in the absence and presence of carrageenan and Ficoll®, the most widely used macromolecular crowding agents (to induce tissue-specific extracellular matrix deposition), and assessed cellular function, via metabolic activity, viability, proliferation and immunofluorescence for collagen related molecules, non-collagenous molecules and transmembrane molecules. At day 7, longest time point assessed, neither carrageenan nor Ficoll® significantly affected metabolic activity, viability and proliferation in either serum and human serum significantly increased metabolic activity and proliferation. At day 7, in the absence of macromolecular crowding, cells in human serum deposited significantly lower collagen type VI, biglycan, versican and tenomodulin than cells in foetal bovine serum. Interestingly, at day 7, in comparison to the no macromolecular crowding group, carrageenan in foetal bovine serum induced the highest effect, as judged by the highest number of significantly increased molecules (collagen type I, collagen type IV, collagen type V, collagen type VI, transforming growth factor β1, matrix metalloproteinase 14, lumican, versican, scleraxis and integrin α2β1). These data, although contradict previous observations where human serum outperformed foetal bovine serum, at the same time, support the use of foetal bovine serum in the development of cell-based medicines.
Collapse
Affiliation(s)
- Andrea Rampin
- Department of Agriculture, University of Ioannina, Arta, Greece; School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece; Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Andrea Rossoni
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Lefki Chaniotaki
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Ioannis S Gkiatas
- Department of Orthopaedic Surgery, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Athina Tzora
- Department of Agriculture, University of Ioannina, Arta, Greece
| | - Ioannis Skoufos
- Department of Agriculture, University of Ioannina, Arta, Greece
| | - Nikolaos Diakakis
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikitas Prassinos
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland.
| |
Collapse
|
27
|
Park KH, Truong TT, Park JH, Park Y, Kim H, Hyun SA, Shim HE, Mallick S, Park HJ, Huh KM, Kang SW. Robust and customizable spheroid culture system for regenerative medicine. Biofabrication 2024; 16:045016. [PMID: 39053497 DOI: 10.1088/1758-5090/ad6795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/25/2024] [Indexed: 07/27/2024]
Abstract
Three-dimensional cell spheroids show promise for the reconstruction of native tissues. Herein, we report a sophisticated, uniform, and highly reproducible spheroid culture system for tissue reconstruction. A mesh-integrated culture system was designed to precisely control the uniformity and reproducibility of spheroid formation. Furthermore, we synthesized hexanoyl glycol chitosan, a material with ultralow cell adhesion properties, to further improve spheroid formation efficiency and biological function. Our results demonstrate improved biological function in various types of cells and ability to generate spheroids with complex structures composed of multiple cell types. In conclusion, our spheroid culture system offers a highly effective and widely applicable approach to generating customized spheroids with desired structural and biological features for a variety of biomedical applications.
Collapse
Affiliation(s)
- Kyoung Hwan Park
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Thuy Trang Truong
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 11765, Republic of Korea
| | - Yujin Park
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hyeok Kim
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 11765, Republic of Korea
| | - Sung-Ae Hyun
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejoen 34114, Republic of Korea
| | - Hye-Eun Shim
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sudipta Mallick
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 11765, Republic of Korea
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Kang Moo Huh
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
- Department of Materials Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sun-Woong Kang
- Research Group for Biomimetic Advanced Technology, Korea Institute of Toxicology (KIT), Daejeon 34114, Republic of Korea
- Human and Environmental Toxicology Program, University of Science and Technology, Daejeon 34114, Republic of Korea
| |
Collapse
|
28
|
Tian JS, Tay A. Progress on Electro-Enhancement of Cell Manufacturing. SMALL METHODS 2024; 8:e2301281. [PMID: 38059759 DOI: 10.1002/smtd.202301281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/09/2023] [Indexed: 12/08/2023]
Abstract
With the long persistence of complex, chronic diseases in society, there is increasing motivation to develop cells as living medicine to treat diseases ranging from cancer to wounds. While cell therapies can significantly impact healthcare, the shortage of starter cells meant that considerable raw materials must be channeled solely for cell expansion, leading to expensive products with long manufacturing time which can prevent accessibility by patients who either cannot afford the treatment or have highly aggressive diseases and cannot wait that long. Over the last three decades, there has been increasing knowledge on the effects of electrical modulation on proliferation, but to the best of the knowledge, none of these studies went beyond how electro-control of cell proliferation may be extended to enhance industrial scale cell manufacturing. Here, this review is started by discussing the importance of maximizing cell yield during manufacturing before comparing strategies spanning biomolecular/chemical/physical to modulate cell proliferation. Next, the authors describe how factors governing invasive and non-invasive electrical stimulation (ES) including capacitive coupling electric field may be modified to boost cell manufacturing. This review concludes by describing what needs to be urgently performed to bridge the gap between academic investigation of ES to industrial applications.
Collapse
Affiliation(s)
- Johann Shane Tian
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, 117599, Singapore
- NUS Tissue Engineering Program, National University of Singapore, Singapore, 117510, Singapore
| |
Collapse
|
29
|
Yoon S, Fuwad A, Jeong S, Cho H, Jeon TJ, Kim SM. Surface Deformation of Biocompatible Materials: Recent Advances in Biological Applications. Biomimetics (Basel) 2024; 9:395. [PMID: 39056836 PMCID: PMC11274418 DOI: 10.3390/biomimetics9070395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/28/2024] Open
Abstract
The surface topography of substrates is a crucial factor that determines the interaction with biological materials in bioengineering research. Therefore, it is important to appropriately modify the surface topography according to the research purpose. Surface topography can be fabricated in various forms, such as wrinkles, creases, and ridges using surface deformation techniques, which can contribute to the performance enhancement of cell chips, organ chips, and biosensors. This review provides a comprehensive overview of the characteristics of soft, hard, and hybrid substrates used in the bioengineering field and the surface deformation techniques applied to the substrates. Furthermore, this review summarizes the cases of cell-based research and other applications, such as biosensor research, that utilize surface deformation techniques. In cell-based research, various studies have reported optimized cell behavior and differentiation through surface deformation, while, in the biosensor and biofilm fields, performance improvement cases due to surface deformation have been reported. Through these studies, we confirm the contribution of surface deformation techniques to the advancement of the bioengineering field. In the future, it is expected that the application of surface deformation techniques to the real-time interaction analysis between biological materials and dynamically deformable substrates will increase the utilization and importance of these techniques in various fields, including cell research and biosensors.
Collapse
Affiliation(s)
- Sunhee Yoon
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.Y.); (H.C.)
- Industry-Academia Interactive R&E Center for Bioprocess Innovation (BK21), Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Ahmed Fuwad
- Department of Mechanical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (A.F.); (S.J.)
| | - Seorin Jeong
- Department of Mechanical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (A.F.); (S.J.)
| | - Hyeran Cho
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.Y.); (H.C.)
| | - Tae-Joon Jeon
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.Y.); (H.C.)
