1
|
Cao L, Liu Y, Lin G. Strategies for Altering Delivery Technologies to Optimize CAR Therapy. Int J Mol Sci 2025; 26:3206. [PMID: 40244018 PMCID: PMC11989270 DOI: 10.3390/ijms26073206] [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: 02/05/2025] [Revised: 03/22/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has been proven to be an effective strategy for the treatment of hematological malignancies. At present, how to prepare CAR-T cells efficiently, quickly, and safely is one of the urgent problems to be solved. The durability and activity of engineered T cells in solid tumors need to be further improved, and the strategy of T cells penetrating the tumor microenvironment also needs to be improved. In addition, although the problems mainly caused by T-cell biology are being solved, the manufacturing mode and process still need to be improved to ensure that CAR-T cell therapy can be widely used. This paper summarizes some strategies that can improve the efficacy of CAR-T cells.
Collapse
Affiliation(s)
- Lili Cao
- Student Counseling Center, Shandong University, Jinan 250012, China;
| | - Yingying Liu
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China;
| | - Guimei Lin
- School of Pharmaceutical Science, Shandong University, Jinan 250012, China;
| |
Collapse
|
2
|
Yadav K, Ebenezer Gnanakani SP, Kumar Sahu K, Sucheta, Dubey A, Minz S, Raza W, Pradhan M. Unleashing the potential of natural protein based nanoparticles for the delivery of therapeutic nucleic Acid: A comprehensive review. Int J Pharm 2025; 669:125049. [PMID: 39674384 DOI: 10.1016/j.ijpharm.2024.125049] [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: 05/14/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
Nucleic acid-based therapeutics represent a revolutionary approach in treating genetic disorders, offering unprecedented potential for addressing pathologies at their molecular level. However, effective cellular delivery remains a critical challenge hindering their clinical implementation. While existing delivery systems, including viral vectors and lipid nanoparticles, have shown utility, they face limitations in immunogenicity, cargo capacity, and manufacturing complexity. Natural protein-based nanoparticles, derived from proteins such as albumin, ferritin, and elastin, have emerged as promising alternative delivery systems. These carriers offer distinct advantages including reduced immunogenicity, enhanced biocompatibility, and optimal biodegradation profiles. Their engineerable nature enables precise control over particle size, surface charge, and ligand conjugation, facilitating selective cellular targeting and improved pharmacokinetics. Recent technological advances have expanded the application of protein nanoparticles across various nucleic acid modalities, including mRNA, siRNA, and plasmid DNA. Extensive research has characterized these systems through rigorous in vitro and in vivo studies, advancing our understanding of their biological behavior and clinical potential. Advanced engineering methodologies have further enhanced their optimization for specific therapeutic applications. This review examines the development and potential of protein-based nanoparticles in nucleic acid delivery, highlighting their advantages and addressing current challenges. By analyzing recent advances and clinical progress, we underscore their significant potential to enhance the safety, specificity, and efficacy of nucleic acid therapeutics, potentially revolutionizing the treatment of genetic disorders.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kurud Road, Kohka, Bhilai 490024, Chhattisgarh, India
| | - S Princely Ebenezer Gnanakani
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Limda, Waghodia, Vadodara, Gujarat 391760, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangaluru 575018, Karnataka, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | - Wasim Raza
- Central Laboratory Facility, Chhattisgarh Council of Science and Technology, Vigyan Bhawan, Raipur, Chhattisgarh, India
| | | |
Collapse
|
3
|
Nair R, Kasturi M, Mathur V, Seetharam RN, S Vasanthan K. Strategies for developing 3D printed ovarian model for restoring fertility. Clin Transl Sci 2024; 17:e13863. [PMID: 38955776 PMCID: PMC11219245 DOI: 10.1111/cts.13863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 05/16/2024] [Accepted: 05/31/2024] [Indexed: 07/04/2024] Open
Abstract
Ovaries play a crucial role in the regulation of numerous essential processes that occur within the intricate framework of female physiology. They are entrusted with the responsibility of both generating a new life and orchestrating a delicate hormonal symphony. Understanding their functioning is crucial for gaining insight into the complexities of reproduction, health, and fertility. In addition, ovaries secrete hormones that are crucial for both secondary sexual characteristics and the maintenance of overall health. A three-dimensional (3D) prosthetic ovary has the potential to restore ovarian function and preserve fertility in younger females who have undergone ovariectomies or are afflicted with ovarian malfunction. Clinical studies have not yet commenced, and the production of 3D ovarian tissue for human implantation is still in the research phase. The main challenges faced while creating a 3D ovary for in vivo implantation include sustenance of ovarian follicles, achieving vascular infiltration into the host tissue, and restoring hormone circulation. The complex ovarian microenvironment that is compartmentalized and rigid makes the biomimicking of the 3D ovary challenging in terms of biomaterial selection and bioink composition. The successful restoration of these properties in animal models has led to expectations for the development of human ovaries for implantation. This review article summarizes and evaluates the optimal 3D models of ovarian structures and their safety and efficacy concerns to provide concrete suggestions for future research.
Collapse
Affiliation(s)
- Ramya Nair
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Meghana Kasturi
- Department of Mechanical EngineeringUniversity of MichiganDearbornMichiganUSA
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Raviraja N. Seetharam
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher EducationManipalKarnatakaIndia
| |
Collapse
|
4
|
Quek J, Vizetto-Duarte C, Teoh SH, Choo Y. Towards Stem Cell Therapy for Critical-Sized Segmental Bone Defects: Current Trends and Challenges on the Path to Clinical Translation. J Funct Biomater 2024; 15:145. [PMID: 38921519 PMCID: PMC11205181 DOI: 10.3390/jfb15060145] [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: 04/24/2024] [Revised: 05/18/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
The management and reconstruction of critical-sized segmental bone defects remain a major clinical challenge for orthopaedic clinicians and surgeons. In particular, regenerative medicine approaches that involve incorporating stem cells within tissue engineering scaffolds have great promise for fracture management. This narrative review focuses on the primary components of bone tissue engineering-stem cells, scaffolds, the microenvironment, and vascularisation-addressing current advances and translational and regulatory challenges in the current landscape of stem cell therapy for critical-sized bone defects. To comprehensively explore this research area and offer insights for future treatment options in orthopaedic surgery, we have examined the latest developments and advancements in bone tissue engineering, focusing on those of clinical relevance in recent years. Finally, we present a forward-looking perspective on using stem cells in bone tissue engineering for critical-sized segmental bone defects.
Collapse
Affiliation(s)
- Jolene Quek
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| | - Catarina Vizetto-Duarte
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| | - Swee Hin Teoh
- Centre for Advanced Medical Engineering, College of Materials Science and Engineering, Hunan University, Changsha 410012, China
| | - Yen Choo
- Developmental Biology and Regenerative Medicine Programme, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore; (J.Q.); (C.V.-D.)
| |
Collapse
|
5
|
Liu Y, Liu H, Guo S, Qi J, Zhang R, Liu X, Sun L, Zong M, Cheng H, Wu X, Li B. Applications of Bacterial Cellulose-Based Composite Materials in Hard Tissue Regenerative Medicine. Tissue Eng Regen Med 2023; 20:1017-1039. [PMID: 37688748 PMCID: PMC10645761 DOI: 10.1007/s13770-023-00575-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Cartilage, bone, and teeth, as the three primary hard tissues in the human body, have a significant application value in maintaining physical and mental health. Since the development of bacterial cellulose-based composite materials with excellent biomechanical strength and good biocompatibility, bacterial cellulose-based composites have been widely studied in hard tissue regenerative medicine. This paper provides an overview of the advantages of bacterial cellulose-based for hard tissue regeneration and reviews the recent progress in the preparation and research of bacterial cellulose-based composites in maxillofacial cartilage, dentistry, and bone. METHOD A systematic review was performed by searching the PubMed and Web of Science databases using selected keywords and Medical Subject Headings search terms. RESULTS Ideal hard tissue regenerative medicine materials should be biocompatible, biodegradable, non-toxic, easy to use, and not burdensome to the human body; In addition, they should have good plasticity and processability and can be prepared into materials of different shapes; In addition, it should have good biological activity, promoting cell proliferation and regeneration. Bacterial cellulose materials have corresponding advantages and disadvantages due to their inherent properties. However, after being combined with other materials (natural/ synthetic materials) to form composite materials, they basically meet the requirements of hard tissue regenerative medicine materials. We believe that it is worth being widely promoted in clinical applications in the future. CONCLUSION Bacterial cellulose-based composites hold great promise for clinical applications in hard tissue engineering. However, there are still several challenges that need to be addressed. Further research is needed to incorporate multiple disciplines and advance biological tissue engineering techniques. By enhancing the adhesion of materials to osteoblasts, providing cell stress stimulation through materials, and introducing controlled release systems into matrix materials, the practical application of bacterial cellulose-based composites in clinical settings will become more feasible in the near future.
Collapse
Affiliation(s)
- Yingyu Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Haiyan Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Susu Guo
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Jin Qi
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Ran Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Xiaoming Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Lingxiang Sun
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Mingrui Zong
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Huaiyi Cheng
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China
| | - Xiuping Wu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China.
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China.
| | - Bing Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China.
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
6
|
Liu H, Chen H, Han Q, Sun B, Liu Y, Zhang A, Fan D, Xia P, Wang J. Recent advancement in vascularized tissue-engineered bone based on materials design and modification. Mater Today Bio 2023; 23:100858. [PMID: 38024843 PMCID: PMC10679779 DOI: 10.1016/j.mtbio.2023.100858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/03/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023] Open
Abstract
Bone is one of the most vascular network-rich tissues in the body and the vascular system is essential for the development, homeostasis, and regeneration of bone. When segmental irreversible damage occurs to the bone, restoring its vascular system by means other than autogenous bone grafts with vascular pedicles is a therapeutic challenge. By pre-generating the vascular network of the scaffold in vivo or in vitro, the pre-vascularization technique enables an abundant blood supply in the scaffold after implantation. However, pre-vascularization techniques are time-consuming, and in vivo pre-vascularization techniques can be damaging to the body. Critical bone deficiencies may be filled quickly with immediate implantation of a supporting bone tissue engineered scaffold. However, bone tissue engineered scaffolds generally lack vascularization, which requires modification of the scaffold to aid in enhancing internal vascularization. In this review, we summarize the relationship between the vascular system and osteogenesis and use it as a basis to further discuss surgical and cytotechnology-based pre-vascularization strategies and to describe the preparation of vascularized bone tissue engineered scaffolds that can be implanted immediately. We anticipate that this study will serve as inspiration for future vascularized bone tissue engineered scaffold construction and will aid in the achievement of clinical vascularized bone.
Collapse
Affiliation(s)
- Hao Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Hao Chen
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Qin Han
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Bin Sun
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Yang Liu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Aobo Zhang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Danyang Fan
- Department of Dermatology, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Peng Xia
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| | - Jincheng Wang
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun 130000, Jilin, China
| |
Collapse
|
7
|
Abalymov A, Pinchasik BE, Akasov RA, Lomova M, Parakhonskiy BV. Strategies for Anisotropic Fibrillar Hydrogels: Design, Cell Alignment, and Applications in Tissue Engineering. Biomacromolecules 2023; 24:4532-4552. [PMID: 37812143 DOI: 10.1021/acs.biomac.3c00503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Efficient cellular alignment in biomaterials presents a considerable challenge, demanding the refinement of appropriate material morphologies, while ensuring effective cell-surface interactions. To address this, biomaterials are continuously researched with diverse coatings, hydrogels, and polymeric surfaces. In this context, we investigate the influence of physicochemical parameters on the architecture of fibrillar hydrogels that significantly orient the topography of flexible hydrogel substrates, thereby fostering cellular adhesion and spatial organization. Our Review comprehensively assesses various techniques for aligning polymer fibrils within hydrogels, specifically interventions applied during and after the cross-linking process. These methodologies include mechanical strains, precise temperature modulation, controlled fluidic dynamics, and chemical modulators, as well as the use of magnetic and electric fields. We highlight the intrinsic appeal of these methodologies in fabricating cell-aligning interfaces and discuss their potential implications within the fields of biomaterials and tissue engineering, particularly concerning the pursuit of optimal cellular alignment.
