1
|
Frączek W, Kotela A, Kotela I, Grodzik M. Nanostructures in Orthopedics: Advancing Diagnostics, Targeted Therapies, and Tissue Regeneration. MATERIALS (BASEL, SWITZERLAND) 2024; 17:6162. [PMID: 39769763 PMCID: PMC11677186 DOI: 10.3390/ma17246162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/08/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
Nanotechnology, delving into the realm of nanometric structures, stands as a transformative force in orthopedics, reshaping diagnostics, and numerous regenerative interventions. Commencing with diagnostics, this scientific discipline empowers accurate analyses of various diseases and implant stability, heralding an era of unparalleled precision. Acting as carriers for medications, nanomaterials introduce novel therapeutic possibilities, propelling the field towards more targeted and effective treatments. In arthroplasty, nanostructural modifications to implant surfaces not only enhance mechanical properties but also promote superior osteointegration and durability. Simultaneously, nanotechnology propels tissue regeneration, with nanostructured dressings emerging as pivotal elements in accelerating wound healing. As we navigate the frontiers of nanotechnology, ongoing research illuminates promising avenues for further advancements, assuring a future where orthopedic practices are not only personalized but also highly efficient, promising a captivating journey through groundbreaking innovations and tailored patient care.
Collapse
Affiliation(s)
- Wiktoria Frączek
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland
| | - Andrzej Kotela
- Faculty of Medicine, Collegium Medicum, Cardinal Stefan Wyszyński University, 01-938 Warsaw, Poland
| | - Ireneusz Kotela
- National Medical Institute of the Ministry of the Interior and Administration, 02-507 Warsaw, Poland
- Collegium Medicum, Jan Kochanowski University in Kielce, 25-369 Kielce, Poland
| | - Marta Grodzik
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences (WULS-SGGW), 02-787 Warsaw, Poland
| |
Collapse
|
2
|
Schwager JM, Di Maggio N, Grosso A, Rasadurai A, Minder N, Hubbell JA, Kappos EA, Schaefer DJ, Briquez PS, Banfi A, Burger MG. Semaphorin 3A promotes the long-term persistence of human SVF-derived microvascular networks in engineered grafts. Front Bioeng Biotechnol 2024; 12:1396450. [PMID: 39234267 PMCID: PMC11371724 DOI: 10.3389/fbioe.2024.1396450] [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: 03/05/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024] Open
Abstract
Introduction The stromal vascular fraction (SVF) of human adipose tissue is an attractive cell source for engineering grafts with intrinsic vascularization potential, as it is rich in vasculogenic progenitors. However, in order to maintain their functional perfusion it is important to promote the in vivo stabilization of newly assembled microvascular networks. We previously found that Semaphorin 3A (Sema3A) promotes the rapid stabilization of new blood vessels induced by VEGF overexpression in skeletal muscle. Here we investigated whether Sema3A could promote the assembly, connection to circulation and persistence of human SVF-derived microvascular networks in engineered grafts. Methods Recombinant Sema3A was engineered with a transglutaminase substrate sequence (TG-Sema3A) to allow cross-linking into fibrin hydrogels. Grafts were prepared with freshly isolated human SVF cells in fibrin hydrogels decorated with 0, 0.1 or 100 μg/ml TG-Sema3A and implanted subcutaneously in immune-deficient mice. Results After 1 week in vivo, the assembly of human-derived networks was similar in all conditions. The outer part of the grafts was populated by blood vessels of both human and mouse origin, which formed abundant hybrid structures within a common basal lamina. About 90% of human-derived blood vessels were functionally connected to the host circulation in all conditions. However, in the control samples human vessels were unstable. In fact, they significantly regressed by 6 weeks and could no longer be found by 12 weeks. In contrast, a low Sema3A dose (0.1 μg/ml) promoted further human vascular expansion by about 2-fold at 6 weeks and protected them from regression until 12 weeks. From a mechanistic point of view, the stabilization of SVF-derived vessels by 0.1 μg/ml of Sema3A correlated with the recruitment of a specific population of monocytes expressing its receptor Neuropilin-1. Discussion In conclusion, Sema3A is a potent stimulator of in vivo long-term persistence of microvascular networks derived from human SVF. Therefore, decoration of matrices with Sema3a can be envisioned to promote the functional support of tissue engineered grafts.
Collapse
Affiliation(s)
- Juan M Schwager
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| | - Nunzia Di Maggio
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Andrea Grosso
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Abeelan Rasadurai
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Nadja Minder
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
| | - Jeffrey A Hubbell
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, United States
| | - Elisabeth A Kappos
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
- Department of Clinical Research, Medical Faculty, University of Basel, Basel, Switzerland
| | - Dirk J Schaefer
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| | - Priscilla S Briquez
- Department of General and Visceral Surgery, Medical Center-University of Freiburg, Freiburg, Germany
| | - Andrea Banfi
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| | - Maximilian G Burger
- Regenerative Angiogenesis Laboratory, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Basel, Switzerland
| |
Collapse
|
3
|
Stanciugelu SI, Patrascu JM, Florescu S, Marian C. Sticky Bone as a New Type of Autologous Bone Grafting in Schatzker Type II Tibial Plateau Fracture Case Report. Life (Basel) 2024; 14:1042. [PMID: 39202784 PMCID: PMC11355391 DOI: 10.3390/life14081042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Schatzker type II fractures usually need to be grafted. Autograft bone from the iliac crest represents the gold standard, but it comes with high rates of morbidity on the donor side. Sticky bone is one of the regenerative therapies that aims to find new solutions to treat bone defects and to overcome the limitation of conventional options regarding bone grafts, due to their content in growth factors, which offer osteo-induction and osteo-conduction properties. Notably, regenerative dentistry has been at the forefront of applying these products in bone regeneration, demonstrating that PRF produces a highly promising "sticky bone" when combined with bone chips. To the best of our knowledge, this grafting technique has not been used in the orthopedic field to date. METHODS The subject was a 53-year-old woman with a Schatzker type II tibial plateau fracture, for which a new autologous bone grafting technique, i.e., sticky bone, was used for the treatment of the fracture. RESULTS This case reports the effectiveness of sticky bone as autologous bone graft used in Shatzker type II tibial plateau fracture. As an indispensable component of regenerative medicine, it seems to be an ideal biologic graft with a fibrin-rich structure that provides effective treatment in impressed tibial plateau fractures. CONCLUSION Sticky bone showed promising results and should be considered in the future as an appropriate bone implant.