- Industry-Academia Interactive R&E Center for Bioprocess Innovation (BK21), Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
- Biohybrid Systems Research Center, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| | - Sun Min Kim
- Department of Biological Sciences and Bioengineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (S.Y.); (H.C.)
- Department of Mechanical Engineering, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea; (A.F.); (S.J.)
- Biohybrid Systems Research Center, Inha University, 100, Inha-ro, Michuhol-gu, Incheon 22212, Republic of Korea
| |
Collapse
|
30
|
Kim HE, Ju HJ, Kim S, Kim YH, Lee S, Choi S, Yoon HC, Choi HS, Kim MS. Amplifying endogenous stem cell migration for in situ bone tissue formation: Substance P analog and BMP mimetic peptide-loaded click-crosslinked hyaluronic acid hydrogel. Mater Today Bio 2024; 26:101070. [PMID: 38711939 PMCID: PMC11070699 DOI: 10.1016/j.mtbio.2024.101070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/08/2024] Open
Abstract
Endogenous stem cell-driven in situ bone tissue formation has recently garnered increasing attention. Therefore, our study sought to refine methods to enhance the migration and subsequent osteogenic differentiation of these cells. Our innovative approach involves using an injectable hydrogel that combines click cross-linking sites and a BMP-2 mimetic peptide (BP) with hyaluronic acid (HA). This injectable formulation, hereinafter referred to as SPa + Cx-HA-BP, incorporates a substance P analog peptide (SPa) with Cx-HA-BP, proving versatile for in vitro and in vivo applications without cytotoxicity. The controlled release of SPa creates a gradient that guides endogenous stem cells towards the Cx-HA scaffold from specific tissue niches. Both Cx-HA and SPa+Cx-HA induced minimal changes in the expression of genes associated with osteogenic differentiation. In contrast, these genes were robustly induced by both SPa + Cx-HA+BP and SPa + Cx-HA-BP, in which BP was respectively integrated via physical and chemical methods. Remarkably, chemically incorporating BP (Cx-HA-BP) resulted in 4-9 times higher osteogenic gene expression than physically mixed BP in Cx-HA+BP. This study validates the role of SPa role in guiding endogenous stem cells toward the hydrogel and underscores the substantial impact of sustained BP presence within the hydrogel. Collectively, our findings offer valuable insights for the development of innovative strategies to promote endogenous stem cell-based tissue regeneration. The developed hydrogel effectively guides stem cells from their natural locations and facilitates sustained osteogenic differentiation, thus holding great promise for applications in regenerative medicine.
Collapse
Affiliation(s)
- Hee Eun Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Hyeon Jin Ju
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Shina Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Young Hun Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Soyeon Lee
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Hyun C. Yoon
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, South Korea
- Research Institute, Medipolymer, Woncheon Dong 274, Suwon, 16522, South Korea
| |
Collapse
|
31
|
Matsuzaka Y, Yashiro R. Current Strategies and Therapeutic Applications of Mesenchymal Stem Cell-Based Drug Delivery. Pharmaceuticals (Basel) 2024; 17:707. [PMID: 38931374 PMCID: PMC11206583 DOI: 10.3390/ph17060707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have emerged as a promising approach for drug delivery strategies because of their unique properties. These strategies include stem cell membrane-coated nanoparticles, stem cell-derived extracellular vesicles, immunomodulatory effects, stem cell-laden scaffolds, and scaffold-free stem cell sheets. MSCs offer advantages such as low immunogenicity, homing ability, and tumor tropism, making them ideal for targeted drug delivery systems. Stem cell-derived extracellular vesicles have gained attention for their immune properties and tumor-homing abilities, presenting a potential solution for drug delivery challenges. The relationship between MSC-based drug delivery and the self-renewal and differentiation capabilities of MSCs lies in the potential of engineered MSCs to serve as effective carriers for therapeutic agents while maintaining their intrinsic properties. MSCs exhibit potent immunosuppressive functions in MSC-based drug delivery strategies. Stem cell-derived EVs have low immunogenicity and strong therapeutic potential for tissue repair and regeneration. Scaffold-free stem cell sheets represent a cutting-edge approach in regenerative medicine, offering a versatile platform for tissue engineering and regeneration across different medical specialties. MSCs have shown great potential for clinical applications in regenerative medicine because of their ability to differentiate into various cell types, secrete bioactive factors, and modulate immune responses. Researchers are exploring these innovative approaches to enhance drug delivery efficiency and effectiveness in treating various diseases.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan;
- Department of Medical Molecular Informatics, Meiji Pharmaceutical University, Kiyose 204-8588, Tokyo, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Tokyo, Japan;
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
32
|
Owaidah A. Induced pluripotent stem cells in cartilage tissue engineering: a literature review. Biosci Rep 2024; 44:BSR20232102. [PMID: 38563479 PMCID: PMC11088306 DOI: 10.1042/bsr20232102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/04/2024] Open
Abstract
Osteoarthritis (OA) is a long-term, persistent joint disorder characterized by bone and cartilage degradation, resulting in tightness, pain, and restricted movement. Current attempts in cartilage regeneration are cell-based therapies using stem cells. Multipotent stem cells, such as mesenchymal stem cells (MSCs), and pluripotent stem cells, such as embryonic stem cells (ESCs), have been used to regenerate cartilage. However, since the discovery of human-induced pluripotent stem cells (hiPSCs) in 2007, it was seen as a potential source for regenerative chondrogenic therapy as it overcomes the ethical issues surrounding the use of ESCs and the immunological and differentiation limitations of MSCs. This literature review focuses on chondrogenic differentiation and 3D bioprinting technologies using hiPSCS, suggesting them as a viable source for successful tissue engineering. METHODS A literature search was conducted using scientific search engines, PubMed, MEDLINE, and Google Scholar databases with the terms 'Cartilage tissue engineering' and 'stem cells' to retrieve published literature on chondrogenic differentiation and tissue engineering using MSCs, ESCs, and hiPSCs. RESULTS hiPSCs may provide an effective and autologous treatment for focal chondral lesions, though further research is needed to explore the potential of such technologies. CONCLUSIONS This review has provided a comprehensive overview of these technologies and the potential applications for hiPSCs in regenerative medicine.