Collapse
Affiliation(s)
- Anatolii Abalymov
- Science Medical Center, Saratov State University, 410012 Saratov, Russia
| | - Bat-El Pinchasik
- School of Mechanical Engineering, Faculty of Engineering, Tel-Aviv University, 69978 Tel-Aviv, Israel
| | - Roman A Akasov
- Sechenov University and Federal Scientific Research Centre "Crystallography and Photonics" of Russian Academy of Sciences, 101000 Moscow, Russia
| | - Maria Lomova
- Science Medical Center, Saratov State University, 410012 Saratov, Russia
| | - Bogdan V Parakhonskiy
- Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
8
|
Anitua E, Zalduendo M, Troya M, Tierno R, Alkhraisat MH. Cellular composition modifies the biological properties and stability of platelet rich plasma membranes for tissue engineering. J Biomed Mater Res A 2023; 111:1710-1721. [PMID: 37318048 DOI: 10.1002/jbm.a.37579] [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: 06/22/2022] [Revised: 03/03/2023] [Accepted: 05/30/2023] [Indexed: 06/16/2023]
Abstract
Scaffolds should provide structural support for tissue regeneration, allowing their gradual biodegradation and interacting with cells and bioactive molecules to promote remodeling. Thus, the scaffold's intrinsic properties affect cellular processes involved in tissue regeneration, including migration, proliferation, differentiation, and protein synthesis. In this sense, due to its biological effect and clinical potential, Platelet Rich Plasma (PRP) fibrin could be considered a successful scaffold. Given the high variability in commercial PRPs formulations, this research focused on assessing the influence of cellular composition on fibrin membrane stability and remodeling cell activity. The stability and biological effect were evaluated at different time points via D-dimer, type I collagen and elastase quantification in culture media conditioned by Plasma Rich in Growth Factors - Fraction 1 (PRGF-F1), Plasma Rich in Growth Factors - Whole Plasma (PRGF-WP) and Leukocyte-rich Platelet Rich Plasma (L-PRP) membranes, and by gingival fibroblast cells seeded on them, respectively. Ultrastructure of PRP membranes was also evaluated. Histological analyses were performed after 5 and 18 days. Additionally, the effect of fibrin membranes on cell proliferation was determined. According to the results, L-PRP fibrin membranes degradation was complete at the end of the study, while PRGF membranes remained practically unchanged. Considering fibroblast behavior, PRGF membranes, in contrast to L-PRP ones, promoted extracellular matrix biosynthesis at the same time as fibrinolysis and enhanced cell proliferation. In conclusion, leukocytes in PRP fibrin membranes drastically reduce scaffold stability and induce behavioral changes in fibroblasts by reducing their proliferation rate and remodeling ability.
Collapse
Affiliation(s)
- Eduardo Anitua
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
| | - Mar Zalduendo
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
| | - María Troya
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
| | - Roberto Tierno
- Regenerative Medicine Laboratory, BTI-Biotechnology Institute, Vitoria, Spain
| | | |
Collapse
|
9
|
Awale GM, Barajaa MA, Kan HM, Seyedsalehi A, Nam GH, Hosseini FS, Ude CC, Schmidt TA, Lo KWH, Laurencin CT. Regenerative engineering of long bones using the small molecule forskolin. Proc Natl Acad Sci U S A 2023; 120:e2219756120. [PMID: 37216527 PMCID: PMC10235978 DOI: 10.1073/pnas.2219756120] [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: 11/18/2022] [Accepted: 04/10/2023] [Indexed: 05/24/2023] Open
Abstract
Bone grafting procedures have become increasingly common in the United States, with approximately 500,000 cases occurring each year at a societal cost exceeding $2.4 billion. Recombinant human bone morphogenetic proteins (rhBMPs) are therapeutic agents that have been widely used by orthopedic surgeons to stimulate bone tissue formation alone and when paired with biomaterials. However, significant limitations such as immunogenicity, high production cost, and ectopic bone growth from these therapies remain. Therefore, efforts have been made to discover and repurpose osteoinductive small-molecule therapeutics to promote bone regeneration. Previously, we have demonstrated that a single-dose treatment with the small-molecule forskolin for just 24 h induces osteogenic differentiation of rabbit bone marrow-derived stem cells in vitro, while mitigating adverse side effects attributed with prolonged small-molecule treatment schemes. In this study, we engineered a composite fibrin-PLGA [poly(lactide-co-glycolide)]-sintered microsphere scaffold for the localized, short-term delivery of the osteoinductive small molecule, forskolin. In vitro characterization studies showed that forskolin released out of the fibrin gel within the first 24 h and retained its bioactivity toward osteogenic differentiation of bone marrow-derived stem cells. The forskolin-loaded fibrin-PLGA scaffold was also able to guide bone formation in a 3-mo rabbit radial critical-sized defect model comparable to recombinant human bone morphogenetic protein-2 (rhBMP-2) treatment, as demonstrated through histological and mechanical evaluation, with minimal systemic off-target side effects. Together, these results demonstrate the successful application of an innovative small-molecule treatment approach within long bone critical-sized defects.
Collapse
Affiliation(s)
- Guleid M. Awale
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Department of Chemical Engineering, University of Connecticut, Storrs, CT06269
| | - Mohammed A. Barajaa
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT06030
- Department of Biomedical Engineering, Imam Abdulrahman Bin Faisal University,31451Dammam, Saudi Arabia
| | - Ho-Man Kan
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
| | - Amir Seyedsalehi
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT06030
| | - Ga Hie Nam
- Department of Pathology and Laboratory Medicine, UConn Health, Farmington, CT06030
| | - Fatemeh S. Hosseini
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Department of Skeletal Biology and Regeneration, UConn Health, Farmington, CT06030
| | - Chinedu C. Ude
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
| | - Tannin A. Schmidt
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT06030
| | - Kevin W.-H. Lo
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Division of Endocrinology, Department of Medicine, UConn Health, Farmington, CT06030
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT06030
| | - Cato T. Laurencin
- The Cato T. Laurencin Institute for Regenerative Engineering, University of Connecticut, Storrs, CT06269
- Department of Chemical Engineering, University of Connecticut, Storrs, CT06269
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT06030
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT06030
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT06269
| |
Collapse
|
10
|
Limelette M, De Fourmestraux C, Despas C, Lafragette A, Veziers J, Le Guennec Y, Touzot-Jourde G, Lefevre FX, Verron E, Bouler JM, Bujoli B, Gauthier O. Calcium Phosphate Cements Combined with Blood as a Promising Tool for the Treatment of Bone Marrow Lesions. J Funct Biomater 2023; 14:jfb14040204. [PMID: 37103294 PMCID: PMC10143268 DOI: 10.3390/jfb14040204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 03/24/2023] [Accepted: 04/03/2023] [Indexed: 04/28/2023] Open
Abstract
The solid phase of a commercial calcium phosphate (Graftys® HBS) was combined with ovine or human blood stabilized either with sodium citrate or sodium heparin. The presence of blood delayed the setting reaction of the cement by ca. 7-15 h, depending on the nature of the blood and blood stabilizer. This phenomenon was found to be directly related to the particle size of the HBS solid phase, since prolonged grinding of the latter resulted in a shortened setting time (10-30 min). Even though ca. 10 h were necessary for the HBS blood composite to harden, its cohesion right after injection was improved when compared to the HBS reference as well as its injectability. A fibrin-based material was gradually formed in the HBS blood composite to end-up, after ca. 100 h, with a dense 3D organic network present in the intergranular space, thus affecting the microstructure of the composite. Indeed, SEM analyses of polished cross-sections showed areas of low mineral density (over 10-20 µm) spread in the whole volume of the HBS blood composite. Most importantly, when the two cement formulations were injected in the tibial subchondral cancellous bone in a bone marrow lesion ovine model, quantitative SEM analyses showed a highly significant difference between the HBS reference versus its analogue combined with blood. After a 4-month implantation, histological analyses clearly showed that the HBS blood composite underwent high resorption (remaining cement: ca. 13.1 ± 7.3%) and new bone formation (newly formed bone: 41.8 ± 14.7%). This was in sharp contrast with the case of the HBS reference for which a low resorption rate was observed (remaining cement: 79.0 ± 6.9%; newly formed bone: 8.6 ± 4.8%). This study suggested that the particular microstructure, induced by the use of blood as the HBS liquid phase, favored quicker colonization of the implant and acceleration of its replacement by newly formed bone. For this reason, the HBS blood composite might be worth considering as a potentially suitable material for subchondroplasty.
Collapse
Affiliation(s)
- Maxence Limelette
- CNRS, CEISAM, UMR 6230, Nantes Université, 44000 Nantes, France
- Graftys SA, Eiffel Park, Pôle d'activités d'Aix en Provence, 13080 Aix en Provence, France
| | - Claire De Fourmestraux
- Department of Small Animal and Equine Surgery and Anesthesia, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering (ONIRIS), 44307 Nantes, France
- Regenerative Medicine and Skeleton, INSERM, University Hospital (CHU), UMR 1229-RMeS, Nantes University, 44000 Nantes, France
| | - Christelle Despas
- LCPME, CNRS UMR 7564, Université de Lorraine, 54800 Villers Lès Nancy, France
| | - Audrey Lafragette
- Department of Small Animal and Equine Surgery and Anesthesia, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering (ONIRIS), 44307 Nantes, France
| | - Joelle Veziers
- Regenerative Medicine and Skeleton, INSERM, University Hospital (CHU), UMR 1229-RMeS, Nantes University, 44000 Nantes, France
| | - Yohan Le Guennec
- Regenerative Medicine and Skeleton, INSERM, University Hospital (CHU), UMR 1229-RMeS, Nantes University, 44000 Nantes, France
| | - Gwenola Touzot-Jourde
- Department of Small Animal and Equine Surgery and Anesthesia, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering (ONIRIS), 44307 Nantes, France
- Regenerative Medicine and Skeleton, INSERM, University Hospital (CHU), UMR 1229-RMeS, Nantes University, 44000 Nantes, France
| | | | - Elise Verron
- CNRS, CEISAM, UMR 6230, Nantes Université, 44000 Nantes, France
| | | | - Bruno Bujoli
- CNRS, CEISAM, UMR 6230, Nantes Université, 44000 Nantes, France
| | - Olivier Gauthier
- Department of Small Animal and Equine Surgery and Anesthesia, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering (ONIRIS), 44307 Nantes, France
- Regenerative Medicine and Skeleton, INSERM, University Hospital (CHU), UMR 1229-RMeS, Nantes University, 44000 Nantes, France
| |
Collapse
|
11
|
Hoveizi E, Naddaf H, Ahmadianfar S, Bernardi S. Using Odontoblasts Derived from Dog Endometrial Stem Cells Encapsulated in Fibrin Gel Associated with BMP-2 in a Rat Pulp-Capping Model. Curr Issues Mol Biol 2023; 45:2984-2999. [PMID: 37185720 PMCID: PMC10136987 DOI: 10.3390/cimb45040196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 04/07/2023] Open
Abstract
This study aimed to treat dental injuries by utilizing one of the most advanced tissue engineering techniques. In this study, an in vitro model was employed to investigate the proliferation and odontogenic differentiation of canine endometrial stem cells (C-EnSCs). Furthermore, the dentin regeneration potential of odontoblast like-cells (OD) derived from C-EnSCs was assessed in rats. The C-EnSCs were isolated by the enzymatic method and identified by flow cytometry. The C-EnSCs were encapsulated in fibrin gel associated with signaling factors to create the proper conditions for cell growth and differentiation. Then, the OD cells were associated with bone morphologic protein-2 (BMP-2) to promote dentin formation in vivo. The animal model used to evaluate the regenerative effect of cells and biomaterials included the preparation of the left maxillary first molar of rats for direct pulp capping operation. Animals were divided into four groups: group 1, a control group without any treatment, group 2, which received fibrin, group 3, which received fibrin with ODs (fibrin/ODs), and group 4, which received fibrin with ODs and BMP-2 (fibrin/ODs/BMP-2). The morphological observations showed the differentiation of C-EnSCs into adipose, bone, neural cells, and ODs. Furthermore, the histomorphometric data of the treated teeth showed how fibrin gel and BMP2 at a concentration of 100 ng/mL provided an optimal microenvironment for regenerating dentin tissue in rats, which was increased significantly with the presence of OD cells within eight weeks. Our study showed that using OD cells derived from C-EnSCs encapsulated in fibrin gel associated with BMP2 can potentially be an appropriate candidate for direct pulp-capping and dentin regeneration.