Collapse
Affiliation(s)
- Stefan Iulian Stanciugelu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
- “Pius Brinzeu” Emergency Clinical County Hospital, Bld. Liviu Rebreanu, No. 156, 300723 Timisoara, Romania;
- Orthopedics II Research Center, “Pius Brinzeu” Emergency Clinical County Hospital, Bld. Liviu Rebreanu, No. 156, 300723 Timisoara, Romania
| | - Jenel Marian Patrascu
- Doctoral School, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
- Prof. Univ. Dr. Teodor Șora Research Centre, Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Department of Orthopedics and Traumatology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Sorin Florescu
- “Pius Brinzeu” Emergency Clinical County Hospital, Bld. Liviu Rebreanu, No. 156, 300723 Timisoara, Romania;
- Prof. Univ. Dr. Teodor Șora Research Centre, Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania
- Department of Orthopedics and Traumatology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Catalin Marian
- Department of Biochemistry and Pharmacology, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
- Center for Complex Networks Science, “Victor Babes” University of Medicine and Pharmacy, Eftimie Murgu Square 2, 300041 Timisoara, Romania
| |
Collapse
|
4
|
Zou Y, Mei X, Wang X, Zhang X, Wang X, Xiang W, Lu N. Fibrin-konjac glucomannan-black phosphorus hydrogel scaffolds loaded with nasal ectodermal mesenchymal stem cells accelerated alveolar bone regeneration. BMC Oral Health 2024; 24:878. [PMID: 39095803 PMCID: PMC11297757 DOI: 10.1186/s12903-024-04649-0] [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: 03/04/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Effective treatments for the alveolar bone defect remain a major concern in dental therapy. The objectives of this study were to develop a fibrin and konjac glucomannan (KGM) composite hydrogel as scaffolds for the osteogenesis of nasal mucosa-derived ectodermal mesenchymal stem cells (EMSCs) for the regeneration of alveolar bone defect, and to investigate the osteogenesis-accelerating effects of black phosphorus nanoparticles (BPNs) embedded in the hydrogels. METHODS Primary EMSCs were isolated from rat nasal mucosa and used for the alveolar bone recovery. Fibrin and KGM were prepared in different ratios for osteomimetic hydrogel scaffolds, and the optimal ratio was determined by mechanical properties and biocompatibility analysis. Then, the optimal hydrogels were integrated with BPNs to obtain BPNs/fibrin-KGM hydrogels, and the effects on osteogenic EMSCs in vitro were evaluated. To explore the osteogenesis-enhancing effects of hydrogels in vivo, the BPNs/fibrin-KGM scaffolds combined with EMSCs were implanted to a rat model of alveolar bone defect. Micro-computed tomography (CT), histological examination, real-time quantitative polymerase chain reaction (RT-qPCR) and western blot were conducted to evaluate the bone morphology and expression of osteogenesis-related genes of the bone regeneration. RESULTS The addition of KGM improved the mechanical properties and biodegradation characteristics of the fibrin hydrogels. In vitro, the BPNs-containing compound hydrogel was proved to be biocompatible and capable of enhancing the osteogenesis of EMSCs by upregulating the mineralization and the activity of alkaline phosphatase. In vivo, the micro-CT analysis and histological evaluation demonstrated that rats implanted EMSCs-BPNs/fibrin-KGM hydrogels exhibited the best bone reconstruction. And compared to the model group, the expression of osteogenesis genes including osteopontin (Opn, p < 0.0001), osteocalcin (Ocn, p < 0.0001), type collagen (Col , p < 0.0001), bone morphogenetic protein-2 (Bmp2, p < 0.0001), Smad1 (p = 0.0006), and runt-related transcription factor 2 (Runx2, p < 0.0001) were all significantly upregulated. CONCLUSIONS EMSCs/BPNs-containing fibrin-KGM hydrogels accelerated the recovery of the alveolar bone defect in rats by effectively up-regulating the expression of osteogenesis-related genes, promoting the formation and mineralisation of bone matrix.
Collapse
Affiliation(s)
- Yin Zou
- Department of Stomatology, Affiliated Children's Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China
| | - Xue Mei
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China
| | - Xinhe Wang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China
| | - Xuan Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China
| | - Xun Wang
- Jiangnan University Medical Center, Wuxi, Jiangsu Province, People's Republic of China
| | - Wen Xiang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China
| | - Naiyan Lu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China.
| |
Collapse
|
5
|
Bahreini M, Moghaddam MM, Ghorbani M, Nourani MR, Mirnejad R. Antimicrobial peptide-fibrin glue mixture for treatment of methicillin-resistant Staphylococcus aureus-infected wounds. Ther Deliv 2024; 15:577-591. [PMID: 39011599 PMCID: PMC11412140 DOI: 10.1080/20415990.2024.2369497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Aim: This study was conducted to investigate the effect of fibrin glue-CM11 antibacterial peptide mixture (FG-P) on the healing of infected wounds in vivo.Materials & methods: We formulated a mixture of FG-P and evaluated its antimicrobial activity in vitro against multidrug-resistant (MDR) bacteria involved in wound infection as well as its healing effect on wound infected by methicillin-resistant S. aureus (MRSA) in vivo.Results: The peptide had an MIC of 8 μg/ml against all bacteria isolates. Growth inhibition zones were evident for FG-P compared with FG. The in vivo study showed that the FG-P could be significantly effective in healing the MRSA-infected wound.Conclusion: The use of FG-P mixture is a very suitable option for treating infected wounds.
Collapse
Affiliation(s)
- Mehran Bahreini
- Student Research Committee, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mehrdad Moosazadeh Moghaddam
- Tissue Engineering & Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Masoud Ghorbani
- Tissue Engineering & Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Nourani
- Tissue Engineering & Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Reza Mirnejad
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Iacono V, Padovani L, Qordja F, De Berardinis L, Screpis D, Gigante AP, Zorzi C. Surgical and Biological Treatment with a Platelet-Rich Fibrin Matrix for Patellar Tendinopathy: Clinical Outcomes and Return to Sport at 2-Year Follow-Up. J Pers Med 2024; 14:567. [PMID: 38929787 PMCID: PMC11204417 DOI: 10.3390/jpm14060567] [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: 04/27/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Patellar tendinopathy (PT) involves anterior knee pain and functional. Platelet-rich fibrin matrix (PRFM) is a promising biological therapy for tendinopathies. We examined a cohort of PT patients treated with tendon debridement and autologous PRFM at the 24-month follow-up to assess whether the combined treatment facilitated return to sports and yielded satisfactory clinical and functional scores. METHODS Baseline and 24-month visual analogue scale (VAS), Victorian Institute of Sport Assessment Scale for Patellar Tendinopathy (VISA-P), Tegner Activity Scale (TAS), and Blazina scores were compared to evaluate treatment effectiveness. The Friedman test was used to compare repeated observations of VAS, VISA-P, TAS, and Blazina Score values. Return to sport rate, Tampa Scale of Kinesiophobia (TKS) score and patient satisfaction were collected at 24 months. RESULTS The postoperative clinical scores demonstrated significant improvement compared with their preoperative values (all p < 0.001). Specifically, the VISA-P score was 80.32 (±20.58), 92.10% of patients had resumed sports activities and patient satisfaction was 9.21 (±1.21) at 24 months. CONCLUSIONS Surgical debridement and autologous PRFM application in patients with chronic PT resulted in a higher rate of return to sports when compared to solely surgical treatment, significantly improved clinical outcomes and excellent patient satisfaction at 24 months.