Collapse
Affiliation(s)
- Amani Y. Owaidah
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
33
|
Kaboodkhani R, Mehrabani D, Moghaddam A, Salahshoori I, Khonakdar HA. Tissue engineering in otology: a review of achievements. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:1105-1153. [PMID: 38386362 DOI: 10.1080/09205063.2024.2318822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 02/09/2024] [Indexed: 02/23/2024]
Abstract
Tissue engineering application in otology spans a distance from the pinna to auditory nerve covered with specialized tissues and functions such as sense of hearing and aesthetics. It holds the potential to address the barriers of lack of donor tissue, poor tissue match, and transplant rejection through provision of new and healthy tissues similar to the host and possesses the capacity to renew, to regenerate, and to repair in-vivo and was shown to be a bypasses for any need to immunosuppression. This review aims to investigate the application of tissue engineering in otology and to evaluate the achievements and challenges in external, middle and inner ear sections. Since gaining the recent knowledge and training on use of different scaffolds is essential for otology specialists and who look for the recovery of ear function and aesthetics of patients, it is shown in this review how utilizing tissue engineering and cell transplantation, regenerative medicine can provide advancements in hearing and ear aesthetics to fit different patients' needs.
Collapse
Affiliation(s)
- Reza Kaboodkhani
- Otorhinolaryngology Research Center, Department of Otorhinolaryngology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | - Davood Mehrabani
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
- Stem Cell Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Fars, Iran
| | | | | | - Hossein Ali Khonakdar
- Iran Polymer and Petrochemical Institute (IPPI), Tehran, Iran
- Max Bergmann Center of Biomaterials and Institute of Materials Science, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
34
|
Anitua E, Troya M, Zalduendo M, Tierno R, Alkhraisat MH, Osinalde N, Fullaondo A, Zubiaga AM. Improving the mechanical and biological functions of cell sheet constructs: The interplay of human-derived periodontal ligament stem cells, endothelial cells and plasma rich in growth factors. Biomed Pharmacother 2024; 174:116599. [PMID: 38640711 DOI: 10.1016/j.biopha.2024.116599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024] Open
Abstract
OBJECTIVE The aim of this study was to produce and characterize triple-layered cell sheet constructs with varying cell compositions combined or not with the fibrin membrane scaffold obtained by the technology of Plasma Rich in Growth Factors (mPRGF). MATERIALS AND METHODS Human primary cultures of periodontal ligament stem cells (hPDLSCs) were isolated, and their stemness nature was evaluated. Three types of triple-layered composite constructs were generated, composed solely of hPDLSCs or combined with human umbilical vein endothelial cells (HUVECs), either as a sandwiched endothelial layer or as coculture sheets of both cell phenotypes. These three triple-layered constructs were also manufactured using mPRGF as cell sheets' support. Necrosis, glucose consumption, secretion of extracellular matrix proteins and synthesis of proangiogenic factors were determined. Histological evaluations and proteomic analyses were also performed. RESULTS The inclusion of HUVECs did not clearly improve the properties of the multilayered constructs and yet hindered their optimal conformation. The presence of mPRGF prevented the shrinkage of cell sheets, stimulated the metabolic activity and increased the matrix synthesis. At the proteome level, mPRGF conferred a dramatic advantage to the hPDLSC constructs in their ability to provide a suitable environment for tissue regeneration by inducing the expression of proteins necessary for bone morphogenesis and cellular proliferation. CONCLUSIONS hPDLSCs' triple-layer construct onto mPRGF emerges as the optimal structure for its use in regenerative therapeutics. CLINICAL RELEVANCE These results suggest the suitability of mPRGF as a promising tool to support cell sheet formation by improving their handling and biological functions.
Collapse
Affiliation(s)
- Eduardo Anitua
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain.
| | - María Troya
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Mar Zalduendo
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Roberto Tierno
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Mohammad H Alkhraisat
- BTI-Biotechnology Institute, Vitoria, Spain; University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain
| | - Nerea Osinalde
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain
| | - Asier Fullaondo
- University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Ana M Zubiaga
- University Institute for Regenerative Medicine & Oral Implantology, UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain; Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
| |
Collapse
|
35
|
Nguyen MA, Dinh NT, Do Thi MH, Nguyen Thi D, Pham UT, Tran TQ, Nguyen VM, Le NH, Nguyen DT, Pham DTN. Simple and Rapid Method of Microwell Array Fabrication for Drug Testing on 3D Cancer Spheroids. ACS OMEGA 2024; 9:16949-16958. [PMID: 38645317 PMCID: PMC11024980 DOI: 10.1021/acsomega.3c05873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/30/2023] [Accepted: 12/12/2023] [Indexed: 04/23/2024]
Abstract
Three-dimensional (3D) cell culture systems are becoming increasingly popular due to their ability to mimic the complex process of angiogenesis in cancer, providing more accurate and physiologically relevant data than traditional two-dimensional (2D) cell culture systems. Microwell systems are particularly useful in this context as they provide a microenvironment that more closely resembles the in vivo environment than traditional microwells. Poly(ethylene glycol) (PEG) microwells are particularly advantageous due to their bio-inertness and the ability to tailor their material characteristics depending on the PEG molecular weight. Although there are several methods available for microwell fabrication, most of them are time-consuming and expensive. The current study utilizes a low-cost laser etching technique on poly(methyl methacrylate) materials followed by molding with PDMS to produce microwells. The optimal conditions for making concave microwells are an engraving parameter speed of 600 mm/s, power of 20%, and a design diameter of the microwell of 0.4 mm. The artificial tumor achieved its full size after 7 days of cell growth in a microwell system, and the cells developed drugs through a live/dead assay test. The results of the drug testing revealed that the IC50 value of zerumbone-loaded liposomes in HepG2 was 4.53 pM, which is greater than the IC50 value of zerumbone. The HepG2 cancer sphere's 3D platform for medication testing revealed that zerumbone-loaded liposomes were very effective at high doses. These findings generally imply that zerumbone-loaded liposomes have the capacity to target the liver and maintain medication delivery.