Collapse
Affiliation(s)
- Elham Hoveizi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 6135783151, Iran
| | - Hadi Naddaf
- Department of Clinical Sciences, Faculty of Veterinary, Shahid Chamran University of Ahvaz, Ahvaz 6135783151, Iran
| | - Sina Ahmadianfar
- Department of Clinical Sciences, Faculty of Veterinary, Shahid Chamran University of Ahvaz, Ahvaz 6135783151, Iran
| | - Sara Bernardi
- Department of Life Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
12
|
Wachendörfer M, Buhl EM, Messaoud GB, Richtering W, Fischer H. pH and Thrombin Concentration Are Decisive in Synthesizing Stiff, Stable, and Open-Porous Fibrin-Collagen Hydrogel Blends without Chemical Cross-Linker. Adv Healthc Mater 2023; 12:e2203302. [PMID: 36546310 PMCID: PMC11468609 DOI: 10.1002/adhm.202203302] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Fibrin-collagen hydrogel blends exhibit high potential for tissue engineering applications. However, it is still unclear whether the underlying cross-linking mechanisms are of chemical or physical nature. It is here hypothesized that chemical cross-linkers play a negligible role and that instead pH and thrombin concentration are decisive for synthetizing blends with high stiffness and hydrolytic stability. Different fibrin-collagen formulations (pure and with additional transglutaminase) are used and the blends' compaction rate, hydrolytic stability, compressive strength, and hydrogel microstructure are investigated. The effect of thrombin concentration on gel compaction is examined and the importance of pH control during synthesis observed. It is revealed that transglutaminase impairs gel stability and it is deduced that fibrin-collagen blends mainly cross-link by mechanical interactions due to physical fibril entanglement as opposed to covalent bonds from chemical cross-linking. High thrombin concentrations and basic pH during synthesis reduce gel compaction and enhance stiffness and long-term stability. Scanning electron microscopy reveals a highly interpenetrating fibrous network with unique, interconnected open-porous microstructures. Endothelial cells proliferate on the blends and form a confluent monolayer. This study reveals the underlying cross-linking mechanisms and presents enhanced fibrin-collagen blends with high stiffness, hydrolytic stability, and large, interconnected pores; findings that offer high potential for advanced tissue engineering applications.
Collapse
Affiliation(s)
- Mattis Wachendörfer
- Department of Dental Materials and Biomaterials ResearchRWTH Aachen University HospitalPauwelsstrasse 3052074AachenGermany
| | - Eva Miriam Buhl
- Electron Microscopy FacilityInstitute of PathologyRWTH Aachen University HospitalPauwelsstrasse 3052074AachenGermany
| | - Ghazi Ben Messaoud
- Institute of Physical ChemistryRWTH Aachen UniversityLandoltweg 252074AachenGermany
- Physical ChemistryDWI–Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052074AachenGermany
| | - Walter Richtering
- Institute of Physical ChemistryRWTH Aachen UniversityLandoltweg 252074AachenGermany
- Physical ChemistryDWI–Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052074AachenGermany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials ResearchRWTH Aachen University HospitalPauwelsstrasse 3052074AachenGermany
| |
Collapse
|
13
|
Zuardi LR, Silva CLA, Rego EM, Carneiro GV, Spriano S, Nanci A, de Oliveira PT. Influence of a Physiologically Formed Blood Clot on Pre-Osteoblastic Cells Grown on a BMP-7-Coated Nanoporous Titanium Surface. Biomimetics (Basel) 2023; 8:biomimetics8010123. [PMID: 36975353 PMCID: PMC10046195 DOI: 10.3390/biomimetics8010123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
Titanium (Ti) nanotopography modulates the osteogenic response to exogenous bone morphogenetic protein 7 (BMP-7) in vitro, supporting enhanced alkaline phosphatase mRNA expression and activity, as well as higher osteopontin (OPN) mRNA and protein levels. As the biological effects of OPN protein are modulated by its proteolytic cleavage by serum proteases, this in vitro study evaluated the effects on osteogenic cells in the presence of a physiological blood clot previously formed on a BMP-7-coated nanostructured Ti surface obtained by chemical etching (Nano-Ti). Pre-osteoblastic MC3T3-E1 cells were cultured during 5 days on recombinant mouse (rm) BMP-7-coated Nano-Ti after it was implanted in adult female C57BI/6 mouse dorsal dermal tissue for 18 h. Nano-Ti without blood clot or with blood clot at time 0 were used as the controls. The presence of blood clots tended to inhibit the expression of key osteoblast markers, except for Opn, and rmBMP-7 functionalization resulted in a tendency towards relatively greater osteoblastic differentiation, which was corroborated by runt-related transcription factor 2 (RUNX2) amounts. Undetectable levels of OPN and phosphorylated suppressor of mothers against decapentaplegic (SMAD) 1/5/9 were noted in these groups, and the cleaved form of OPN was only detected in the blood clot immediately prior to cell plating. In conclusion, the strategy to mimic in vitro the initial interfacial in vivo events by forming a blood clot on a Ti nanoporous surface resulted in the inhibition of pre-osteoblastic differentiation, which was minimally reverted with an rmBMP-7 coating.
Collapse
Affiliation(s)
- Leonardo Raphael Zuardi
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Cleide Lúcia Araújo Silva
- Haematology Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, SP, Brazil
| | - Eduardo Magalhães Rego
- Haematology Division, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14051-060, SP, Brazil
| | - Giovana Vacilotto Carneiro
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
| | - Silvia Spriano
- Department of Applied Science and Technology, Politecnico di Torino, 10129 Torino, Italy
| | - Antonio Nanci
- Faculté de médecine dentaire, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Paulo Tambasco de Oliveira
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-904, SP, Brazil
- Correspondence: ; Tel.: +55-16-99623-3663
| |
Collapse
|
14
|
Fibrin-Rhamnogalacturonan I Composite Gel for Therapeutic Enzyme Delivery to Intestinal Tumors. Int J Mol Sci 2023; 24:ijms24020926. [PMID: 36674440 PMCID: PMC9862006 DOI: 10.3390/ijms24020926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/22/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Therapy of colorectal cancer with protein drugs, including targeted therapy using monoclonal antibodies, requires the preservation of the drug's structure and activity in the gastrointestinal tract or bloodstream. Here, we confirmed experimentally the fundamental possibility of creating composite protein-polysaccharide hydrogels based on non-degrading rhamnogalacturonan I (RG) and fibrin as a delivery vehicle for antitumor RNase binase. The method is based on enzymatic polymerization of fibrin in the presence of RG with the inclusion of liposomes, containing an encapsulated enzyme drug, into the gel network. The proposed method for fabricating a gel matrix does not require the use of cytotoxic chemical cross-linking agents and divalent cations, and contains completely biocompatible and biodegradable components. The process proceeds under physiological conditions, excluding the effect of high temperatures, organic solvents and ultrasound on protein components. Immobilization of therapeutic enzyme binase in the carrier matrix by encapsulating it in liposomes made from uncharged lipid made it possible to achieve its prolonged release with preservation of activity for a long time. The release time of binase from the composite carrier can be regulated by variation of the fibrin and RG concentration.
Collapse
|
15
|
Bone Marrow-Derived Fibrin Clots Stimulate Healing of a Meniscal Defect in a Rabbit Model. Arthroscopy 2022:S0749-8063(22)00838-6. [PMID: 36574822 DOI: 10.1016/j.arthro.2022.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/25/2022] [Accepted: 12/01/2022] [Indexed: 12/26/2022]
Abstract
PURPOSE To determine the in vivo effectiveness of bone marrow aspirate-derived (BMA) fibrin clots for avascular meniscal defect healing in a rabbit model. METHODS In 42 Japanese white rabbits, a 2.0-mm cylindrical defect was introduced into the avascular zone of the anterior part of the medial meniscus in the bilateral knees. The rabbits were grouped according to implantation of a BMA fibrin clot (BMA group) or a peripheral blood (PB)-derived clot (PB group) into the defect and nonimplantation (control group). Macroscopic and histological assessments were performed using a scoring system at 4 and 12 weeks after surgery. At 12 weeks after surgery, compressive stress was analyzed biomechanically. RESULTS The meniscal score in the BMA group (12.1) was greater than that in the PB group (5.5; P = .031) and control group (4.4; P = .013) at 4 weeks. The meniscal score in the BMA group (13.1) was greater than that in the control group (6.4; BMA = 13.1; P = .0046) at 12 weeks. In the biomechanical analysis, the BMA group demonstrated significantly higher compressive strength than the PB group (6.6 MPa) (BMA = 15.4 MPa; P = .0201) and control group (3.6 MPa; BMA = 15.4 MPa; P = .007). CONCLUSIONS Implantation of BMA fibrin clots into the meniscal defect of the avascular zone in a rabbit model improved the meniscal score at 4 weeks and strengthened the reparative meniscal tissue at 12 weeks compared with the implantation of PB fibrin clots. CLINICAL RELEVANCE Healing in the avascular zone of the meniscus can be problematic. Approaches to improving this healing response have had variable results. This study provides additional information that may help improve the outcomes in patients with these injuries.
Collapse
|
16
|
Novel Dental Restorative Solutions for Natural Teeth and Implants. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9120772. [PMID: 36550979 PMCID: PMC9774112 DOI: 10.3390/bioengineering9120772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
The long-term survival of restorations in the oral cavity has always been one of the most significant challenges in modern dental practice [...].
Collapse
|
17
|
Moise S, Dolcetti L, Dazzi F, Roach P, Buttery L, MacNeil S, Medcalf N. Assessing the immunosuppressive activity of alginate-encapsulated mesenchymal stromal cells on splenocytes. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2022; 50:168-176. [PMID: 35726746 DOI: 10.1080/21691401.2022.2088547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/09/2022] [Accepted: 06/05/2022] [Indexed: 06/15/2023]
Abstract
Mesenchymal stromal cells (MSCs) show immunosuppressive effects both via cell-to-cell contact (direct) with immune cells and by producing paracrine factors and extracellular vesicles (indirect). A key challenge in delivering this therapeutic effect in vivo is retaining the MSCs at the site of injection. One way to address this is by encapsulating the MSCs within suitable biomaterial scaffolds. Here, we assess the immunosuppressive effect of alginate-encapsulated murine MSCs on proliferating murine splenocytes. Our results show that MSCs are able to significantly suppress splenocyte proliferation by ∼50% via the indirect mechanism and almost completely (∼98%) via the direct mechanism. We also show for the first time that MSCs as monolayers on tissue culture plastic or encapsulated within alginate, when physically isolated from the splenocytes via transwells, are able to sustain immunosuppressive activity with repeated exposure to fresh splenocytes, for as long as 9 days. These results indicate the need to identify design strategies to simultaneously deliver both modes of MSC immunosuppression. By designing cell-biomaterial constructs with tailored degradation profiles, we can achieve a more sustained (avoiding MSCs migration and apoptosis) and controlled release of both the paracrine signals and eventually the cells themselves enabling efficient MSC-based immunosuppressive therapies for wound healing.