Collapse
Affiliation(s)
- Venanzio Iacono
- Department of Orthopaedics, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy; (V.I.); (L.P.); (D.S.); (C.Z.)
| | - Luca Padovani
- Department of Orthopaedics, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy; (V.I.); (L.P.); (D.S.); (C.Z.)
| | - Fjorela Qordja
- Clinical Orthopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.D.B.); (A.P.G.)
| | - Luca De Berardinis
- Clinical Orthopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.D.B.); (A.P.G.)
| | - Daniele Screpis
- Department of Orthopaedics, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy; (V.I.); (L.P.); (D.S.); (C.Z.)
| | - Antonio Pompilio Gigante
- Clinical Orthopaedics, Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, 60126 Ancona, Italy; (L.D.B.); (A.P.G.)
| | - Claudio Zorzi
- Department of Orthopaedics, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar, Italy; (V.I.); (L.P.); (D.S.); (C.Z.)
| |
Collapse
|
7
|
Bauso LV, La Fauci V, Longo C, Calabrese G. Bone Tissue Engineering and Nanotechnology: A Promising Combination for Bone Regeneration. BIOLOGY 2024; 13:237. [PMID: 38666849 PMCID: PMC11048357 DOI: 10.3390/biology13040237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/27/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024]
Abstract
Large bone defects are the leading contributor to disability worldwide, affecting approximately 1.71 billion people. Conventional bone graft treatments show several disadvantages that negatively impact their therapeutic outcomes and limit their clinical practice. Therefore, much effort has been made to devise new and more effective approaches. In this context, bone tissue engineering (BTE), involving the use of biomaterials which are able to mimic the natural architecture of bone, has emerged as a key strategy for the regeneration of large defects. However, although different types of biomaterials for bone regeneration have been developed and investigated, to date, none of them has been able to completely fulfill the requirements of an ideal implantable material. In this context, in recent years, the field of nanotechnology and the application of nanomaterials to regenerative medicine have gained significant attention from researchers. Nanotechnology has revolutionized the BTE field due to the possibility of generating nanoengineered particles that are able to overcome the current limitations in regenerative strategies, including reduced cell proliferation and differentiation, the inadequate mechanical strength of biomaterials, and poor production of extrinsic factors which are necessary for efficient osteogenesis. In this review, we report on the latest in vitro and in vivo studies on the impact of nanotechnology in the field of BTE, focusing on the effects of nanoparticles on the properties of cells and the use of biomaterials for bone regeneration.
Collapse
Affiliation(s)
- Luana Vittoria Bauso
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (V.L.F.); (C.L.)
| | | | | | - Giovanna Calabrese
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98168 Messina, Italy; (V.L.F.); (C.L.)
| |
Collapse
|
8
|
Almalki WH. An Up-to-date Review on Protein-based Nanocarriers in the Management of Cancer. Curr Drug Deliv 2024; 21:509-524. [PMID: 37165498 DOI: 10.2174/1567201820666230509101020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 02/19/2023] [Accepted: 02/27/2023] [Indexed: 05/12/2023]
Abstract
BACKGROUND A big health issue facing the world's population is cancer. An alarming increase in cancer patients was anticipated by worldwide demographic statistics, which showed that the number of patients with different malignancies was rapidly increasing. By 2025, probably 420 million cases were projected to be achieved. The most common cancers diagnosed are breast, colorectal, prostate, and lung. Conventional treatments, such as surgery, chemotherapy, and radiation therapy, have been practiced. OBJECTIVE In recent years, the area of cancer therapy has changed dramatically with expanded studies on the molecular-level detection and treatment of cancer. Recent advances in cancer research have seen significant advances in therapies such as chemotherapy and immunotherapy, although both have limitations in effectiveness and toxicity. METHODS The development of nanotechnology for anticancer drug delivery has developed several potentials as nanocarriers, which may boost the pharmacokinetic and pharmacodynamic effects of the drug product and substantially reduce the side effects. RESULTS The advancement in non-viral to viral-based protein-based nanocarriers for treating cancer has earned further recognition in this respect. Many scientific breakthroughs have relied on protein-based nanocarriers, and proteins are essential organic macromolecules for life. It allows targeted delivery of passive or active tumors using non-viral-based protein-based nanocarriers to viral-based protein nanocarriers. When targeting cancer cells, both animal and plant proteins may be used in a formulation process to create self-assembled viruses and platforms that can successfully eradicate metastatic cancer cells. CONCLUSION This review, therefore, explores in depth the applications of non-viral to viral proteinbased noncarriers with a specific focus on intracellular drug delivery and anti-cancer drug targeting ability.
Collapse
Affiliation(s)
- Waleed H Almalki
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Aal-qura University, Saudi Arabia
| |
Collapse
|
9
|
Bindi B, Perioli A, Melo P, Mattu C, Ferreira AM. Bioinspired Collagen/Hyaluronic Acid/Fibrin-Based Hydrogels for Soft Tissue Engineering: Design, Synthesis, and In Vitro Characterization. J Funct Biomater 2023; 14:495. [PMID: 37888160 PMCID: PMC10607851 DOI: 10.3390/jfb14100495] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/12/2023] [Accepted: 09/16/2023] [Indexed: 10/28/2023] Open
Abstract
A major challenge for future drug development comprises finding alternative models for drug screening. The use of animal models in research is highly controversial, with an ongoing debate on their ethical acceptability. Also, animal models are often poorly predictive of therapeutic outcomes due to the differences between animal and human physiological environments. In this study, we aimed to develop a biomimetic hydrogel that replicates the composition of skin for potential use in in vitro modeling within tissue engineering. The hydrogel was fabricated through the crosslinking of collagen type I, hyaluronic acid, four-arm PEG succinimidyl glutarate (4S-StarPEG), and fibrinogen. Various ratios of these components were systematically optimized to achieve a well-interconnected porosity and desirable rheological properties. To evaluate the hydrogel's cytocompatibility, fibroblasts were embedded within the matrix. The resulting hydrogel exhibited promising properties as a scaffold, also facilitating the growth of and proliferation of the cells. This biomimetic hydrogel holds great potential for tissue engineering applications, particularly in skin regeneration and cancer research. The study used melanoma spheroids fabricated using the 96-round bottom well plate method as a potential application. The results demonstrate that the developed hydrogels allowed the maintenance of spheroid integrity and viability, meaning it has a promising use as a three-dimensional in vitro model of melanoma for both tissue engineering and drug screening applications.