Collapse
Affiliation(s)
- Mai Anh Nguyen
- Institute
for Tropical Technology, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
| | - Nhung Thi Dinh
- Hanoi
University of Science and Technology (HUST), 1 Dai Co Viet st., Hai Ba Trung
dist., Hanoi 100000, Vietnam
| | - My Hanh Do Thi
- Institute
for Tropical Technology, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
| | - Dung Nguyen Thi
- Institute
for Tropical Technology, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
| | - Uyen Thu Pham
- Institute
for Tropical Technology, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
- University
of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay
dist., Hanoi 100000, Vietnam
| | - Toan Quoc Tran
- Graduate
University of Science and Technology, Vietnam
Academy of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
- Institute
of Natural Products Chemistry, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 100000, Vietnam
| | - Vuong Minh Nguyen
- Institute
of Natural Products Chemistry, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 100000, Vietnam
| | - Nhung Hong Le
- Graduate
University of Science and Technology, Vietnam
Academy of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
- Institute
of Natural Products Chemistry, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet St., Cau Giay Dist., Hanoi 100000, Vietnam
| | - Duong Thanh Nguyen
- Institute
for Tropical Technology, Vietnam Academy
of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
- Graduate
University of Science and Technology, Vietnam
Academy of Science and Technology (VAST), 18 Hoang Quoc Viet st., Cau Giay dist., Hanoi 100000, Vietnam
| | - Dung Thuy Nguyen Pham
- NTT Institute
of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho
Chi Minh City 70000, Vietnam
- Faculty
of Environmental and Food Engineering, Nguyen
Tat Thanh University, Ho Chi Minh
City 70000, Vietnam
| |
Collapse
|
36
|
Scalzone A, Imparato G, Urciuolo F, Netti PA. Bioprinting of human dermal microtissues precursors as building blocks for endogenous in vitroconnective tissue manufacturing. Biofabrication 2024; 16:035009. [PMID: 38574552 DOI: 10.1088/1758-5090/ad3aa5] [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: 12/06/2023] [Accepted: 04/04/2024] [Indexed: 04/06/2024]
Abstract
The advent of 3D bioprinting technologies in tissue engineering has unlocked the potential to fabricatein vitrotissue models, overcoming the constraints associated with the shape limitations of preformed scaffolds. However, achieving an accurate mimicry of complex tissue microenvironments, encompassing cellular and biochemical components, and orchestrating their supramolecular assembly to form hierarchical structures while maintaining control over tissue formation, is crucial for gaining deeper insights into tissue repair and regeneration. Building upon our expertise in developing competent three-dimensional tissue equivalents (e.g. skin, gut, cervix), we established a two-step bottom-up approach involving the dynamic assembly of microtissue precursors (μTPs) to generate macroscopic functional tissue composed of cell-secreted extracellular matrix (ECM). To enhance precision and scalability, we integrated extrusion-based bioprinting technology into our established paradigm to automate, control and guide the coherent assembly ofμTPs into predefined shapes. Compared to cell-aggregated bioink, ourμTPs represent a functional unit where cells are embedded in their specific ECM.μTPs were derived from human dermal fibroblasts dynamically seeded onto gelatin-based microbeads. After 9 days,μTPs were suspended (50% v/v) in Pluronic-F127 (30% w/v) (µTP:P30), and the obtained formulation was loaded as bioink into the syringe of the Dr.INVIVO-4D6 extrusion based bioprinter.µTP:P30 bioink showed shear-thinning behavior and temperature-dependent viscosity (gel atT> 30 °C), ensuringµTPs homogenous dispersion within the gel and optimal printability. The bioprinting involved extruding several geometries (line, circle, and square) into Pluronic-F127 (40% w/v) (P40) support bath, leveraging its shear-recovery property. P40 effectively held the bioink throughout and after the bioprinting procedure, untilµTPs fused into a continuous connective tissue.µTPs fusion dynamics was studied over 8 days of culture, while the resulting endogenous construct underwent 28 days culture. Histological, immunofluorescence analysis, and second harmonic generation reconstruction revealed an increase in endogenous collagen and fibronectin production within the bioprinted construct, closely resembling the composition of the native connective tissues.
Collapse
Affiliation(s)
- Annachiara Scalzone
- Center for Advanced Biomaterials for Health Care, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples 80125, Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for Health Care, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples 80125, Italy
| | - Francesco Urciuolo
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, P.le Tecchio 80, Naples 80125, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, Naples 80125, Italy
| | - Paolo A Netti
- Center for Advanced Biomaterials for Health Care, Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples 80125, Italy
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, P.le Tecchio 80, Naples 80125, Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Napoli Federico II, P.le Tecchio 80, Naples 80125, Italy
| |
Collapse
|
37
|
Djalali-Cuevas A, Rettel M, Stein F, Savitski M, Kearns S, Kelly J, Biggs M, Skoufos I, Tzora A, Prassinos N, Diakakis N, Zeugolis DI. Macromolecular crowding in human tenocyte and skin fibroblast cultures: A comparative analysis. Mater Today Bio 2024; 25:100977. [PMID: 38322661 PMCID: PMC10846491 DOI: 10.1016/j.mtbio.2024.100977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/22/2023] [Accepted: 01/24/2024] [Indexed: 02/08/2024] Open
Abstract
Although human tenocytes and dermal fibroblasts have shown promise in tendon engineering, no tissue engineered medicine has been developed due to the prolonged ex vivo time required to develop an implantable device. Considering that macromolecular crowding has the potential to substantially accelerate the development of functional tissue facsimiles, herein we compared human tenocyte and dermal fibroblast behaviour under standard and macromolecular crowding conditions to inform future studies in tendon engineering. Basic cell function analysis made apparent the innocuousness of macromolecular crowding for both cell types. Gene expression analysis of the without macromolecular crowding groups revealed expression of tendon related molecules in human dermal fibroblasts and tenocytes. Protein electrophoresis and immunocytochemistry analyses showed significantly increased and similar deposition of collagen fibres by macromolecular crowding in the two cell types. Proteomics analysis demonstrated great similarities between human tenocyte and dermal fibroblast cultures, as well as the induction of haemostatic, anti-microbial and tissue-protective proteins by macromolecular crowding in both cell populations. Collectively, these data rationalise the use of either human dermal fibroblasts or tenocytes in combination with macromolecular crowding in tendon engineering.
Collapse
Affiliation(s)
- Adrian Djalali-Cuevas
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Mandy Rettel
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Frank Stein
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Mikhail Savitski
- Proteomics Core Facility, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | | | - Jack Kelly
- Galway University Hospital, Galway, Ireland
| | - Manus Biggs
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | - Ioannis Skoufos
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece
| | - Athina Tzora
- Laboratory of Animal Science, Nutrition and Biotechnology, School of Agriculture, University of Ioannina, Arta, Greece
| | - Nikitas Prassinos
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Diakakis
- School of Veterinary Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dimitrios I. Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
38
|
Rimal R, Muduli S, Desai P, Marquez AB, Möller M, Platzman I, Spatz J, Singh S. Vascularized 3D Human Skin Models in the Forefront of Dermatological Research. Adv Healthc Mater 2024; 13:e2303351. [PMID: 38277705 PMCID: PMC11468127 DOI: 10.1002/adhm.202303351] [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: 10/02/2023] [Revised: 12/04/2023] [Indexed: 01/28/2024]
Abstract
In vitro engineered skin models are emerging as an alternative platform to reduce and replace animal testing in dermatological research. Despite the progress made in recent years, considerable challenges still exist for the inclusion of diverse cell types within skin models. Blood vessels, in particular, are essential in maintaining tissue homeostasis and are one of many primary contributors to skin disease inception and progression. Substantial efforts in the past have allowed the successful fabrication of vascularized skin models that are currently utilized for disease modeling and drugs/cosmetics testing. This review first discusses the need for vascularization within tissue-engineered skin models, highlighting their role in skin grafting and disease pathophysiology. Second, the review spotlights the milestones and recent progress in the fabrication and utilization of vascularized skin models. Additionally, advances including the use of bioreactors, organ-on-a-chip devices, and organoid systems are briefly explored. Finally, the challenges and future outlook for vascularized skin models are addressed.