Collapse
Affiliation(s)
- Sandhya Moise
- Centre for Integrated Bioprocessing Research (CIBR), Department of Chemical Engineering, University of Bath, Bath, UK
- Centre for Therapeutic Innovation (CTI), University of Bath, Bath, UK
| | - Luigi Dolcetti
- Department of Medicine and Pharmaceutical Science, King's College London, London, UK
| | - Francesco Dazzi
- Department of Haematological malignancies and stem cell transplant, Kings College hospital NHS trust, London, UK
| | - Paul Roach
- Department of Chemistry, Loughborough University, Loughborough, UK
| | - Lee Buttery
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Sheila MacNeil
- Biomaterials and Tissue Engineering Group, Department of Materials Science and Engineering, Kroto Research Institute, University of Sheffield, Sheffield, UK
| | - Nick Medcalf
- Centre for Biological Engineering, Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough, UK
| |
Collapse
|
18
|
Biomaterials as regenerative medicine in Poly Cystic Ovarian Syndrome (PCOS) treatment. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
19
|
Wachendörfer M, Schräder P, Buhl EM, Palkowitz AL, Ben Messaoud G, Richtering W, Fischer H. A defined heat pretreatment of gelatin enables control of hydrolytic stability, stiffness, and microstructural architecture of fibrin-gelatin hydrogel blends. Biomater Sci 2022; 10:5552-5565. [PMID: 35969162 DOI: 10.1039/d2bm00214k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Fibrin-gelatin hydrogel blends exhibit high potential for tissue engineering in vitro applications. However, the means to tailor these blends in order to control their properties, thus opening up a broad range of new target applications, have been insufficiently explored. We hypothesized that a controlled heat treatment of gelatin prior to blend synthesis enables control of hydrolytic swelling and shrinking, stiffness, and microstructural architecture of fibrin-gelatin based hydrogel blends while providing tremendous long-term stability. We investigated these hydrogel blends' compressive strength, in vitro degradation stability, and microstructure in order to test this hypothesis. In addition, we examined the gel's ability to support endothelial cell proliferation and stretching of encapsulated smooth muscle cells. This research showed that a controlled heat pretreatment of the gelatin component strongly influenced the stiffness, swelling, shrinking, and microstructural architecture of the final blends regardless of identical gelatin mass fractions. All blends offered high long-term hydrolytic stability. In conclusion, the results of this study open the possibility to use this technique in order to tune low-concentrated, open-porous fibrin-based hydrogels, even in long-term tissue engineering in vitro experiments.
Collapse
Affiliation(s)
- Mattis Wachendörfer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Philipp Schräder
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Eva Miriam Buhl
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Alena L Palkowitz
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| | - Ghazi Ben Messaoud
- Institute of Physical Chemistry, RWTH Aachen University, Landoltweg 2, 52074 Aachen, Germany.,DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany
| | - Walter Richtering
- Institute of Physical Chemistry, RWTH Aachen University, Landoltweg 2, 52074 Aachen, Germany.,DWI-Leibniz Institute for Interactive Materials, Forckenbeckstr. 50, 52074 Aachen, Germany
| | - Horst Fischer
- Department of Dental Materials and Biomaterials Research, RWTH Aachen University Hospital, Pauwelsstrasse 30, 52074 Aachen, Germany.
| |
Collapse
|
20
|
Advances in Fibrin-Based Materials in Wound Repair: A Review. Molecules 2022; 27:molecules27144504. [PMID: 35889381 PMCID: PMC9322155 DOI: 10.3390/molecules27144504] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/28/2022] [Accepted: 07/08/2022] [Indexed: 11/29/2022] Open
Abstract
The first bioprocess that occurs in response to wounding is the deterrence of local hemorrhage. This is accomplished by platelet aggregation and initiation of the hemostasis cascade. The resulting blood clot immediately enables the cessation of bleeding and then functions as a provisional matrix for wound healing, which begins a few days after injury. Here, fibrinogen and fibrin fibers are the key players, because they literally serve as scaffolds for tissue regeneration and promote the migration of cells, as well as the ingrowth of tissues. Fibrin is also an important modulator of healing and a host defense system against microbes that effectively maintains incoming leukocytes and acts as reservoir for growth factors. This review presents recent advances in the understanding and applications of fibrin and fibrin-fiber-incorporated biomedical materials applied to wound healing and subsequent tissue repair. It also discusses how fibrin-based materials function through several wound healing stages including physical barrier formation, the entrapment of bacteria, drug and cell delivery, and eventual degradation. Pure fibrin is not mechanically strong and stable enough to act as a singular wound repair material. To alleviate this problem, this paper will demonstrate recent advances in the modification of fibrin with next-generation materials exhibiting enhanced stability and medical efficacy, along with a detailed look at the mechanical properties of fibrin and fibrin-laden materials. Specifically, fibrin-based nanocomposites and their role in wound repair, sustained drug release, cell delivery to wound sites, skin reconstruction, and biomedical applications of drug-loaded fibrin-based materials will be demonstrated and discussed.
Collapse
|
21
|
A Molecular View on Biomaterials and Dental Stem Cells Interactions: Literature Review. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12125815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Biomaterials and stem cells are essential components in the field of regenerative medicine. Various biomaterials have been designed that have appropriate biochemical and biophysical characteristics to mimic the microenvironment of an extracellular matrix. Dental stem cells (DT-MSCs) represent a novel source for the development of autologous therapies due to their easy availability. Although research on biomaterials and DT-MSCs has progressed, there are still challenges in the characteristics of biomaterials and the molecular mechanisms involved in regulating the behavior of DT-MSCs. In this review, the characteristics of biomaterials are summarized, and their classification according to their source, bioactivity, and different biological effects on the expansion and differentiation of DT-MSCs is summarized. Finally, advances in research on the interaction of biomaterials and the molecular components involved (mechanosensors and mechanotransduction) in DT-MSCs during their proliferation and differentiation are analyzed. Understanding the molecular dynamics of DT-MSCs and biomaterials can contribute to research in regenerative medicine and the development of autologous stem cell therapies.
Collapse
|
22
|
Neural Differentiation Potential of Mesenchymal Stem Cells Enhanced by Biocompatible Chitosan-Gold Nanocomposites. Cells 2022; 11:cells11121861. [PMID: 35740991 PMCID: PMC9221394 DOI: 10.3390/cells11121861] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/28/2022] [Accepted: 06/04/2022] [Indexed: 02/04/2023] Open
Abstract
Chitosan (Chi) is a natural polymer that has been demonstrated to have potential as a promoter of neural regeneration. In this study, Chi was prepared with various amounts (25, 50, and 100 ppm) of gold (Au) nanoparticles for use in in vitro and in vivo assessments. Each as-prepared material was first characterized by UV-visible spectroscopy (UV-Vis), Fourier-transform infrared spectroscopy (FTIR), atomic force microscopy (AFM), scanning electron microscopy (SEM), and Dynamic Light Scattering (DLS). Through the in vitro experiments, Chi combined with 50 ppm of Au nanoparticles demonstrated better biocompatibility. The platelet activation, monocyte conversion, and intracellular ROS generation was remarkably decreased by Chi–Au 50 pm treatment. Furthermore, Chi–Au 50 ppm could facilitate colony formation and strengthen matrix metalloproteinase (MMP) activation in mesenchymal stem cells (MSCs). The lower expression of CD44 in Chi–Au 50 ppm treatment demonstrated that the nanocomposites could enhance the MSCs undergoing differentiation. Chi–Au 50 ppm was discovered to significantly induce the expression of GFAP, β-Tubulin, and nestin protein in MSCs for neural differentiation, which was verified by real-time PCR analysis and immunostaining assays. Additionally, a rat model involving subcutaneous implantation was used to evaluate the superior anti-inflammatory and endothelialization abilities of a Chi–Au 50 ppm treatment. Capsule formation and collagen deposition were decreased. The CD86 expression (M1 macrophage polarization) and leukocyte filtration (CD45) were remarkably reduced as well. In summary, a Chi polymer combined with 50 ppm of Au nanoparticles was proven to enhance the neural differentiation of MSCs and showed potential as a biosafe nanomaterial for neural tissue engineering.
Collapse
|
23
|
Fan J, Abedi-Dorcheh K, Sadat Vaziri A, Kazemi-Aghdam F, Rafieyan S, Sohrabinejad M, Ghorbani M, Rastegar Adib F, Ghasemi Z, Klavins K, Jahed V. A Review of Recent Advances in Natural Polymer-Based Scaffolds for Musculoskeletal Tissue Engineering. Polymers (Basel) 2022; 14:polym14102097. [PMID: 35631979 PMCID: PMC9145843 DOI: 10.3390/polym14102097] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/09/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
The musculoskeletal (MS) system consists of bone, cartilage, tendon, ligament, and skeletal muscle, which forms the basic framework of the human body. This system plays a vital role in appropriate body functions, including movement, the protection of internal organs, support, hematopoiesis, and postural stability. Therefore, it is understandable that the damage or loss of MS tissues significantly reduces the quality of life and limits mobility. Tissue engineering and its applications in the healthcare industry have been rapidly growing over the past few decades. Tissue engineering has made significant contributions toward developing new therapeutic strategies for the treatment of MS defects and relevant disease. Among various biomaterials used for tissue engineering, natural polymers offer superior properties that promote optimal cell interaction and desired biological function. Natural polymers have similarity with the native ECM, including enzymatic degradation, bio-resorb and non-toxic degradation products, ability to conjugate with various agents, and high chemical versatility, biocompatibility, and bioactivity that promote optimal cell interaction and desired biological functions. This review summarizes recent advances in applying natural-based scaffolds for musculoskeletal tissue engineering.
Collapse
Affiliation(s)
- Jingzhi Fan
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia
| | - Keyvan Abedi-Dorcheh
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Asma Sadat Vaziri
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Fereshteh Kazemi-Aghdam
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Saeed Rafieyan
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Masoume Sohrabinejad
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Mina Ghorbani
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Fatemeh Rastegar Adib
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Zahra Ghasemi
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Kristaps Klavins
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia
- Correspondence: (K.K.); (V.J.)
| | - Vahid Jahed
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia
- Correspondence: (K.K.); (V.J.)
| |
Collapse
|
24
|
Wang K, Camman M, Mosser G, Haye B, Trichet L, Coradin T. Synthesis of Fibrin-Type I Collagen Biomaterials via an Acidic Gel. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27072099. [PMID: 35408498 PMCID: PMC9000341 DOI: 10.3390/molecules27072099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/17/2022] [Accepted: 03/23/2022] [Indexed: 11/16/2022]
Abstract
Fibrin-Type I collagen composite gels have been widely studied as biomaterials, in which both networks are usually formed simultaneously at a neutral pH. Here, we describe a new protocol in which mixed concentrated solutions of collagen and fibrinogen were first incubated at acidic pH to induce fibrinogen gel formation, followed by a pH change to neutral inducing collagen fiber formation. Thrombin was then added to form fibrin-collagen networks. Using this protocol, mixed gels containing 20 mg.mL−1 fibrin and up to 10 mg.mL−1 collagen could be prepared. Macroscopic observations evidenced that increasing the content of collagen increases the turbidity of the gels and decreases their shrinkage during the fibrinogen-to-fibrin conversion. The presence of collagen had a minor influence on the rheological properties of the gels. Electron microscopy allowed for observation of collagen fibers within the fibrin network. 2D cultures of C2C12 myoblasts on mixed gels revealed that the presence of collagen favors proliferation and local alignment of the cells. However, it interferes with cell differentiation and myotube formation, suggesting that further control of in-gel collagen self-assembly is required to elaborate fully functional biomaterials.