Collapse
Affiliation(s)
- Bianca Bindi
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Annalisa Perioli
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Priscila Melo
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Ana Marina Ferreira
- School of Engineering, Newcastle University, Newcastle upon Tyne NE1 7RU, UK
| |
Collapse
|
10
|
Morey M, Larrañaga A, Abbah SA, Bohara R, Aljaabary A, Pandit A. Glucose-Responsive Fibrin Hydrogel-Based Multimodal Nucleic Acid Delivery System. Adv Biol (Weinh) 2023; 7:e2300161. [PMID: 37401646 DOI: 10.1002/adbi.202300161] [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: 04/27/2023] [Indexed: 07/05/2023]
Abstract
Nucleic acid therapy has emerged as a potential alternative for promoting wound healing by gene expression modification. On the other hand, protecting the nucleic acid payload from degradation, efficient bioresponsive delivery and effective transfection into cells remain challenging. A glucose-responsive gene delivery system for treating diabetic wounds would be advantageous as it would be responsive to the underlying pathology giving a regulated payload delivery with fewer side effects. Herein a GOx-based glucose-responsive delivery system is designed based on fibrin-coated polymeric microcapsules (FCPMC) using the layer-by-layer (LbL) approach that simultaneously delivers two nucleic acids in diabetic wounds. The designed FCPMC displays an ability to effectively load many nucleic acids in polyplexes and release it over a prolonged period with no cytotoxic effects seen in in vitro studies. Furthermore, the developed system does not show any undesired effects in vivo. When applied to wounds in genetically diabetic db/db mice, the fabricated system on its own improves reepithelialization and angiogenesis while decreasing inflammation. Also, key proteins involved in the wound healing process, i.e., Actn2, MYBPC1, and desmin, are upregulated in the glucose-responsive fibrin hydrogel (GRFHG) treated group of animals. In conclusion, the fabricated hydrogel promotes wound healing. Furthermore, the system may be encapsulated with various therapeutic nucleic acids that aid wound healing.
Collapse
Affiliation(s)
- Mangesh Morey
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Aitor Larrañaga
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Sunny Akogwu Abbah
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Raghvendra Bohara
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Amal Aljaabary
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
11
|
Cui JT, Wang XY, Mu XD, Huang M, Wang YD, Luo Q, He HX. Bone marrow stromal cell-derived exosome combinate with fibrin on tantalum coating titanium implant accelerates osseointegration. Front Bioeng Biotechnol 2023; 11:1198545. [PMID: 37496851 PMCID: PMC10367419 DOI: 10.3389/fbioe.2023.1198545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/20/2023] [Indexed: 07/28/2023] Open
Abstract
This study aims to present a sustainably releasing system of exosomes-fibrin combinate loaded on tantalum-coating titanium implants. We hope to investigate potential effects of the system on osseointegration between tantalum coating titanium implants and its surrounding bone tissue. Exosomes derived from rabbit bone marrow stromal cells (rBMSCs) and fibrin were deposited onto the micro-nanostructure tantalum coating surface (Ta + exo + FI) and compared to control groups, including tantalum coating (Ta), tantalum coating loaded exosomes (Ta + exo) and tantalum coating loaded fibrin (Ta + FI). The optimal concentration of loading exosomes, exosomes uptake capacity by BMSCs, and the effect of controlled-release by fibrin were assessed by laser scanning confocal microscope (LCSM) and microplate reader. The optimal concentration of exosomes was 1 μg/μL. Adhesion, proliferation, and osteogenic differentiation ability of BMSCs on different materials were assessed in vitro. Finally, osseointegrative capacity of Ta, Ta + exo, Ta + FI, Ta + exo + FI implants in rabbit tibia were respectively evaluated with histology and bone-implant contact ratio (BIC%). It was demonstrated that exosome sustained-release system with fibrin loading on the tantalum coating was successfully established. Fibrin contribute to exosomes release extension from 2d to 6d. Furthermore, Ta + exo + FI significantly promoted adhesion, proliferation, and osteogenic differentiation of BMSCs. In vivo, the implants in Ta + exo + FI group displayed the highest osseointegrative capability than those in other groups. It is concluded that this exosome delivery system on the implants may be an effective way for tantalum coating titanium implants to promote osseointegration between implant and its surrounding bone tissue.
Collapse
Affiliation(s)
- Jian-Tong Cui
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Shannxi Provincial Crops Hospital of Chinese People’s Armed Police Forces, Xian, China
| | - Xin-Yuan Wang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Xiao-Dan Mu
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Meng Huang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Ya-Di Wang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Qiang Luo
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hui-Xia He
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
12
|
Stahn L, Rasińska J, Dehne T, Schreyer S, Hakus A, Gossen M, Steiner B, Hemmati-Sadeghi S. Sleeping Beauty transposon system for GDNF overexpression of entrapped stem cells in fibrin hydrogel in a rat model of Parkinson's disease. Drug Deliv Transl Res 2023; 13:1745-1765. [PMID: 36853436 PMCID: PMC10125957 DOI: 10.1007/s13346-023-01289-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2022] [Indexed: 03/01/2023]
Abstract
There is currently no causal treatment available for Parkinson's disease (PD). However, the use of glial cell line-derived neurotrophic factor (GDNF) to provide regenerative effects for neurons is promising. Such approaches require translational delivery systems that are functional in diseased tissue. To do so, we used a non-viral Sleeping Beauty (SB) transposon system to overexpress GDNF in adipose tissue-derived mesenchymal stromal cells (adMSCs). Entrapment of cells in fibrin hydrogel was used to boost potential neurorestorative effects. Functional GDNF-adMSCs were able to secrete 1066.8 ± 169.4 ng GDNF/120,000 cells in vitro. The GDNF-adMSCs were detectable for up to 1 month after transplantation in a mild 6-hydroxydopamine (6-OHDA) hemiparkinson male rat model. Entrapment of GDNF-adMSCs enabled GDNF secretion in surrounding tissue in a more concentrated manner, also tending to prolong GDNF secretion relatively. GDNF-adMSCs entrapped in hydrogel also led to positive immunomodulatory effects via an 83% reduction of regional IL-1β levels compared to the non-entrapped GDNF-adMSC group after 1 month. Furthermore, GDNF-adMSC-treated groups showed higher recovery of tyrosine hydroxylase (TH)-expressing cells, indicating a neuroprotective function, although this was not strong enough to show significant improvement in motor performance. Our findings establish a promising GDNF treatment system in a PD model. Entrapment of GDNF-adMSCs mediated positive immunomodulatory effects. Although the durability of the hydrogel needs to be extended to unlock its full potential for motor improvements, the neuroprotective effects of GDNF were evident and safe. Further motor behavioral tests and other disease models are necessary to evaluate this treatment option adequately.
Collapse
Affiliation(s)
- Laura Stahn
- Department of Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Justyna Rasińska
- Department of Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Tilo Dehne
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology & Clinical Immunology, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Stefanie Schreyer
- Department of Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Aileen Hakus
- Department of Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Manfred Gossen
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Institute of Active Polymers, Helmholtz-Zentrum Hereon, 21502 Teltow, Germany
| | - Barbara Steiner
- Department of Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
| | - Shabnam Hemmati-Sadeghi
- Department of Neurology, Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, 10115 Berlin, Germany
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies, Department of Rheumatology & Clinical Immunology, Charité – Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
13
|
Wang J, Song Y, Xie W, Zhao J, Wang Y, Yu W. Therapeutic angiogenesis based on injectable hydrogel for protein delivery in ischemic heart disease. iScience 2023; 26:106577. [PMID: 37192972 PMCID: PMC10182303 DOI: 10.1016/j.isci.2023.106577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Ischemic heart disease (IHD) remains the leading cause of death and disability worldwide and leads to myocardial necrosis and negative myocardial remodeling, ultimately leading to heart failure. Current treatments include drug therapy, interventional therapy, and surgery. However, some patients with severe diffuse coronary artery disease, complex coronary artery anatomy, and other reasons are unsuitable for these treatments. Therapeutic angiogenesis stimulates the growth of the original blood vessels by using exogenous growth factors to generate more new blood vessels, which provides a new treatment for IHD. However, direct injection of these growth factors can cause a short half-life and serious side effects owing to systemic spread. Therefore, to overcome this problem, hydrogels have been developed for temporally and spatially controlled delivery of single or multiple growth factors to mimic the process of angiogenesis in vivo. This paper reviews the mechanism of angiogenesis, some important bioactive molecules, and natural and synthetic hydrogels currently being applied for bioactive molecule delivery to treat IHD. Furthermore, the current challenges of therapeutic angiogenesis in IHD and its potential solutions are discussed to facilitate real translation into clinical applications in the future.