Collapse
Affiliation(s)
- Rahul Rimal
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Saradaprasan Muduli
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
| | - Prachi Desai
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Andrea Bonnin Marquez
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Martin Möller
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Ilia Platzman
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
| | - Joachim Spatz
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
- Max Planck School Matter to LifeJahnstrasse 2969120HeidelbergGermany
| | - Smriti Singh
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
| |
Collapse
|
39
|
Yang H, Zheng M, Zhang Y, Li C, Lai JHC, Zhang Q, Chan KWY, Wang H, Zhao X, Yang Z, Xu C. Enhanced angiogenesis in porous poly(ε-caprolactone) scaffolds fortified with methacrylated hyaluronic acid hydrogel after subcutaneous transplantation. BIOMATERIALS TRANSLATIONAL 2024; 5:59-68. [PMID: 39220667 PMCID: PMC11362355 DOI: 10.12336/biomatertransl.2024.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/20/2024] [Accepted: 03/08/2024] [Indexed: 09/04/2024]
Abstract
A composite scaffold composed of a porous scaffold and hydrogel filling can facilitate engraftment, survival, and retention in cell transplantation processes. This study presents a composite scaffold made of poly(ε-caprolactone) (PCL) and methacrylated hyaluronic acid (MeHA) hydrogel and describes the corresponding physical properties (surface area, porosity, and mechanical strength) and host response (angiogenesis and fibrosis) after subcutaneous transplantation. Specifically, we synthesise MeHA with different degrees of substitution and fabricate a PCL scaffold with different porosities. Subsequently, we construct a series of PCL/MeHA composite scaffolds by combining these hydrogels and scaffolds. In experiments with mice, the scaffold composed of 3% PCL and 10-100 kDa, degree of substitution 70% MeHA results in the least fibrosis and a higher degree of angiogenesis. This study highlights the potential of PCL/MeHA composite scaffolds for subcutaneous cell transplantation, given their desirable physical properties and host response.
Collapse
Affiliation(s)
- Huaxin Yang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Mengjia Zheng
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yuyue Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Chaochang Li
- Center for Pluripotent Stem Cell Research and Engineering, Research Institute of Tsinghua, Guangzhou, Guangdong Province, China
| | - Joseph Ho Chi Lai
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Qizheng Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kannie WY Chan
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Hao Wang
- Center for Pluripotent Stem Cell Research and Engineering, Research Institute of Tsinghua, Guangzhou, Guangdong Province, China
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| | - Zijiang Yang
- Center for Pluripotent Stem Cell Research and Engineering, Research Institute of Tsinghua, Guangzhou, Guangdong Province, China
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
40
|
Bodio C, Milesi A, Lonati PA, Chighizola CB, Mauro A, Pradotto LG, Meroni PL, Borghi MO, Raschi E. Fibroblasts and Endothelial Cells in Three-Dimensional Models: A New Tool for Addressing the Pathogenesis of Systemic Sclerosis as a Prototype of Fibrotic Vasculopathies. Int J Mol Sci 2024; 25:2780. [PMID: 38474040 DOI: 10.3390/ijms25052780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
Two-dimensional in vitro cultures have represented a milestone in biomedical and pharmacological research. However, they cannot replicate the architecture and interactions of in vivo tissues. Moreover, ethical issues regarding the use of animals have triggered strategies alternative to animal models. The development of three-dimensional (3D) models offers a relevant tool to investigate disease pathogenesis and treatment, modeling in vitro the in vivo environment. We aimed to develop a dynamic 3D in vitro model for culturing human endothelial cells (ECs) and skin fibroblasts, simulating the structure of the tissues mainly affected in systemic sclerosis (SSc), a prototypical autoimmune fibrotic vasculopathy. Dermal fibroblasts and umbilical vein ECs grown in scaffold or hydrogel, respectively, were housed in bioreactors under flow. Fibroblasts formed a tissue-like texture with the deposition of a new extracellular matrix (ECM) and ECs assembled tube-shaped structures with cell polarization. The fine-tuned dynamic modular system allowing 3D fibroblast/EC culture connection represents a valuable model of the in vivo interplay between the main players in fibrotic vasculopathy as SSc. This model can lead to a more accurate study of the disease's pathogenesis, avoiding the use of animals, and to the development of novel therapies, possibly resulting in improved patient management.
Collapse
Affiliation(s)
- Caterina Bodio
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Auxologico Italiano, 20095 Cusano Milanino, Italy
| | - Alessandra Milesi
- Laboratory of Clinical Neurobiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Auxologico Italiano, 28824 Piancavallo, Italy
| | - Paola Adele Lonati
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Auxologico Italiano, 20095 Cusano Milanino, Italy
| | - Cecilia Beatrice Chighizola
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
- U.O.C. Clinica Reumatologica Pediatrica, ASST G. Pini-CTO, 20122 Milan, Italy
| | - Alessandro Mauro
- Laboratory of Clinical Neurobiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Auxologico Italiano, 28824 Piancavallo, Italy
- Department of Neuroscience, University of Turin, 10124 Turin, Italy
| | - Luca Guglielmo Pradotto
- Laboratory of Clinical Neurobiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Auxologico Italiano, 28824 Piancavallo, Italy
- Department of Neuroscience, University of Turin, 10124 Turin, Italy
| | - Pier Luigi Meroni
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Auxologico Italiano, 20095 Cusano Milanino, Italy
| | - Maria Orietta Borghi
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Auxologico Italiano, 20095 Cusano Milanino, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Elena Raschi
- Experimental Laboratory of Immunological and Rheumatologic Researches, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Auxologico Italiano, 20095 Cusano Milanino, Italy
| |
Collapse
|
41
|
Purbantoro SD, Taephatthanasagon T, Purwaningrum M, Hirankanokchot T, Peralta S, Fiani N, Sawangmake C, Rattanapuchpong S. Trends of regenerative tissue engineering for oral and maxillofacial reconstruction in veterinary medicine. Front Vet Sci 2024; 11:1325559. [PMID: 38450027 PMCID: PMC10915013 DOI: 10.3389/fvets.2024.1325559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 02/05/2024] [Indexed: 03/08/2024] Open
Abstract
Oral and maxillofacial (OMF) defects are not limited to humans and are often encountered in other species. Reconstructing significant tissue defects requires an excellent strategy for efficient and cost-effective treatment. In this regard, tissue engineering comprising stem cells, scaffolds, and signaling molecules is emerging as an innovative approach to treating OMF defects in veterinary patients. This review presents a comprehensive overview of OMF defects and tissue engineering principles to establish proper treatment and achieve both hard and soft tissue regeneration in veterinary practice. Moreover, bench-to-bedside future opportunities and challenges of tissue engineering usage are also addressed in this literature review.