Collapse
|
25
|
Hashimoto Y, Nishino K, Orita K, Yamasaki S, Nishida Y, Kinoshita T, Nakamura H. Biochemical Characteristics and Clinical Result of Bone Marrow-Derived Fibrin Clot for Repair of Isolated Meniscal Injury in the Avascular Zone. Arthroscopy 2022; 38:441-449. [PMID: 34052371 DOI: 10.1016/j.arthro.2021.05.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 05/06/2021] [Accepted: 05/12/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE To characterize bone marrow aspirate-derived fibrin clot (BMA clot) and evaluate the clinical result of meniscal repair with a BMA clot for isolated meniscal injury in the avascular zone. METHODS Blood counts of total leukocytes, platelets, and concentrations of basic fibroblast growth factor (bFGF), transforming growth factor β (TGF-β), and stromal cell-derived factor 1 (SDF-1) were analyzed with BMA, peripheral blood (PB), BMA clot, and PB clot from 5 patients treated for meniscal repair. In addition, a retrospective analysis of 30 patients with isolated avascular meniscal injuries who underwent repair with a BMA clot was performed to assess rate failure. Avascular meniscal injury was identified as horizontal tear, radial tear, and flap tear. Clinical failure was defined as the presence of 1 or more of Barrett's criteria. Anatomic failure was defined as the existence of equivalent signal intensity to intra-articular fluid along the repair area on follow-up magnetic resonance imaging (MRI). Patients' demographic and clinical data were compared between the overall failure group and the success group. RESULTS The bFGF, TGF-β, and SDF-1 levels of BMA clots were more highly concentrated compared with PB clots. The Lysholm scores and meniscal status evaluated by MRI were significantly improved from preoperatively to postoperatively (both P < .001). The Kellgren-Lawrence grading of knee radiographs did not significantly differ pre- and postoperatively (P = .140). Rates of clinical failure, anatomic failure, and retear were 10%, 6.7%, and 3.3%, respectively. The demographic characteristics and surgical and postoperative status did not significantly differ between the overall failure group and the success group. CONCLUSIONS BMA clots had increased levels of cytokines compared to PB clots. The retrospective analysis revealed that the rates of clinical failure and anatomic failure after meniscal repair with a BMA clot for isolated avascular meniscal injury were 10% and 6.7%, respectively. LEVEL OF EVIDENCE Level IV, case series.
Collapse
Affiliation(s)
- Yusuke Hashimoto
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kazuya Nishino
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kumi Orita
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shinya Yamasaki
- Department of Orthopaedic Surgery, Osaka City General Hospital, Osaka, Japan
| | - Yohei Nishida
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Takuya Kinoshita
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Nakamura
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
26
|
Łukowicz K, Zagrajczuk B, Wieczorek J, Millan-Ciesielska K, Polkowska I, Cholewa-Kowalska K, Osyczka AM. Molecular Indicators of Biomaterials Osteoinductivity - Cell Migration, BMP Production and Signalling Turns a Key. Stem Cell Rev Rep 2022; 18:672-690. [PMID: 34782949 PMCID: PMC8930966 DOI: 10.1007/s12015-021-10300-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2021] [Indexed: 12/05/2022]
Abstract
In this work we dissected the osteoinductive properties of selected, PLGA-based scaffolds enriched with gel-derived bioactive glasses (SBGs) of either binary SiO2-CaO or ternary SiO2-CaO-P2O5 system, differing in CaO/SiO2 ratio (i.e. high -or low-calcium SBGs). To assess the inherent ability of the scaffolds to induce osteogenesis of human bone marrow stromal cells (BMSC), the study was designed to avoid any osteogenic stimuli beyond the putative osteogenic SBG component of the studied scaffolds. The bioactivity and porosity of scaffolds were confirmed by SBF test and porosimetry. Condition media (CM) from BMSC-loaded scaffolds exhibited increased Ca and decreased P content corresponding to SBGs CaO/SiO2 ratio, whereas Si content was relatively stable and overall lower in CM from scaffolds containing binary SBGs. CM from cell-loaded scaffolds containing high-calcium, binary SBGs promoted migration of BMSC and BMP-response in reporter osteoblast cell line. BMSC culture on these scaffolds or the ones containing ternary, low-calcium SBGs resulted in the activation of BMP-related signaling and expression of several osteogenic markers. Ectopic bone formation was induced by scaffolds containing binary SBGs, but high-calcium ones produced significantly more osteoid. Scaffolds containing ternary SBGs negatively influenced the expression of osteogenic transcription factors and Cx43, involved in cell-cell interactions. High-calcium scaffolds stimulated overall higher Cx43 expression. We believe the initial cell-cell communication may be crucial to induce and maintain osteogenesis and high BMP signaling on the studied scaffolds. The presented scaffolds' biological properties may also constitute new helpful markers to predict osteoinductive potential of other bioactive implant materials.
Collapse
Affiliation(s)
- Krzysztof Łukowicz
- Faculty of Biology, Institute of Zoology and Biomedical Research, Department of Biology and Cell Imaging, Jagiellonian University, 9 Gronostajowa St, 30-387, Krakow, Poland
| | - Barbara Zagrajczuk
- Faculty of Materials Science and Ceramics, Department of Glass Technology and Amorphous Coatings, AGH University of Science and Technology, 30 Mickiewicza Ave, 30-059, Krakow, Poland
| | - Jarosław Wieczorek
- University Centre of Veterinary Medicine UJ-UR, University of Agriculture in Krakow, 24/28 Mickiewicza Ave., 30-059, Krakow, Poland
| | - Katarzyna Millan-Ciesielska
- Faculty of Medicine, Department of Pathomorphology, Jagiellonian University Medical College, 2 Jakubowskiego St., 30-688, Krakow, Poland
| | - Izabela Polkowska
- Faculty of Veterinary Medicine, Department and Clinic of Animal Surgery, University of Life Sciences in Lublin, 13 Akademicka St., 20-950, Lublin, Poland
| | - Katarzyna Cholewa-Kowalska
- Faculty of Materials Science and Ceramics, Department of Glass Technology and Amorphous Coatings, AGH University of Science and Technology, 30 Mickiewicza Ave, 30-059, Krakow, Poland.
| | - Anna M Osyczka
- Faculty of Biology, Institute of Zoology and Biomedical Research, Department of Biology and Cell Imaging, Jagiellonian University, 9 Gronostajowa St, 30-387, Krakow, Poland.
| |
Collapse
|
27
|
Niu Q, He J, Wu M, Liu J, Lu X, Zhang L, Jin Z. Transplantation of bone marrow mesenchymal stem cells and fibrin glue into extraction socket in maxilla promoted bone regeneration in osteoporosis rat. Life Sci 2022; 290:119480. [PMID: 33862113 DOI: 10.1016/j.lfs.2021.119480] [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: 10/18/2020] [Revised: 03/03/2021] [Accepted: 03/31/2021] [Indexed: 11/20/2022]
Abstract
AIMS Bone defect repair in osteoporosis remains a tremendous challenge for clinicians due to increased bone metabolism resulted from estrogen deficiency. This study aims to investigate the effect of bone marrow mesenchymal stem cells (BMSCs) combined with fibrin glue (FG) in the extraction socket healing process of osteoporosis rats, as well as estimate the role of estrogen receptors (ERs) played in BMSCs differentiation in vitro and in the alveolar bone reconstruction process in vivo. MAIN METHODS Forty rats were randomly divided into four groups, under general anesthesia, three groups underwent bilateral ovariectomy(OVX) and one group with the sham operation. Three months later, the osteogenic ability of BMSCs, isolated from healthy and osteoporosis rats, respectively, was tested. The ERα and ERβ mRNA expression in BMSCs was also evaluated by RT-PCR analysis. In vivo experiment, Micro-CT detection, histological and immunofluorescent analysis, tissue PCR was conducted up to 2, 4 and 6 weeks after transplantation of BMSCs/FG to assess the newly formed bone in the extraction socket. KEY FINDINGS The BMSCs from osteoporosis rats displayed weaker osteogenic potential and lower ERs expression compared with the BMSCs from healthy rats. Newly formed bone tissue filled the socket defect in BMSCs/FG treated VOX rats after six weeks, which was comparable to the sham group, while reduced ERs expression was found in the regenerated bone of the OVX group. SIGNIFICANCE The BMSCs seeded within FG might provide an alternative therapeutic method for repairing the extraction socket defect in osteoporosis condition.
Collapse
Affiliation(s)
- Qiannan Niu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jiaojiao He
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Minke Wu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Pediatric Dentistry, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Jia Liu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Xiaolin Lu
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Liang Zhang
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Stomatology, The Air Force 986 Hospital, Xi'an, People's Republic of China.
| | - Zuolin Jin
- State Key Laboratory of Military Stomatology, National Clinical Research Center for Oral Diseases, Shaanxi Clinical Research Center for Oral Diseases, Fourth Military Medical University, Xi'an, People's Republic of China; Department of Orthodontics, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China.
| |
Collapse
|
28
|
Bahraminasab M, Janmohammadi M, Arab S, Talebi A, Nooshabadi VT, Koohsarian P, Nourbakhsh MS. Bone Scaffolds: An Incorporation of Biomaterials, Cells, and Biofactors. ACS Biomater Sci Eng 2021; 7:5397-5431. [PMID: 34797061 DOI: 10.1021/acsbiomaterials.1c00920] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Large injuries to bones are still one of the most challenging musculoskeletal problems. Tissue engineering can combine stem cells, scaffold biomaterials, and biofactors to aid in resolving this complication. Therefore, this review aims to provide information on the recent advances made to utilize the potential of biomaterials for making bone scaffolds and the assisted stem cell therapy and use of biofactors for bone tissue engineering. The requirements and different types of biomaterials used for making scaffolds are reviewed. Furthermore, the importance of stem cells and biofactors (growth factors and extracellular vesicles) in bone regeneration and their use in bone scaffolds and the key findings are discussed. Lastly, some of the main obstacles in bone tissue engineering and future trends are highlighted.