Collapse
Affiliation(s)
- Junke Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Yancheng Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
| | - Wenjie Xie
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Shandong, Qingdao, Shandong 26000, China
| | - Jiang Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ying Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong 26000, China
- Corresponding author
| | - Wenzhou Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26003, China
- Corresponding author
| |
Collapse
|
14
|
Griffanti G, McKee MD, Nazhat SN. Mineralization of Bone Extracellular Matrix-like Scaffolds Fabricated as Silk Sericin-Functionalized Dense Collagen–Fibrin Hybrid Hydrogels. Pharmaceutics 2023; 15:pharmaceutics15041087. [PMID: 37111573 PMCID: PMC10142947 DOI: 10.3390/pharmaceutics15041087] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/20/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023] Open
Abstract
The design of hydrogels that combine both the biochemical cues needed to direct seeded cellular functions and mineralization to provide the structural and mechanical properties approaching those of mineralized native bone extracellular matrix (ECM) represents a significant challenge in bone tissue engineering. While fibrous hydrogels constituting of collagen or fibrin (and their hybrids) can be considered as scaffolds that mimic to some degree native bone ECM, their insufficient mechanical properties limit their application. In the present study, an automated gel aspiration–ejection (automated GAE) method was used to generate collagen–fibrin hybrid gel scaffolds with micro-architectures and mechanical properties approaching those of native bone ECM. Moreover, the functionalization of these hybrid scaffolds with negatively charged silk sericin accelerated their mineralization under acellular conditions in simulated body fluid and modulated the proliferation and osteoblastic differentiation of seeded MC3T3-E1 pre-osteoblastic cells. In the latter case, alkaline phosphatase activity measurements indicated that the hybrid gel scaffolds with seeded cells showed accelerated osteoblastic differentiation, which in turn led to increased matrix mineralization. In summary, the design of dense collagen–fibrin hybrid gels through an automated GAE process can provide a route to tailoring specific biochemical and mechanical properties to different types of bone ECM-like scaffolds, and can provide a model to better understand cell–matrix interactions in vitro for bioengineering purposes.
Collapse
Affiliation(s)
- Gabriele Griffanti
- Department of Mining and Materials Engineering, McGill University, Montréal, QC H3A 0C5, Canada;
| | - Marc D. McKee
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montréal, QC H3A 0C7, Canada;
- Department of Anatomy and Cell Biology, McGill University, Montréal, QC H3A 0C7, Canada
| | - Showan N. Nazhat
- Department of Mining and Materials Engineering, McGill University, Montréal, QC H3A 0C5, Canada;
- Correspondence: ; Tel.: +514-398-5524; Fax: 514-398-4492
| |
Collapse
|
15
|
Ortiz-Arrabal O, Irastorza-Lorenzo A, Campos F, Martín-Piedra MÁ, Carriel V, Garzón I, Ávila-Fernández P, de Frutos MJ, Esteban E, Fernández J, Janer A, Campos A, Chato-Astrain J, Alaminos M. Fibrin and Marine-Derived Agaroses for the Generation of Human Bioartificial Tissues: An Ex Vivo and In Vivo Study. Mar Drugs 2023; 21:md21030187. [PMID: 36976236 PMCID: PMC10058299 DOI: 10.3390/md21030187] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/09/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Development of an ideal biomaterial for clinical use is one of the main objectives of current research in tissue engineering. Marine-origin polysaccharides, in particular agaroses, have been widely explored as scaffolds for tissue engineering. We previously developed a biomaterial based on a combination of agarose with fibrin, that was successfully translated to clinical practice. However, in search of novel biomaterials with improved physical and biological properties, we have now generated new fibrin-agarose (FA) biomaterials using 5 different types of agaroses at 4 different concentrations. First, we evaluated the cytotoxic effects and the biomechanical properties of these biomaterials. Then, each bioartificial tissue was grafted in vivo and histological, histochemical and immunohistochemical analyses were performed after 30 days. Ex vivo evaluation showed high biocompatibility and differences in their biomechanical properties. In vivo, FA tissues were biocompatible at the systemic and local levels, and histological analyses showed that biointegration was associated to a pro-regenerative process with M2-type CD206-positive macrophages. These results confirm the biocompatibility of FA biomaterials and support their clinical use for the generation of human tissues by tissue engineering, with the possibility of selecting specific agarose types and concentrations for applications requiring precise biomechanical properties and in vivo reabsorption times.
Collapse
Grants
- FIS PI20/0317 FIS PI20/0318 FIS PI21/0980 ICI19/00024 ICI21/00010 Spanish Plan Nacional de Investigación Científica, Desarrollo e Innovación Tecnológica (I+D+I) of the Spanish Ministry of Science and Innovation (Instituto de Salud Carlos III),
- PE-0395-2019 PI-0442-2019 Consejería de Salud y Familias, Junta de Andalucía, Spain
- IDI-20180052 Hispanagar SA, Burgos, Spain, through CDTI, Ministry of Science and Innovation, Spain, Pro-grama Operativo Plurirregional de Crecimiento Inteligente (CRIN)
- B-CTS-504-UGR20 B-CTS-450-UGR20 marco del Programa Operativo FEDER Andalucía 2014-2020, University of Granada and Conseje-ría de Transformación Económica, Industria, Conocimiento y Universidades
Collapse
Affiliation(s)
- Olimpia Ortiz-Arrabal
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain
- Doctoral Program in Biochemistry and Molecular Biology, University of Granada, E18016 Granada, Spain
| | - Ainhoa Irastorza-Lorenzo
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain
| | - Fernando Campos
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain
| | - Miguel Ángel Martín-Piedra
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain
| | - Víctor Carriel
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain
| | - Ingrid Garzón
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain
| | - Paula Ávila-Fernández
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain
| | | | | | | | | | - Antonio Campos
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain
| | - Jesús Chato-Astrain
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain
| | - Miguel Alaminos
- Tissue Engineering Group, Department of Histology, University of Granada and Instituto de Investigación Biosanitaria ibs.GRANADA, E18016 Granada, Spain
| |
Collapse
|
16
|
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
|
17
|
Blando S, Anchesi I, Mazzon E, Gugliandolo A. Can a Scaffold Enriched with Mesenchymal Stem Cells Be a Good Treatment for Spinal Cord Injury? Int J Mol Sci 2022; 23:ijms23147545. [PMID: 35886890 PMCID: PMC9319719 DOI: 10.3390/ijms23147545] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 12/10/2022] Open
Abstract
Spinal cord injury (SCI) is a worldwide highly crippling disease that can lead to the loss of motor and sensory neurons. Among the most promising therapies, there are new techniques of tissue engineering based on stem cells that promote neuronal regeneration. Among the different types of stem cells, mesenchymal stem cells (MSCs) seem the most promising. Indeed, MSCs are able to release trophic factors and to differentiate into the cell types that can be found in the spinal cord. Currently, the most common procedure to insert cells in the lesion site is infusion. However, this causes a low rate of survival and engraftment in the lesion site. For these reasons, tissue engineering is focusing on bioresorbable scaffolds to help the cells to stay in situ. Scaffolds do not only have a passive role but become fundamental for the trophic support of cells and the promotion of neuroregeneration. More and more types of materials are being studied as scaffolds to decrease inflammation and increase the engraftment as well as the survival of the cells. Our review aims to highlight how the use of scaffolds made from biomaterials enriched with MSCs gives positive results in in vivo SCI models as well as the first evidence obtained in clinical trials.