Collapse
Affiliation(s)
- Steven Dwi Purbantoro
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Teeanutree Taephatthanasagon
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Medania Purwaningrum
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Department of Biochemistry, Faculty of Veterinary Medicine, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Thanyathorn Hirankanokchot
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Santiago Peralta
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Nadine Fiani
- Department of Clinical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Chenphop Sawangmake
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Sirirat Rattanapuchpong
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Veterinary Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
- Academic Affairs, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
42
|
Chen W, Nie M, Gan J, Xia N, Wang D, Sun L. Tailoring cell sheets for biomedical applications. SMART MEDICINE 2024; 3:e20230038. [PMID: 39188516 PMCID: PMC11235941 DOI: 10.1002/smmd.20230038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/04/2024] [Indexed: 08/28/2024]
Abstract
Cell sheet technology has emerged as a novel scaffold-free approach for cell-based therapies in regenerative medicine. Techniques for harvesting cell sheets are essential to preserve the integrity of living cell sheets. This review provides an overview of fundamental technologies to fabricate cell sheets and recent advances in cell sheet-based tissue engineering. In addition to the commonly used temperature-responsive systems, we introduce alternative approaches, such as ROS-induced, magnetic-controlled, and light-induced cell sheet technologies. Moreover, we discuss the modification of the cell sheet to improve its function, including stacking, genetic modification, and vascularization. With the significant advances in cell sheet technology, cell sheets have been widely applied in various tissues and organs, including but not limited to the lung, cornea, cartilage, periodontium, heart, and liver. This review further describes both the preclinical and clinical applications of cell sheets. We believe that the progress in cell sheet technology would further propel its biomedical applications.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| | - Min Nie
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| | - Jingjing Gan
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| | - Nan Xia
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| | - Dandan Wang
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
| | - Lingyun Sun
- Department of Rheumatology and ImmunologyNanjing Drum Tower HospitalMedical SchoolNanjing UniversityNanjingChina
- Department of Rheumatology and ImmunologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| |
Collapse
|
43
|
Santa Cruz-Pavlovich FJ, Bolaños-Chang AJ, Del Rio-Murillo XI, Aranda-Preciado GA, Razura-Ruiz EM, Santos A, Navarro-Partida J. Beyond Vision: An Overview of Regenerative Medicine and Its Current Applications in Ophthalmological Care. Cells 2024; 13:179. [PMID: 38247870 PMCID: PMC10814238 DOI: 10.3390/cells13020179] [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: 12/05/2023] [Revised: 12/23/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
Regenerative medicine (RM) has emerged as a promising and revolutionary solution to address a range of unmet needs in healthcare, including ophthalmology. Moreover, RM takes advantage of the body's innate ability to repair and replace pathologically affected tissues. On the other hand, despite its immense promise, RM faces challenges such as ethical concerns, host-related immune responses, and the need for additional scientific validation, among others. The primary aim of this review is to present a high-level overview of current strategies in the domain of RM (cell therapy, exosomes, scaffolds, in vivo reprogramming, organoids, and interspecies chimerism), centering around the field of ophthalmology. A search conducted on clinicaltrials.gov unveiled a total of at least 209 interventional trials related to RM within the ophthalmological field. Among these trials, there were numerous early-phase studies, including phase I, I/II, II, II/III, and III trials. Many of these studies demonstrate potential in addressing previously challenging and degenerative eye conditions, spanning from posterior segment pathologies like Age-related Macular Degeneration and Retinitis Pigmentosa to anterior structure diseases such as Dry Eye Disease and Limbal Stem Cell Deficiency. Notably, these therapeutic approaches offer tailored solutions specific to the underlying causes of each pathology, thus allowing for the hopeful possibility of bringing forth a treatment for ocular diseases that previously seemed incurable and significantly enhancing patients' quality of life. As advancements in research and technology continue to unfold, future objectives should focus on ensuring the safety and prolonged viability of transplanted cells, devising efficient delivery techniques, etc.
Collapse
Affiliation(s)
- Francisco J. Santa Cruz-Pavlovich
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Mexico; (F.J.S.C.-P.); (A.J.B.-C.); (X.I.D.R.-M.); (E.M.R.-R.); (A.S.)
| | - Andres J. Bolaños-Chang
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Mexico; (F.J.S.C.-P.); (A.J.B.-C.); (X.I.D.R.-M.); (E.M.R.-R.); (A.S.)
| | - Ximena I. Del Rio-Murillo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Mexico; (F.J.S.C.-P.); (A.J.B.-C.); (X.I.D.R.-M.); (E.M.R.-R.); (A.S.)
| | | | - Esmeralda M. Razura-Ruiz
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Mexico; (F.J.S.C.-P.); (A.J.B.-C.); (X.I.D.R.-M.); (E.M.R.-R.); (A.S.)
| | - Arturo Santos
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Mexico; (F.J.S.C.-P.); (A.J.B.-C.); (X.I.D.R.-M.); (E.M.R.-R.); (A.S.)
| | - Jose Navarro-Partida
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64849, Mexico; (F.J.S.C.-P.); (A.J.B.-C.); (X.I.D.R.-M.); (E.M.R.-R.); (A.S.)
| |
Collapse
|
44
|
Bhattacharya A, Alam K, Roy NS, Kaur K, Kaity S, Ravichandiran V, Roy S. Exploring the interaction between extracellular matrix components in a 3D organoid disease model to replicate the pathophysiology of breast cancer. J Exp Clin Cancer Res 2023; 42:343. [PMID: 38102637 PMCID: PMC10724947 DOI: 10.1186/s13046-023-02926-4] [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: 10/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
In vitro models are necessary to study the pathophysiology of the disease and the development of effective, tailored treatment methods owing to the complexity and heterogeneity of breast cancer and the large population affected by it. The cellular connections and tumor microenvironments observed in vivo are often not recapitulated in conventional two-dimensional (2D) cell cultures. Therefore, developing 3D in vitro models that mimic the complex architecture and physiological circumstances of breast tumors is crucial for advancing our understanding of the illness. A 3D scaffold-free in vitro disease model mimics breast cancer pathophysiology by allowing cells to self-assemble/pattern into 3D structures, in contrast with other 3D models that rely on artificial scaffolds. It is possible that this model, whether applied to breast tumors using patient-derived primary cells (fibroblasts, endothelial cells, and cancer cells), can accurately replicate the observed heterogeneity. The complicated interactions between different cell types are modelled by integrating critical components of the tumor microenvironment, such as the extracellular matrix, vascular endothelial cells, and tumor growth factors. Tissue interactions, immune cell infiltration, and the effects of the milieu on drug resistance can be studied using this scaffold-free 3D model. The scaffold-free 3D in vitro disease model for mimicking tumor pathophysiology in breast cancer is a useful tool for studying the molecular basis of the disease, identifying new therapeutic targets, and evaluating treatment modalities. It provides a more physiologically appropriate high-throughput platform for screening large compound library in a 96-384 well format. We critically discussed the rapid development of personalized treatment strategies and accelerated drug screening platforms to close the gap between traditional 2D cell culture and in vivo investigations.