Collapse
Affiliation(s)
- Marjan Bahraminasab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran.,Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Mahsa Janmohammadi
- Department of Biomedical Engineering, Faculty of New Sciences and Technologies, Semnan University, Semnan 3513119111, Iran
| | - Samaneh Arab
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran.,Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Athar Talebi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran.,Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | - Parisa Koohsarian
- Department of Biochemistry and Hematology, School of Medicine, Semnan University of Medical Sciences, Semnan 3513138111, Iran
| | | |
Collapse
|
29
|
Mathieu L, Murison JC, de Rousiers A, de l’Escalopier N, Lutomski D, Collombet JM, Durand M. The Masquelet Technique: Can Disposable Polypropylene Syringes be an Alternative to Standard PMMA Spacers? A Rat Bone Defect Model. Clin Orthop Relat Res 2021; 479:2737-2751. [PMID: 34406150 PMCID: PMC8726567 DOI: 10.1097/corr.0000000000001939] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Usually, the two-stage Masquelet induced-membrane technique for extremity reconstruction begins with a polymethylmethacrylate (PMMA) cement spacer-driven membrane, followed by an autologous cancellous bone graft implanted into the membrane cavity to promote healing of large bone defects. In exceptional cases, spacers made of polypropylene disposable syringes were successfully used instead of the usual PMMA spacers because of a PMMA cement shortage caused by a lack of resources. However, this approach lacks clinical evidence and requires experimental validation before being recommended as an alternative to the conventional technique. QUESTIONS/PURPOSES To (1) develop and (2) validate a critical-sized femoral defect model in rats for two stages of the Masquelet technique and to (3) compare the biological and bone healing properties of polypropylene-induced membranes and PMMA-induced membranes in this model. METHODS Fifty male Sprague Dawley rats aged 8 weeks old received a 6-mm femur defect, which was stabilized with an external fixator that was converted into an internal device. In the development phase, the defect was filled with PMMA in 16 rats to determine the most favorable timing for bone grafting. Two rats were excluded since they died of anesthetic complications. The other 14 were successively euthanized after 2 weeks (n = 3), 4 weeks (n = 4), 6 weeks (n = 4), and 8 weeks (n = 3) for induced membrane analyses. In the validation phase, 12 rats underwent both stages of the procedure using a PMMA spacer and were randomly assigned to two groups, whether the induced membrane was preserved or removed before grafting. To address our final objective, we implanted either polypropylene or PMMA spacers into the defect (Masquelet technique Stage 1; n = 11 rats per group) for the period established by the development phase. In each group, 6 of 11 rats were euthanized to compare the biological properties of polypropylene-induced membranes and PMMA-induced membranes using histological qualitative analysis, semiquantitative assessment of the bone morphogenic protein-2 content by immunostaining, and qualitative assessment of the mesenchymal stromal cell (MSC; CD31-, CD45-, CD90+, and CD73+ phenotypes) content by flow cytometry. Quantitative measurements from serum bone turnover markers were also performed. The five remaining rats of each group were used for Masquelet technique Stage 2, in which rat bone allografts were implanted in the induced membrane cavity after the polypropylene or PMMA spacers were removed. These rats recovered for 10 weeks before being euthanized for microCT quantitative measurements and bone histology qualitative assessment to evaluate and compare the extent of bone regeneration between groups. RESULTS Induced membrane analyses together with serum bone turnover measurements indicated that a 4-week interval time between stages was the most favorable. Removal of the induced membrane before grafting led to almost constant early implant failures with poor bone formation. Four-week-old rats with polypropylene-triggered induced membranes displayed similar histologic organization as rats with PMMA-driven induced membranes, without any difference in the cell density of the extracellular matrix (4933 ± 916 cells per mm2 for polypropylene versus 4923 ± 1284 cells per mm2 for PMMA; p = 0.98). Induced membrane-derived MSCs were found in both groups with no difference (4 of 5 with polypropylene versus 3 of 3 with PMMA; p > 0.99). Induced membrane bone morphogenic protein-2 immunolabeling and serum bone turnover marker levels were comparable between the polypropylene and PMMA groups. MicroCT analysis found that bone regeneration in the polypropylene group seemed comparable with that in the PMMA group (29 ± 26 mm3 for polypropylene versus 24 ± 18 mm3 for PMMA; p > 0.99). Finally, qualitative histological assessment revealed a satisfactory endochondral ossification maturation in both groups. CONCLUSION Using a critical-sized femoral defect model in rats, we demonstrated that polypropylene spacers could induce membrane encapsulation with histologic characteristics and bone regenerative capacities that seem like those of PMMA spacers. CLINICAL RELEVANCE In a same bone site, polymers with close physical properties seem to lead to similar foreign body reactions and induce encapsulating membranes with comparable bone healing properties. Polypropylene spacers made from disposable syringes could be a valuable alternative to PMMA. These results support the possibility of a cementless Masquelet technique in cases of PMMA shortage caused by a lack of resources.
Collapse
Affiliation(s)
- Laurent Mathieu
- Department of Orthopedic, Trauma and Reconstructive Surgery, Percy Military Hospital, Clamart, France
- French Military Health Service Academy, Ecole du Val-de-Grâce, Paris, France
| | - James Charles Murison
- Department of Orthopedic, Trauma and Reconstructive Surgery, Percy Military Hospital, Clamart, France
| | - Arnaud de Rousiers
- Department of Orthopedic, Trauma and Reconstructive Surgery, Percy Military Hospital, Clamart, France
| | - Nicolas de l’Escalopier
- Department of Orthopedic, Trauma and Reconstructive Surgery, Percy Military Hospital, Clamart, France
| | - Didier Lutomski
- Tissue Engineering and Proteomics Team, Université Paris, Bobigny, France
| | | | - Marjorie Durand
- Military Biomedical Research Institute, Brétigny-sur-Orge, France
| |
Collapse
|
30
|
Zastosowanie fibryny w inżynierii tkankowej. Osiągnięcia i perspektywy. POSTEP HIG MED DOSW 2021. [DOI: 10.2478/ahem-2021-0017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstrakt
W ostatnich latach istotnym obszarem zastosowania fibryny stała się inżynieria tkankowa, w której wykorzystuje się naturalne właściwości biostatyczne i bioaktywne fibryny, a także możliwość pułapkowania i wiązania w jej strukturze czynników wzrostu. Fibryna jest najczęściej stosowana w postaci żeli i dysków. Jednak każda postać wskutek pochłaniania wody docelowo przyjmuje postać żelu. Białko to w warunkach in vivo spełnia rolę rusztowania dla komórek, a także może być aplikowane w miejsca trudno dostępne – może wypełniać ubytki tkanek i podtrzymywać tkanki okalające, zapobiegając ich zapadaniu się. Ponadto fibryna hamuje krwawienie i inicjuje proces odnowy, jak również pełni rolę stymulatora wzrostu komórek. Przez modyfikacje struktury fibryny cząsteczkami adhezyjnymi, można przyspieszyć odbudowę prawidłowej struktury tkanek. Jej właściwości strukturalne mogą być także wykorzystywane jako rezerwuar czynników wzrostu i system ich przedłużonego uwalniania. Fibryna jest materiałem biodegradowalnym, umożliwiając skorelowanie ubytku matrycy fibrynowej z odbudową tkanek własnych pacjenta. Wprowadzenie metod druku 3D i elektroprzędzenia umożliwia formulację dopasowanych do uszkodzeń kształtek oraz włóknin bez utraty bioaktywnych funkcji fibryny. Metody te umożliwiają także poprawę właściwości mechanicznych przez otrzymywanie m.in. włóknin fibryny z innymi polimerami, co jest szczególnie uzasadnione w przypadku materiałów stosowanych w odbudowie takich struktur jak ścięgna czy kości. Biotechnologiczna synteza fibrynogenu może w przyszłości uniezależnić pozyskiwanie go z krwi i zwiększyć popularność wyrobów medycznych otrzymywanych z fibryny.
Collapse
|
31
|
Pliszczak-Król A, Kiełbowicz Z, Król J, Antończyk A, Gemra M, Skrzypczak P, Prządka P, Zalewski D, Bieżyński J, Nicpoń J. Parameters of Hemostasis in Sheep Implanted with Composite Scaffold Settled by Stimulated Mesenchymal Stem Cells-Evaluation of the Animal Model. MATERIALS 2021; 14:ma14226934. [PMID: 34832335 PMCID: PMC8622787 DOI: 10.3390/ma14226934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 11/26/2022]
Abstract
Implantation of composite scaffolds could be potentially associated with the risk of hemostatic disturbances in a recipient. However, there is a lack of information on possible alterations in clotting mechanisms resulting from such a procedure. The aim of the present work was to investigate changes in hemostatic parameters in sheep implanted with a scaffold composed of poly(ε-caprolactone) and hydroxyapatite and tricalcium phosphate (9:4.5:4.5), settled previously with mesenchymal stem cells stimulated by fibroblast growth factor-2 and bone morphogenetic protein-2. Nine Merino sheep were examined for 7 days, and measurements of clotting times (PT, aPTT), activities of antithrombin, protein C and clotting factors II-XII, and concentrations of fibrinogen and D-dimer were carried out before and 1 h, 24 h, 3 days and 7 days after scaffold implantation. The introduction of scaffold initially resulted in a slowdown of the clotting processes (most evident 24 h after surgery); PT and aPTT increased to 14.8 s and 33.9 s, respectively. From the third day onwards, most of these alterations began to return to normal values. The concentration of fibrinogen rose throughout the observation period (up to 8.4 g/L), mirroring the ongoing inflammatory reaction. However, no signals of significant disturbances in hemostatic processes were detected in the sheep tested.
Collapse
Affiliation(s)
- Aleksandra Pliszczak-Król
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland;
- Correspondence: ; Tel.: +48-6-6409-2994
| | - Zdzisław Kiełbowicz
- Department and Clinic of Surgery, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland; (Z.K.); (A.A.); (P.S.); (P.P.); (J.B.); (J.N.)
| | - Jarosław Król
- Department of Pathology, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland;
| | - Agnieszka Antończyk
- Department and Clinic of Surgery, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland; (Z.K.); (A.A.); (P.S.); (P.P.); (J.B.); (J.N.)
| | - Marianna Gemra
- Department of Immunology, Pathophysiology and Veterinary Preventive Medicine, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland;
| | - Piotr Skrzypczak
- Department and Clinic of Surgery, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland; (Z.K.); (A.A.); (P.S.); (P.P.); (J.B.); (J.N.)
| | - Przemysław Prządka
- Department and Clinic of Surgery, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland; (Z.K.); (A.A.); (P.S.); (P.P.); (J.B.); (J.N.)
| | - Dariusz Zalewski
- Department of Genetics, Plant Breeding and Seed Production, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland;
| | - Janusz Bieżyński
- Department and Clinic of Surgery, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland; (Z.K.); (A.A.); (P.S.); (P.P.); (J.B.); (J.N.)
| | - Jakub Nicpoń
- Department and Clinic of Surgery, Wroclaw University of Environmental and Life Sciences, 50-375 Wroclaw, Poland; (Z.K.); (A.A.); (P.S.); (P.P.); (J.B.); (J.N.)
| |
Collapse
|
32
|
Dadashzadeh A, Moghassemi S, Shavandi A, Amorim CA. A review on biomaterials for ovarian tissue engineering. Acta Biomater 2021; 135:48-63. [PMID: 34454083 DOI: 10.1016/j.actbio.2021.08.026] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/26/2021] [Accepted: 08/18/2021] [Indexed: 12/19/2022]
Abstract
Considerable challenges in engineering the female reproductive tissue are the follicle's unique architecture, the need to recapitulate the extracellular matrix, and tissue vascularization. Over the years, various strategies have been developed for preserving fertility in women diagnosed with cancer, such as embryo, oocyte, or ovarian tissue cryopreservation. While autotransplantation of cryopreserved ovarian tissue is a viable choice to restore fertility in prepubertal girls and women who need to begin chemo- or radiotherapy soon after the cancer diagnosis, it is not suitable for all patients due to the risk of having malignant cells present in the ovarian fragments in some types of cancer. Advances in tissue engineering such as 3D printing and ovary-on-a-chip technologies have the potential to be a translational strategy for precisely recapitulating normal tissue in terms of physical structure, vascularization, and molecular and cellular spatial distribution. This review first introduces the ovarian tissue structure, describes suitable properties of biomaterials for ovarian tissue engineering, and highlights recent advances in tissue engineering for developing an artificial ovary. STATEMENT OF SIGNIFICANCE: The increase of survival rates in young cancer patients has been accompanied by a rise in infertility/sterility in cancer survivors caused by the gonadotoxic effect of some chemotherapy regimens or radiotherapy. Such side-effect has a negative impact on these patients' quality of life as one of their main concerns is generating biologically related children. To aid female cancer patients, several research groups have been resorting to tissue engineering strategies to develop an artificial ovary. In this review, we discuss the numerous biomaterials cited in the literature that have been tested to encapsulate and in vitro culture or transplant isolated preantral follicles from human and different animal models. We also summarize the recent advances in tissue engineering that can potentially be optimal strategies for developing an artificial ovary.