Collapse
|
18
|
Ze Y, Li Y, Huang L, Shi Y, Li P, Gong P, Lin J, Yao Y. Biodegradable Inks in Indirect Three-Dimensional Bioprinting for Tissue Vascularization. Front Bioeng Biotechnol 2022; 10:856398. [PMID: 35402417 PMCID: PMC8990266 DOI: 10.3389/fbioe.2022.856398] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/09/2022] [Indexed: 02/05/2023] Open
Abstract
Mature vasculature is important for the survival of bioengineered tissue constructs, both in vivo and in vitro; however, the fabrication of fully vascularized tissue constructs remains a great challenge in tissue engineering. Indirect three-dimensional (3D) bioprinting refers to a 3D printing technique that can rapidly fabricate scaffolds with controllable internal pores, cavities, and channels through the use of sacrificial molds. It has attracted much attention in recent years owing to its ability to create complex vascular network-like channels through thick tissue constructs while maintaining endothelial cell activity. Biodegradable materials play a crucial role in tissue engineering. Scaffolds made of biodegradable materials act as temporary templates, interact with cells, integrate with native tissues, and affect the results of tissue remodeling. Biodegradable ink selection, especially the choice of scaffold and sacrificial materials in indirect 3D bioprinting, has been the focus of several recent studies. The major objective of this review is to summarize the basic characteristics of biodegradable materials commonly used in indirect 3D bioprinting for vascularization, and to address recent advances in applying this technique to the vascularization of different tissues. Furthermore, the review describes how indirect 3D bioprinting creates blood vessels and vascularized tissue constructs by introducing the methodology and biodegradable ink selection. With the continuous improvement of biodegradable materials in the future, indirect 3D bioprinting will make further contributions to the development of this field.
Collapse
Affiliation(s)
- Yiting Ze
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanxi Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Linyang Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yixin Shi
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Peiran Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jie Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yang Yao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Fibrin-based factor delivery for therapeutic angiogenesis: friend or foe? Cell Tissue Res 2022; 387:451-460. [PMID: 35175429 PMCID: PMC8975770 DOI: 10.1007/s00441-022-03598-w] [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: 10/15/2021] [Accepted: 02/07/2022] [Indexed: 12/28/2022]
Abstract
Therapeutic angiogenesis aims at promoting the growth of blood vessels to restore perfusion in ischemic tissues or aid tissue regeneration. Vascular endothelial growth factor (VEGF) is the master regulator of angiogenesis in development, repair, and disease. However, exploiting VEGF for therapeutic purposes has been challenging and needs to take into account some key aspects of VEGF biology. In particular, the spatial localization of angiogenic signals within the extracellular matrix is crucial for physiological assembly and function of new blood vessels. Fibrin is the provisional matrix that is universally deposited immediately after injury and supports the initial steps of tissue regeneration. It provides therefore several ideal features as a substrate to promote therapeutic vascularization, especially through its ability to present growth factors in their physiological matrix-bound state and to modulate their availability for signaling. Here, we provide an overview of fibrin uses as a tissue-engineering scaffold material and as a tunable platform to finely control dose and duration of delivery of recombinant factors in therapeutic angiogenesis. However, in some cases, fibrin has also been associated with undesirable outcomes, namely the promotion of fibrosis and scar formation that actually prevent physiological tissue regeneration. Understanding the mechanisms that tip the balance between the pro- and anti-regenerative functions of fibrin will be the key to fully exploit its therapeutic potential.
Collapse
|
20
|
Fatima MT, Islam Z, Ahmad E, Hoque M, Yamin M. Plasma Bead Entrapped Liposomes as a Potential Drug Delivery System to Combat Fungal Infections. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27031105. [PMID: 35164370 PMCID: PMC8840493 DOI: 10.3390/molecules27031105] [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: 12/20/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 11/17/2022]
Abstract
Fibrin-based systems offer promises in drug and gene delivery as well as tissue engineering. We established earlier a fibrin-based plasma beads (PB) system as an efficient carrier of drugs and antigens. In the present work, attempts were made to further improve its therapeutic efficacy exploiting innovative ideas, including the use of plasma alginate composite matrices, proteolytic inhibitors, cross linkers, and dual entrapment in various liposomal formulations. In vitro efficacy of the different formulations was examined. Pharmacokinetics of the formulations encapsulating Amphotericin B (AmpB), an antifungal compound, were investigated in Swiss albino mice. While administration of the free AmpB led to its rapid elimination (<72 h), PB/liposome-PB systems were significantly effective in sustaining AmpB release in the circulation (>144 h) and its gradual accumulation in the vital organs, also compared to the liposomal formulations alone. Interestingly, the slow release of AmpB from PB was unusual compared to other small molecules in our earlier findings, suggesting strong interaction with plasma proteins. Molecular interaction studies of bovine serum albumin constituting approximately 60% of plasma with AmpB using isothermal titration calorimetry and in silico docking verify these interactions, explaining the slow release of AmpB entrapped in PB alone. The above findings suggest that PB/liposome-PB could be used as safe and effective delivery systems to combat fungal infections in humans.
Collapse
Affiliation(s)
- Munazza Tamkeen Fatima
- Interdisciplinary Biotechnology Unit (IBU), Aligarh Muslim University (AMU), Aligarh 202002, India; (E.A.); (M.H.)