Collapse
Affiliation(s)
- Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
45
|
Nikdouz A, Orso F. Emerging roles of 3D-culture systems in tackling tumor drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2023; 6:788-804. [PMID: 38263982 PMCID: PMC10804388 DOI: 10.20517/cdr.2023.93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/01/2023] [Accepted: 11/14/2023] [Indexed: 01/25/2024]
Abstract
Drug resistance that affects patients universally is a major challenge in cancer therapy. The development of drug resistance in cancer cells is a multifactor event, and its process involves numerous mechanisms that allow these cells to evade the effect of treatments. As a result, the need to understand the molecular mechanisms underlying cancer drug sensitivity is imperative. Traditional 2D cell culture systems have been utilized to study drug resistance, but they often fail to mimic the 3D milieu and the architecture of real tissues and cell-cell interactions. As a result of this, 3D cell culture systems are now considered a comprehensive model to study drug resistance in vitro. Cancer cells exhibit an in vivo behavior when grown in a three-dimensional environment and react to therapy more physiologically. In this review, we discuss the relevance of main 3D culture systems in the study of potential approaches to overcome drug resistance and in the identification of personalized drug targets with the aim of developing patient-specific treatment strategies that can be put in place when resistance emerges.
Collapse
Affiliation(s)
| | - Francesca Orso
- Department of Translational Medicine, University of Eastern Piedmont, Novara 28100, Italy
| |
Collapse
|
46
|
Du S, Elliman SJ, Zeugolis DI, O'Brien T. Carrageenan as a macromolecular crowding agent in human umbilical cord derived mesenchymal stromal cell culture. Int J Biol Macromol 2023; 251:126353. [PMID: 37591431 DOI: 10.1016/j.ijbiomac.2023.126353] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
Cell sheet tissue engineering requires prolonged in vitro culture for the development of implantable devices. Unfortunately, lengthy in vitro culture is associated with cell phenotype loss and substantially higher cost of goods, which collectively hinder clinical translation and commercialisation of tissue engineered medicines. Although macromolecular crowding has been shown to enhance and accelerate extracellular matrix deposition, whilst maintaining cellular phenotype, the optimal macromolecular crowding agent still remains elusive. Herein, we evaluated the biophysical properties of seven different carrageenan molecules at five different concentrations and their effect on human umbilical cord-derived mesenchymal stromal cell morphology, viability, metabolic activity, proliferation, extracellular matrix deposition and surface marker expression. All types of carrageenan (CR) assessed demonstrated a hydrodynamic radius increase as a function of increasing concentration; high polydispersity; and negative charge. Two iota CRs were excluded from further analysis due to poor solubility in cell culture. Among the remaining five carrageenans, the lambda medium viscosity type at concentrations of 10 and 50 μg/ml did not affect cell morphology, viability, metabolic activity, proliferation and expression of surface markers and significantly increased the deposition of collagen types I, III and IV, fibronectin and laminin. Our data highlight the potential of lambda medium viscosity carrageenan as a macromolecular crowding agent for the accelerated development of functional tissue engineered medicines.
Collapse
Affiliation(s)
- Shanshan Du
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland
| | | | - Dimitrios I Zeugolis
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland; Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, University of Galway, Galway, Ireland; Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research, School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute (REMEDI), Biomedical Sciences Building, University of Galway, Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, University of Galway, Galway, Ireland; Orbsen Therapeutics Ltd, IDA Business Park, Dangan, Galway, Ireland.
| |
Collapse
|
47
|
Mahajan A, Nengroo MA, Datta D, Katti DS. Converse modulation of Wnt/β-catenin signaling during expansion and differentiation phases of Infrapatellar fat pad-derived MSCs for improved engineering of hyaline cartilage. Biomaterials 2023; 302:122296. [PMID: 37696204 DOI: 10.1016/j.biomaterials.2023.122296] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 08/14/2023] [Accepted: 08/25/2023] [Indexed: 09/13/2023]
Abstract
Mesenchymal stem cells (MSCs) are potential candidates in cell-based therapy for cartilage repair and regeneration. However, during chondrogenic differentiation, MSCs undergo undesirable hypertrophic maturation. This poses a risk of ossification in the neo-tissue formed that eventually impedes the clinical use of MSCs for cartilage repair. TGF-β is a potent growth factor used for chondrogenic differentiation of MSCs, however, its role in hypertrophy remains ambiguous. In the present work, we decipher that TGF-β activates Wnt/β-catenin signaling through SMAD3 and increases the propensity of Infrapatellar fat pad derived MSCs (IFP-MSCs) towards hypertrophy. Notably, inhibiting TGF-β induced Wnt/β-catenin signaling suppresses hypertrophic progression and enhances chondrogenic ability of IFP-MSCs in plasma hydrogels. Additionally, we demonstrate that activating Wnt signaling during expansion phase, promotes proliferation and reduces senescence, while improving stemness of IFP-MSCs. Thus, conversely modulating Wnt signaling in vitro during expansion and differentiation phases generates hyaline-like cartilage with minimal hypertrophy. Importantly, pre-treatment of IFP-MSCs encapsulated in plasma hydrogel with Wnt modulators followed by subcutaneous implantation in nude mice resulted in formation of a cartilage tissue with negligible calcification. Overall, this study provides technological advancement on targeting Wnt/β-catenin pathway in a 3D scaffold, while maintaining the standard chondro-induction protocol to overcome the challenges associated with the clinical use of MSCs to engineer hyaline cartilage.