Collapse
|
33
|
Foroutan T. Application of some graphene derivatives to increase the efficiency of stem cell therapy. Curr Stem Cell Res Ther 2021; 17:294-300. [PMID: 34727863 DOI: 10.2174/1574888x16666211102085246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 08/24/2021] [Accepted: 09/12/2021] [Indexed: 11/22/2022]
Abstract
Graphene and its derivatives have application potential in many areas such as environmental technology, catalysis, biomedicine, and in particular, stem cell-based differentiation and regenerative therapies. Mesenchymal stem cell transplantation has emerged as a potential therapy for some diseases, such as acute kidney damage, liver failure and myocardial infarction. However, the poor survival of transplanted stem cells in such applications has significantly limited their therapeutic effectiveness. Graphene-based materials can improve the therapeutic efficacy of stem cells as they prevent the death of implanted cells by attaching them prior to implantation and increasing their paracrine secretion. In this review, we will highlight a number of recent studies that have investigated the potential use of graphene or its derivatives in stem cell applications and the prevention of transplanted stem cells from cell death, thereby improving their therapeutic efficacy.
Collapse
Affiliation(s)
- Tahereh Foroutan
- Department of animal biology, Faculty of biological sciences, Kharazmi University, Tehran. Iran
| |
Collapse
|
34
|
Ogunnaike EA, Valdivia A, Yazdimamaghani M, Leon E, Nandi S, Hudson H, Du H, Khagi S, Gu Z, Savoldo B, Ligler FS, Hingtgen S, Dotti G. Fibrin gel enhances the antitumor effects of chimeric antigen receptor T cells in glioblastoma. SCIENCE ADVANCES 2021; 7:eabg5841. [PMID: 34613775 PMCID: PMC8494441 DOI: 10.1126/sciadv.abg5841] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 08/16/2021] [Indexed: 05/14/2023]
Abstract
Regional delivery of chimeric antigen receptor (CAR) T cells in glioblastoma represents a rational therapeutic approach as an alternative to intravenous administration to avoid the blood-brain barrier impediment. Here, we developed a fibrin gel that accommodates CAR-T cell loading and promotes their gradual release. Using a model of subtotal glioblastoma resection, we demonstrated that the fibrin-based gel delivery of CAR-T cells within the surgical cavity enables superior antitumor activity compared to CAR-T cells directly inoculated into the tumor resection cavity.
Collapse
Affiliation(s)
- Edikan A. Ogunnaike
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC 27695, USA
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Alain Valdivia
- Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mostafa Yazdimamaghani
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ernesto Leon
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Seema Nandi
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC 27695, USA
| | - Hannah Hudson
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hongwei Du
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Simon Khagi
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Medicine, Division of Medical Oncology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Barbara Savoldo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC 27514, USA
| | - Frances S. Ligler
- Joint Department of Biomedical Engineering, North Carolina State University and University of North Carolina at Chapel Hill, Raleigh, NC 27695, USA
- Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Shawn Hingtgen
- Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Gianpietro Dotti
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
35
|
Ruhoff AM, Hong JK, Gao L, Singh J, Tran C, Mackie G, Waterhouse A. Biomaterial Wettability Affects Fibrin Clot Structure and Fibrinolysis. Adv Healthc Mater 2021; 10:e2100988. [PMID: 34423587 DOI: 10.1002/adhm.202100988] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/29/2021] [Indexed: 12/20/2022]
Abstract
Thrombosis on blood-contacting medical devices can cause patient fatalities through device failure and unstable thrombi causing embolism. The effect of material wettability on fibrin network formation, structure, and stability is poorly understood. Under static conditions, fibrin fiber network volume and density increase in clots formed on hydrophilic compared to hydrophobic polystyrene surfaces. This correlates with reduced plasma clotting time and increased factor XIIa (FXIIa) activity. These structural differences are consistent up to 50 µm away from the material surface and are FXIIa dependent. Fibrin forms fibers immediately at the material interface on hydrophilic surfaces but are incompletely formed in the first 5 µm above hydrophobic surfaces. Additionally, fibrin clots on hydrophobic surfaces have increased susceptibility to fibrinolysis compared to clots formed on hydrophilic surfaces. Under low-flow conditions, clots are still denser on hydrophilic surfaces, but only 5 µm above the surface, showing the combined effect of the surface wettability and coagulation factor dilution with low flow. Overall, wettability affects fibrin fiber formation at material interfaces, which leads to differences in bulk fibrin clot density and susceptibility to fibrinolysis. These findings have implications for thrombus formed in stagnant or low-flow regions of medical devices and the design of nonthrombogenic materials.
Collapse
Affiliation(s)
- Alexander M. Ruhoff
- Heart Research Institute 7 Eliza Street Newtown NSW 2042 Australia
- The Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
- Faculty of Medicine and Health The University of Sydney Sydney NSW 2006 Australia
| | - Jun Ki Hong
- Heart Research Institute 7 Eliza Street Newtown NSW 2042 Australia
- The Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
- School of Chemistry Faculty of Science The University of Sydney Sydney NSW 2006 Australia
- School of Medical Sciences Faculty of Medicine and Health The University of Sydney Sydney NSW 2006 Australia
- The University of Sydney Nano Institute The University of Sydney Sydney NSW 2006 Australia
| | - Lingzi Gao
- Heart Research Institute 7 Eliza Street Newtown NSW 2042 Australia
- The Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
- Faculty of Medicine and Health The University of Sydney Sydney NSW 2006 Australia
| | - Jasneil Singh
- Heart Research Institute 7 Eliza Street Newtown NSW 2042 Australia
- The Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
- School of Medical Sciences Faculty of Medicine and Health The University of Sydney Sydney NSW 2006 Australia
- School of Biomedical Engineering Faculty of Engineering The University of Sydney Sydney NSW 2006 Australia
| | - Clara Tran
- School of Biomedical Engineering Faculty of Engineering The University of Sydney Sydney NSW 2006 Australia
- School of Physics Faculty of Science The University of Sydney Sydney NSW 2006 Australia
| | - Grace Mackie
- Heart Research Institute 7 Eliza Street Newtown NSW 2042 Australia
- The Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
- Faculty of Medicine and Health The University of Sydney Sydney NSW 2006 Australia
| | - Anna Waterhouse
- Heart Research Institute 7 Eliza Street Newtown NSW 2042 Australia
- The Charles Perkins Centre The University of Sydney Sydney NSW 2006 Australia
- School of Medical Sciences Faculty of Medicine and Health The University of Sydney Sydney NSW 2006 Australia
- The University of Sydney Nano Institute The University of Sydney Sydney NSW 2006 Australia
| |
Collapse
|
36
|
The Cellular Choreography of Osteoblast Angiotropism in Bone Development and Homeostasis. Int J Mol Sci 2021; 22:ijms22147253. [PMID: 34298886 PMCID: PMC8305002 DOI: 10.3390/ijms22147253] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/23/2021] [Accepted: 06/29/2021] [Indexed: 12/11/2022] Open
Abstract
Interaction between endothelial cells and osteoblasts is essential for bone development and homeostasis. This process is mediated in large part by osteoblast angiotropism, the migration of osteoblasts alongside blood vessels, which is crucial for the homing of osteoblasts to sites of bone formation during embryogenesis and in mature bones during remodeling and repair. Specialized bone endothelial cells that form "type H" capillaries have emerged as key interaction partners of osteoblasts, regulating osteoblast differentiation and maturation and ensuring their migration towards newly forming trabecular bone areas. Recent revolutions in high-resolution imaging methodologies for bone as well as single cell and RNA sequencing technologies have enabled the identification of some of the signaling pathways and molecular interactions that underpin this regulatory relationship. Similarly, the intercellular cross talk between endothelial cells and entombed osteocytes that is essential for bone formation, repair, and maintenance are beginning to be uncovered. This is a relatively new area of research that has, until recently, been hampered by a lack of appropriate analysis tools. Now that these tools are available, greater understanding of the molecular relationships between these key cell types is expected to facilitate identification of new drug targets for diseases of bone formation and remodeling.
Collapse
|
37
|
Fibrin Glue Implants Seeded with Dental Pulp and Periodontal Ligament Stem Cells for the Repair of Periodontal Bone Defects: A Preclinical Study. Bioengineering (Basel) 2021; 8:bioengineering8060075. [PMID: 34206126 PMCID: PMC8226811 DOI: 10.3390/bioengineering8060075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 05/25/2021] [Indexed: 12/30/2022] Open
Abstract
A technology to create a cell-seeded fibrin-based implant matching the size and shape of bone defect is required to create an anatomical implant. The aim of the study was to develop a technology of cell-seeded fibrin gel implant creation that has the same shape and size as the bone defect at the site of implantation. Using computed tomography (CT) images, molds representing bone defects were created by 3D printing. The form was filled with fibrin glue and human dental pulp stem cells (DPSC). The viability, set of surface markers and osteogenic differentiation of DPSC grown in fibrin gel along with the clot retraction time were evaluated. In mice, an alveolar bone defect was created. The defect was filled with fibrin gel seeded with mouse DPSC. After 28 days, the bone repair was analyzed with cone beam CT and by histological examination. The proliferation rate, set of surface antigens and osteogenic potential of cells grown inside the scaffold and in 2D conditions did not differ. In mice, both cell-free and mouse DPSC-seeded implants increased the bone tissue volume and vascularization. In mice with cell-seeded gel implants, the bone remodeling process was more prominent than in animals with a cell-free implant. The technology of 3D-printed forms for molding implants can be used to prepare implants using components that are not suitable for 3D printing.
Collapse
|
38
|
Olaru M, Sachelarie L, Calin G. Hard Dental Tissues Regeneration-Approaches and Challenges. MATERIALS 2021; 14:ma14102558. [PMID: 34069265 PMCID: PMC8156070 DOI: 10.3390/ma14102558] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 12/13/2022]
Abstract
With the development of the modern concept of tissue engineering approach and the discovery of the potential of stem cells in dentistry, the regeneration of hard dental tissues has become a reality and a priority of modern dentistry. The present review reports the recent advances on stem-cell based regeneration strategies for hard dental tissues and analyze the feasibility of stem cells and of growth factors in scaffolds-based or scaffold-free approaches in inducing the regeneration of either the whole tooth or only of its component structures.
Collapse
Affiliation(s)
- Mihaela Olaru
- “Petru Poni” Institute of Macromolecular Chemistry, 41 A Grigore Ghica Voda Alley, 700487 Iasi, Romania;
| | - Liliana Sachelarie
- Faculty of Medical Dentistry, “Apollonia” University of Iasi, 2 Muzicii Str., 700399 Iasi, Romania;
- Correspondence:
| | - Gabriela Calin
- Faculty of Medical Dentistry, “Apollonia” University of Iasi, 2 Muzicii Str., 700399 Iasi, Romania;
| |
Collapse
|
39
|
Ogay V, Mun EA, Kudaibergen G, Baidarbekov M, Kassymbek K, Zharkinbekov Z, Saparov A. Progress and Prospects of Polymer-Based Drug Delivery Systems for Bone Tissue Regeneration. Polymers (Basel) 2020; 12:E2881. [PMID: 33271770 PMCID: PMC7760650 DOI: 10.3390/polym12122881] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the high regenerative capacity of bone tissue, there are some cases where bone repair is insufficient for a complete functional and structural recovery after damage. Current surgical techniques utilize natural and synthetic bone grafts for bone healing, as well as collagen sponges loaded with drugs. However, there are certain disadvantages associated with these techniques in clinical usage. To improve the therapeutic efficacy of bone tissue regeneration, a number of drug delivery systems based on biodegradable natural and synthetic polymers were developed and examined in in vitro and in vivo studies. Recent studies have demonstrated that biodegradable polymers play a key role in the development of innovative drug delivery systems and tissue engineered constructs, which improve the treatment and regeneration of damaged bone tissue. In this review, we discuss the most recent advances in the field of polymer-based drug delivery systems for the promotion of bone tissue regeneration and the physical-chemical modifications of polymers for controlled and sustained release of one or more drugs. In addition, special attention is given to recent developments on polymer nano- and microparticle-based drug delivery systems for bone regeneration.