- Correspondence:
| | - Zeyaul Islam
- Laboratório Nacional de Biociências (LNBio), Centro Nacional de Pesquisa em Energia e Materiais (CNPEM), Campinas, Sao Paulo 13083-970, Brazil;
| | - Ejaj Ahmad
- Interdisciplinary Biotechnology Unit (IBU), Aligarh Muslim University (AMU), Aligarh 202002, India; (E.A.); (M.H.)
| | - Mehboob Hoque
- Interdisciplinary Biotechnology Unit (IBU), Aligarh Muslim University (AMU), Aligarh 202002, India; (E.A.); (M.H.)
| | - Marriam Yamin
- Functional and Molecular Biology, Biochemistry, UNICAMP, Campinas, Sao Paulo 13083-970, Brazil;
| |
Collapse
|
21
|
Safina I, Childress LT, Myneni SR, Vang KB, Biris AS. Cell-Biomaterial Constructs for Wound Healing and Skin Regeneration. Drug Metab Rev 2022; 54:63-94. [PMID: 35129408 DOI: 10.1080/03602532.2021.2025387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Over the years, conventional skin grafts, such as full-thickness, split-thickness, and pre-sterilized grafts from human or animal sources, have been at the forefront of skin wound care. However, these conventional grafts are associated with major challenges, including supply shortage, rejection by the immune system, and disease transmission following transplantation. Due to recent progress in nanotechnology and material sciences, advanced artificial skin grafts-based on the fundamental concepts of tissue engineering-are quickly evolving for wound healing and regeneration applications, mainly because they can be uniquely tailored to meet the requirements of specific injuries. Despite tremendous progress in tissue engineering, many challenges and uncertainties still face skin grafts in vivo, such as how to effectively coordinate the interaction between engineered biomaterials and the immune system to prevent graft rejection. Furthermore, in-depth studies on skin regeneration at the molecular level are lacking; as a consequence, the development of novel biomaterial-based systems that interact with the skin at the core level has also been slow. This review will discuss 1) the biological aspects of wound healing and skin regeneration, 2) important characteristics and functions of biomaterials for skin regeneration applications, and 3) synthesis and applications of common biomaterials for skin regeneration. Finally, the current challenges and future directions of biomaterial-based skin regeneration will be addressed.
Collapse
Affiliation(s)
- Ingrid Safina
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR 72204 USA
| | - Luke T Childress
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR 72204 USA
| | - Srinivas R Myneni
- Department of Periodontology, Stony Brook University, Stony Brook, NY 11794 USA
| | - Kieng Bao Vang
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR 72204 USA
| | - Alexandru S Biris
- Center for Integrative Nanotechnology Sciences, University of Arkansas at Little Rock, 2801 S. University Avenue, Little Rock, AR 72204 USA
| |
Collapse
|
22
|
Plasminogen-Loaded Fibrin Scaffold as Drug Delivery System for Wound Healing Applications. Pharmaceutics 2022; 14:pharmaceutics14020251. [PMID: 35213982 PMCID: PMC8879571 DOI: 10.3390/pharmaceutics14020251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/19/2021] [Accepted: 01/18/2022] [Indexed: 01/13/2023] Open
Abstract
Plasminogen is a protein involved in intravascular and extravascular fibrinolysis, as well as in wound healing, cell migration, tissue formation and angiogenesis. In recent years its role in healing of tympanic perforations has been demonstrated in plasminogen deficient mice. The aim of this work was to fabricate a fibrin-based drug delivery system able to provide a local and sustained release of plasminogen at the wound site. Initially, the biological activity of plasminogen was evaluated by in vitro experiments on cell cultures. A metabolic assay (MTT) was carried out on L929 mouse fibroblast to determine the concentration that does not affect cell viability, which turned out to be 64 nM. The effect of plasminogen on cell migration was evaluated through a scratch test on human keratinocytes: cells treated with 64 nM plasminogen showed faster scratch closure than in complete medium. Fibrin scaffold loaded with plasminogen was fabricated by a spray process. SEM analysis showed the typical nano-fibrillar structure of a fibrin scaffold. Tensile tests highlighted significantly higher value of the ultimate stress and strain of fibrin scaffold with respect to fibrin clot. The in-vitro release kinetic showed an initial plasminogen burst, after that the release slowed, reaching a plateau at 7 days. Plasminogen-loaded fibrin scaffold applied in full-thickness diabetic mouse lesions showed a significantly higher closure rate at 14 days than scaffold used as a reference material. Histological analysis demonstrated an improved reepithelization and collagen deposition in granulation tissue in mouse treated with plasminogen-loaded fibrin scaffold in comparison to unloaded fibrin scaffold. The obtained results demonstrated the suitability of the fibrin scaffold loaded with plasminogen as drug delivery system and suggest its use in wound healing applications, such as for the treatment of chronic diabeticulcers.
Collapse
|
23
|
Ortiz ADC, Fideles SOM, Pomini KT, Bellini MZ, Pereira EDSBM, Reis CHB, Pilon JPG, de Marchi MÂ, Trazzi BFDM, da Silva WS, da Cunha MR, Buchaim DV, Buchaim RL. Potential of Fibrin Glue and Mesenchymal Stem Cells (MSCs) to Regenerate Nerve Injuries: A Systematic Review. Cells 2022; 11:221. [PMID: 35053336 PMCID: PMC8773549 DOI: 10.3390/cells11020221] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 02/04/2023] Open
Abstract
Cell-based therapy is a promising treatment to favor tissue healing through less invasive strategies. Mesenchymal stem cells (MSCs) highlighted as potential candidates due to their angiogenic, anti-apoptotic and immunomodulatory properties, in addition to their ability to differentiate into several specialized cell lines. Cells can be carried through a biological delivery system, such as fibrin glue, which acts as a temporary matrix that favors cell-matrix interactions and allows local and paracrine functions of MSCs. Thus, the aim of this systematic review was to evaluate the potential of fibrin glue combined with MSCs in nerve regeneration. The bibliographic search was performed in the PubMed/MEDLINE, Web of Science and Embase databases, using the descriptors ("fibrin sealant" OR "fibrin glue") AND "stem cells" AND "nerve regeneration", considering articles published until 2021. To compose this review, 13 in vivo studies were selected, according to the eligibility criteria. MSCs favored axonal regeneration, remyelination of nerve fibers, as well as promoted an increase in the number of myelinated fibers, myelin sheath thickness, number of axons and expression of growth factors, with significant improvement in motor function recovery. This systematic review showed clear evidence that fibrin glue combined with MSCs has the potential to regenerate nervous system lesions.
Collapse
Affiliation(s)
- Adriana de Cássia Ortiz
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.)
| | - Simone Ortiz Moura Fideles
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.)
| | - Karina Torres Pomini
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marilia (UNIMAR), Marília 17525-902, Brazil; (E.d.S.B.M.P.); (D.V.B.)
| | - Márcia Zilioli Bellini
- Pro-Rectory of Research and Graduate Studies, University Center of Adamantina (UniFAI), Adamantina 17800-000, Brazil;
| | - Eliana de Souza Bastos Mazuqueli Pereira
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marilia (UNIMAR), Marília 17525-902, Brazil; (E.d.S.B.M.P.); (D.V.B.)
| | - Carlos Henrique Bertoni Reis
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.)