Collapse
Affiliation(s)
- Aman Mahajan
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology-Kanpur, Kanpur, 208016, Uttar Pradesh, India; The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology-Kanpur, Kanpur, 208016, Uttar Pradesh, India
| | - Mushtaq A Nengroo
- Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Dipak Datta
- Cancer Biology Division, CSIR-Central Drug Research Institute, Lucknow, 226031, Uttar Pradesh, India
| | - Dhirendra S Katti
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology-Kanpur, Kanpur, 208016, Uttar Pradesh, India; The Mehta Family Centre for Engineering in Medicine, Indian Institute of Technology-Kanpur, Kanpur, 208016, Uttar Pradesh, India.
| |
Collapse
|
48
|
Ota T, Takao T, Iwai R, Moriwaki T, Kitaguchi Y, Fujisawa Y, Yamada D, Kimata Y, Takarada T. Fabrication of shape-designable cartilage from human induced pluripotent stem cell-derived chondroprogenitors using a cell self-aggregation technique. Biomed Mater 2023; 18:065019. [PMID: 37827163 DOI: 10.1088/1748-605x/ad02d1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/12/2023] [Indexed: 10/14/2023]
Abstract
With the advancement of tissue engineering technologies, implantable materials have been developed for use in facial plastic surgery, including auriculoplasty and rhinoplasty. Tissue-engineered cartilage comprising only cells and cell-produced extracellular matrix is considered valuable as there is no need to consider problems associated with scaffold absorption or immune responses commonly related to conventional artificial materials. However, it is exceedingly difficult to produce large-sized complex shapes of cartilage without the use of scaffolds. In this study, we describe the production of shape-designable cartilage using a novel cell self-aggregation technique (CAT) and chondroprogenitor cells derived from human induced pluripotent stem cells as the source. The method described does not require special equipment such as bio-3D printers, and the produced tissue can be induced into well-matured cartilage with abundant cartilage matrixin vitro. Using CAT, we were able to generate cartilage in the form of rings or tubes with adjustable inner diameter and curvature, over a range of several centimeters, without the use of scaffolds. Thein vitrofabrication of shape-designable cartilage using CAT is a promising development in facial plastic surgery.
Collapse
Affiliation(s)
- Tomoyuki Ota
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
- Department of Plastic and Reconstructive Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Tomoka Takao
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Ryosuke Iwai
- Okayama University of Science, Institute of Frontier Science and Technology, 1-1, Ridai-cho, Kita-ku, Okayama-shi, Okayama 700-0005, Japan
| | - Takeshi Moriwaki
- Department of Mechanical Science and Engineering, Faculty of Science and Technology, Hirosaki University, 3, Bunkyo-cho, Hirosaki-shi, Aomori 036-8561, Japan
| | - Yohei Kitaguchi
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
- Department of Plastic and Reconstructive Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Yuki Fujisawa
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Daisuke Yamada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Yoshihiro Kimata
- Department of Plastic and Reconstructive Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1, Shikata-cho, Kita-ku, Okayama-shi, Okayama 700-8558, Japan
| |
Collapse
|
49
|
Roman V, Mihaila M, Radu N, Marineata S, Diaconu CC, Bostan M. Cell Culture Model Evolution and Its Impact on Improving Therapy Efficiency in Lung Cancer. Cancers (Basel) 2023; 15:4996. [PMID: 37894363 PMCID: PMC10605536 DOI: 10.3390/cancers15204996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Optimizing cell culture conditions is essential to ensure experimental reproducibility. To improve the accuracy of preclinical predictions about the response of tumor cells to different classes of drugs, researchers have used 2D or 3D cell cultures in vitro to mimic the cellular processes occurring in vivo. While 2D cell culture provides valuable information on how therapeutic agents act on tumor cells, it cannot quantify how the tumor microenvironment influences the response to therapy. This review presents the necessary strategies for transitioning from 2D to 3D cell cultures, which have facilitated the rapid evolution of bioengineering techniques, leading to the development of microfluidic technology, including organ-on-chip and tumor-on-chip devices. Additionally, the study aims to highlight the impact of the advent of 3D bioprinting and microfluidic technology and their implications for improving cancer treatment and approaching personalized therapy, especially for lung cancer. Furthermore, implementing microfluidic technology in cancer studies can generate a series of challenges and future perspectives that lead to the discovery of new predictive markers or targets for antitumor treatment.
Collapse
Affiliation(s)
- Viviana Roman
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (V.R.); (M.B.)
| | - Mirela Mihaila
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (V.R.); (M.B.)
| | - Nicoleta Radu
- Department of Biotechnology, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 011464 Bucharest, Romania
- Biotechnology Department, National Institute for Chemistry and Petrochemistry R&D of Bucharest, 060021 Bucharest, Romania
| | - Stefania Marineata
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila, 050471 Bucharest, Romania;
| | - Carmen Cristina Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, 030304 Bucharest, Romania;
| | - Marinela Bostan
- Center of Immunology, Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (V.R.); (M.B.)
- Department of Immunology, ‘Victor Babeș’ National Institute of Pathology, 050096 Bucharest, Romania
| |
Collapse
|
50
|
Noh S, Jin YJ, Shin DI, Kwon HJ, Yun HW, Kim KM, Park JY, Chung JY, Park DY. Selective Extracellular Matrix Guided Mesenchymal Stem Cell Self-Aggregate Engineering for Replication of Meniscal Zonal Tissue Gradient in a Porcine Meniscectomy Model. Adv Healthc Mater 2023; 12:e2301180. [PMID: 37463568 DOI: 10.1002/adhm.202301180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/13/2023] [Accepted: 07/14/2023] [Indexed: 07/20/2023]
Abstract
Degenerative meniscus tears (DMTs) are prevalent findings in osteoarthritic knees, yet current treatment is mostly limited to arthroscopic partial meniscectomy rather than regeneration, which further exacerbates arthritic changes. Translational research regarding meniscus regeneration is hindered by the complex, composite nature of the meniscus which exhibit a gradient from inner cartilage-like tissue to outer fibrous tissue, as well as engineering hurdles often requiring growth factors and cross-linking agents. Here, a meniscus zonal tissue gradient is proposed using zone-specific decellularized meniscus extracellular matrix (DMECM) and autologous synovial mesenchymal stem cells (SMSC) via self-aggregation without the use of growth factors or cross-linking agents. Combination with zone-specific DMECM during self-aggregation of MSCs forms zone-specific meniscus tissue that reflects the respective DMECM harvest site. The implantation of these constructs leads to the regeneration of meniscus tissue resembling the native meniscus, demonstrating inner cartilaginous and outer fibrous characteristics as well as recovery of native meniscal microarchitecture in a porcine partial meniscectomy model at 6 months. In all, the findings offer a potential regenerative therapy for DMTs that may improve current partial meniscectomy-based patient care.
Collapse
Affiliation(s)
- Sujin Noh
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, 16499, Republic of Korea
| | - Yong Jun Jin
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Dong Il Shin
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Hyeon Jae Kwon
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Hee-Woong Yun
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Republic of Korea
| | - Kyu Min Kim
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Republic of Korea
| | - Jae-Young Park
- Department of Orthopedics Surgery, CHA University Bundang Medical Center, Bundang-gu, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Jun Young Chung
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Do Young Park
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, 16499, Republic of Korea
- Department of Orthopedic Surgery, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
- Cell Therapy Center, Ajou Medical Center, Suwon, 16499, Republic of Korea
- Ajou University, Leading Convergence of Healthcare and Medicine, Institute of Science & Technology (ALCHeMIST), Suwon, 16499, Republic of Korea
| |
Collapse
|