Collapse
Affiliation(s)
- Vyacheslav Ogay
- Stem Cell Laboratory, National Center for Biotechnology, Nur-Sultan 010000, Kazakhstan; (V.O.); (G.K.)
| | - Ellina A. Mun
- School of Sciences and Humanities, Nazarbayev University, Nur-Sultan 010000, Kazakhstan;
| | - Gulshakhar Kudaibergen
- Stem Cell Laboratory, National Center for Biotechnology, Nur-Sultan 010000, Kazakhstan; (V.O.); (G.K.)
| | - Murat Baidarbekov
- Research Institute of Traumatology and Orthopedics, Nur-Sultan 010000, Kazakhstan;
| | - Kuat Kassymbek
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (K.K.); (Z.Z.)
| | - Zharylkasyn Zharkinbekov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (K.K.); (Z.Z.)
| | - Arman Saparov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan 010000, Kazakhstan; (K.K.); (Z.Z.)
| |
Collapse
|
40
|
de Melo BA, Jodat YA, Cruz EM, Benincasa JC, Shin SR, Porcionatto MA. Strategies to use fibrinogen as bioink for 3D bioprinting fibrin-based soft and hard tissues. Acta Biomater 2020; 117:60-76. [PMID: 32949823 DOI: 10.1016/j.actbio.2020.09.024] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/03/2020] [Accepted: 09/11/2020] [Indexed: 12/16/2022]
Abstract
Fibrin gel has been widely used for engineering various types of tissues due to its biocompatible nature, biodegradability, and tunable mechanical and nanofibrous structural properties. Despite their promising regenerative capacity and extensive biocompatibility with various tissue types, fibrin-based biomaterials are often notoriously known as burdensome candidates for 3D biofabrication and bioprinting. The high viscosity of fibrin (crosslinked form) hinders proper ink extrusion, and its pre-polymer form, fibrinogen, is not capable of maintaining shape fidelity. To overcome these limitations and empower fibrinogen-based bioinks for fibrin biomimetics and regenerative applications, different strategies can be practiced. The aim of this review is to report the strategies that bring fabrication compatibility to these bioinks through mixing fibrinogen with printable biomaterials, using supporting bath supplemented with crosslinking agents, and crosslinking fibrin in situ. Moreover, the review discusses some of the recent advances in 3D bioprinting of biomimetic soft and hard tissues using fibrinogen-based bioinks, and highlights the impacts of these strategies on fibrin properties, its bioactivity, and the functionality of the consequent biomimetic tissue. Statement of Significance Due to its biocompatible nature, biodegradability, and tunable mechanical and nanofibrous structural properties, fibrin gel has been widely employed in tissue engineering and more recently, used as in 3D bioprinting. The fibrinogen's poor printable properties make it difficult to maintain the 3D shape of bioprinted constructs. Our work describes the strategies employed in tissue engineering to allow the 3D bioprinting of fibrinogen-based bioinks, such as the combination of fibrinogen with printable biomaterials, the in situ fibrin crosslinking, and the use of supporting bath supplemented with crosslinking agents. Further, this review discuss the application of 3D bioprinting technology to biofabricate fibrin-based soft and hard tissues for biomedical applications, and discuss current limitations and future of such in vitro models.
Collapse
|
41
|
Kim MG, Park CH. Tooth-Supporting Hard Tissue Regeneration Using Biopolymeric Material Fabrication Strategies. Molecules 2020; 25:molecules25204802. [PMID: 33086674 PMCID: PMC7587995 DOI: 10.3390/molecules25204802] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/08/2020] [Accepted: 10/16/2020] [Indexed: 12/13/2022] Open
Abstract
The mineralized tissues (alveolar bone and cementum) are the major components of periodontal tissues and play a critical role to anchor periodontal ligament (PDL) to tooth-root surfaces. The integrated multiple tissues could generate biological or physiological responses to transmitted biomechanical forces by mastication or occlusion. However, due to periodontitis or traumatic injuries, affect destruction or progressive damage of periodontal hard tissues including PDL could be affected and consequently lead to tooth loss. Conventional tissue engineering approaches have been developed to regenerate or repair periodontium but, engineered periodontal tissue formation is still challenging because there are still limitations to control spatial compartmentalization for individual tissues and provide optimal 3D constructs for tooth-supporting tissue regeneration and maturation. Here, we present the recently developed strategies to induce osteogenesis and cementogenesis by the fabrication of 3D architectures or the chemical modifications of biopolymeric materials. These techniques in tooth-supporting hard tissue engineering are highly promising to promote the periodontal regeneration and advance the interfacial tissue formation for tissue integrations of PDL fibrous connective tissue bundles (alveolar bone-to-PDL or PDL-to-cementum) for functioning restorations of the periodontal complex.
Collapse
Affiliation(s)
- Min Guk Kim
- Department of Dental Science, Graduate School, Kyungpook National University, Daegu 41940, Korea;
- Department of Dental Biomaterials, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
| | - Chan Ho Park
- Department of Dental Science, Graduate School, Kyungpook National University, Daegu 41940, Korea;
- Department of Dental Biomaterials, School of Dentistry, Kyungpook National University, Daegu 41940, Korea
- Institute for Biomaterials Research and Development, Kyungpook National University, Daegu 41940, Korea
- Correspondence: ; Tel.: +82-53-660-6890
| |
Collapse
|
42
|
Bone marrow aspirate clot: A feasible orthobiologic. J Clin Orthop Trauma 2020; 11:S789-S794. [PMID: 32999557 PMCID: PMC7503156 DOI: 10.1016/j.jcot.2020.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 07/05/2020] [Indexed: 12/29/2022] Open
Abstract
Musculoskeletal disorders are one of the major health burdens and a leading source of disability worldwide, affecting both juvenile and elderly populations either as a consequence of ageing or extrinsic factors such as physical injuries. This condition often involves a group of locomotor structures such as the bones, joints and muscles and may therefore cause significant economic and emotional impact. Some pharmacological and non-pharmacological treatments have been considered as potential solutions, however, these alternatives have provided quite limited efficacy due to the short-term effect on pain management and inability to restore damaged tissue. The emergence of novel therapeutic alternatives such as the application of orthobiologics, particularly bone marrow aspirate (BMA) clot, have bestowed medical experts with considerable optimism as evidenced by the significant results found in numerous studies addressed in this manuscript. Although other products have been proposed for the treatment of musculoskeletal injuries, the peculiar interest in BMA, fibrin clot and associated fibrinolytic mechanisms continues to expand. BMA is a rich source of various cellular and molecular components which have demonstrated positive effects on tissue regeneration in many in vitro and in vivo models of musculoskeletal injuries. In addition to being able to undergo self-renewal and differentiation, the hematopoietic and mesenchymal stem cells present in this orthobiologic elicit key immunomodulatory and paracrine roles in inflammatory responses in tissue injury and drive the coagulation cascade towards tissue repair via different mechanisms. Although promising, these complex regenerative mechanisms have not yet been fully elucidated.
Collapse
|
43
|
Matsarskaia O, Roosen‐Runge F, Schreiber F. Multivalent ions and biomolecules: Attempting a comprehensive perspective. Chemphyschem 2020; 21:1742-1767. [PMID: 32406605 PMCID: PMC7496725 DOI: 10.1002/cphc.202000162] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/13/2020] [Indexed: 12/13/2022]
Abstract
Ions are ubiquitous in nature. They play a key role for many biological processes on the molecular scale, from molecular interactions, to mechanical properties, to folding, to self-organisation and assembly, to reaction equilibria, to signalling, to energy and material transport, to recognition etc. Going beyond monovalent ions to multivalent ions, the effects of the ions are frequently not only stronger (due to the obviously higher charge), but qualitatively different. A typical example is the process of binding of multivalent ions, such as Ca2+ , to a macromolecule and the consequences of this ion binding such as compaction, collapse, potential charge inversion and precipitation of the macromolecule. Here we review these effects and phenomena induced by multivalent ions for biological (macro)molecules, from the "atomistic/molecular" local picture of (potentially specific) interactions to the more global picture of phase behaviour including, e. g., crystallisation, phase separation, oligomerisation etc. Rather than attempting an encyclopedic list of systems, we rather aim for an embracing discussion using typical case studies. We try to cover predominantly three main classes: proteins, nucleic acids, and amphiphilic molecules including interface effects. We do not cover in detail, but make some comparisons to, ion channels, colloidal systems, and synthetic polymers. While there are obvious differences in the behaviour of, and the relevance of multivalent ions for, the three main classes of systems, we also point out analogies. Our attempt of a comprehensive discussion is guided by the idea that there are not only important differences and specific phenomena with regard to the effects of multivalent ions on the main systems, but also important similarities. We hope to bridge physico-chemical mechanisms, concepts of soft matter, and biological observations and connect the different communities further.
Collapse
Affiliation(s)
| | - Felix Roosen‐Runge
- Department of Biomedical Sciences and Biofilms-Research Center for Biointerfaces (BRCB), Faculty of Health and SocietyMalmö UniversitySweden
- Division of Physical ChemistryLund UniversitySweden
| | | |
Collapse
|
44
|
Kondiah PJ, Kondiah PPD, Choonara YE, Marimuthu T, Pillay V. A 3D Bioprinted Pseudo-Bone Drug Delivery Scaffold for Bone Tissue Engineering. Pharmaceutics 2020; 12:E166. [PMID: 32079221 PMCID: PMC7076403 DOI: 10.3390/pharmaceutics12020166] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/27/2020] [Accepted: 02/06/2020] [Indexed: 12/16/2022] Open
Abstract
A 3D bioprinted pseudo-bone drug delivery scaffold was fabricated to display matrix strength, matrix resilience, as well as porous morphology of healthy human bone. Computer-aided design (CAD) software was employed for developing the 3D bioprinted scaffold. Further optimization of the scaffold was undertaken using MATLAB® software and artificial neural networks (ANN). Polymers employed for formulating the 3D scaffold comprised of polypropylene fumarate (PPF), free radical polymerized polyethylene glycol- polycaprolactone (PEG-PCL-PEG), and pluronic (PF127). Simvastatin was incorporated into the 3D bioprinted scaffolds to further promote bone healing and repair properties. The 3D bioprinted scaffold was characterized for its chemical, morphological, mechanical, and in vitro release kinetics for evaluation of its behavior for application as an implantable scaffold at the site of bone fracture. The ANN-optimized 3D bioprinted scaffold displayed significant properties as a controlled release platform, demonstrating drug release over 20 days. The 3D bioprinted scaffold further displayed formation as a pseudo-bone matrix, using a human clavicle bone model, induced with a butterfly fracture. The strength of the pseudo-bone matrix, evaluated for its matrix hardness (MH) and matrix resilience (MR), was evaluated to be as strong as original bone, having a 99% MH and 98% MR property, to healthy human clavicle bones.
Collapse
Affiliation(s)
| | | | | | | | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa; (P.J.K.); (P.P.D.K.); (Y.E.C.); (T.M.)
| |
Collapse
|