- UNIMAR Beneficent Hospital (HBU), Medical School, University of Marilia (UNIMAR), Marília 17525-160, Brazil;
| | - João Paulo Galletti Pilon
- UNIMAR Beneficent Hospital (HBU), Medical School, University of Marilia (UNIMAR), Marília 17525-160, Brazil;
- Postgraduate Program in Speech Therapy, Sao Paulo State University (UNESP—Univ Estadual Paulista), Marília 17525-900, Brazil
| | - Miguel Ângelo de Marchi
- Coordination of the Medical School, University Center of Adamantina (UniFAI), Adamantina 17800-000, Brazil;
| | | | | | - Marcelo Rodrigues da Cunha
- Interunit Postgraduate Program in Bioengineering (EESC/FMRP/IQSC), University of São Paulo (USP), São Carlos 13566-590, Brazil;
- Department of Morphology and Pathology, Jundiaí Medical School, Jundiaí 13202-550, Brazil
| | - Daniela Vieira Buchaim
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marilia (UNIMAR), Marília 17525-902, Brazil; (E.d.S.B.M.P.); (D.V.B.)
- Teaching and Research Coordination of the Medical School, University Center of Adamantina (UniFAI), Adamantina 17800-000, Brazil
| | - Rogerio Leone Buchaim
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.)
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, Brazil
| |
Collapse
|
24
|
Ortiz ADC, Fideles SOM, Pomini KT, Reis CHB, Bueno CRDS, Pereira EDSBM, Rossi JDO, Novais PC, Pilon JPG, Rosa Junior GM, Buchaim DV, Buchaim RL. Effects of Therapy with Fibrin Glue combined with Mesenchymal Stem Cells (MSCs) on Bone Regeneration: A Systematic Review. Cells 2021; 10:2323. [PMID: 34571972 PMCID: PMC8468169 DOI: 10.3390/cells10092323] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/26/2021] [Accepted: 08/31/2021] [Indexed: 12/17/2022] Open
Abstract
Cell therapy strategies using mesenchymal stem cells (MSCs) carried in fibrin glue have shown promising results in regenerative medicine. MSCs are crucial for tissue healing because they have angiogenic, anti-apoptotic and immunomodulatory properties, in addition to the ability to differentiate into several specialized cell lines. Fibrin sealant or fibrin glue is a natural polymer involved in the coagulation process. Fibrin glue provides a temporary structure that favors angiogenesis, extracellular matrix deposition and cell-matrix interactions. Additionally, fibrin glue maintains the local and paracrine functions of MSCs, providing tissue regeneration through less invasive clinical procedures. Thus, the objective of this systematic review was to assess the potential of fibrin glue combined with MSCs in bone or cartilage regeneration. The bibliographic search was performed in the PubMed/MEDLINE, LILACS and Embase databases, using the descriptors ("fibrin sealant" OR "fibrin glue") AND "stem cells" AND "bone regeneration", considering articles published until 2021. In this case, 12 preclinical and five clinical studies were selected to compose this review, according to the eligibility criteria. In preclinical studies, fibrin glue loaded with MSCs, alone or associated with bone substitute, significantly favored bone defects regeneration compared to scaffold without cells. Similarly, fibrin glue loaded with MSCs presented considerable potential to regenerate joint cartilage injuries and multiple bone fractures, with significant improvement in clinical parameters and absence of postoperative complications. Therefore, there is clear evidence in the literature that fibrin glue loaded with MSCs, alone or combined with bone substitute, is a promising strategy for treating lesions in bone or cartilaginous tissue.
Collapse
Affiliation(s)
- Adriana de Cássia Ortiz
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, SP, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.); (C.R.d.S.B.); (J.d.O.R.)
| | - Simone Ortiz Moura Fideles
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, SP, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.); (C.R.d.S.B.); (J.d.O.R.)
| | - Karina Torres Pomini
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, SP, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.); (C.R.d.S.B.); (J.d.O.R.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marilia (UNIMAR), Marília 17525-902, SP, Brazil; (E.d.S.B.M.P.); (P.C.N.); (J.P.G.P.); (D.V.B.)
| | - Carlos Henrique Bertoni Reis
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, SP, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.); (C.R.d.S.B.); (J.d.O.R.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marilia (UNIMAR), Marília 17525-902, SP, Brazil; (E.d.S.B.M.P.); (P.C.N.); (J.P.G.P.); (D.V.B.)
| | - Cleuber Rodrigo de Souza Bueno
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, SP, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.); (C.R.d.S.B.); (J.d.O.R.)
| | - Eliana de Souza Bastos Mazuqueli Pereira
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marilia (UNIMAR), Marília 17525-902, SP, Brazil; (E.d.S.B.M.P.); (P.C.N.); (J.P.G.P.); (D.V.B.)
| | - Jéssica de Oliveira Rossi
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, SP, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.); (C.R.d.S.B.); (J.d.O.R.)
| | - Paulo Cezar Novais
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marilia (UNIMAR), Marília 17525-902, SP, Brazil; (E.d.S.B.M.P.); (P.C.N.); (J.P.G.P.); (D.V.B.)
| | - João Paulo Galletti Pilon
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marilia (UNIMAR), Marília 17525-902, SP, Brazil; (E.d.S.B.M.P.); (P.C.N.); (J.P.G.P.); (D.V.B.)
| | - Geraldo Marco Rosa Junior
- Pro-rectory of Graduate Studies and Research, Sacred Heart University Center (UNISAGRADO), Bauru 17011-160, SP, Brazil;
| | - Daniela Vieira Buchaim
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, Postgraduate Department, University of Marilia (UNIMAR), Marília 17525-902, SP, Brazil; (E.d.S.B.M.P.); (P.C.N.); (J.P.G.P.); (D.V.B.)
- Department of Human Anatomy and Neuroanatomy, University Center of Adamantina (UniFAI), Medical School, Adamantina 17800-000, SP, Brazil
| | - Rogerio Leone Buchaim
- Department of Biological Sciences, Bauru School of Dentistry (FOB/USP), University of São Paulo, Bauru 17012-901, SP, Brazil; (A.d.C.O.); (S.O.M.F.); (K.T.P.); (C.H.B.R.); (C.R.d.S.B.); (J.d.O.R.)
- Graduate Program in Anatomy of Domestic and Wild Animals, Faculty of Veterinary Medicine and Animal Science, University of São Paulo (FMVZ/USP), São Paulo 05508-270, SP, Brazil
| |
Collapse
|
25
|
Uehara K, Zhao C, Gingery A, Thoreson AR, An KN, Amadio PC. The effect of fibrin formulation on cell migration in an in vitro tendon repair model. J Orthop Sci 2021; 26:902-907. [PMID: 32814661 PMCID: PMC7884481 DOI: 10.1016/j.jos.2020.07.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 05/18/2020] [Accepted: 07/15/2020] [Indexed: 02/09/2023]
Abstract
BACKGROUND The purpose of this study was to determine the effect of fibrinogen concentration on cell viability and migration in a tissue culture tendon healing model. METHODS Forty-eight canine flexor digitorum profundus tendons were randomly divided into three groups. In each group the tendons were lacerated and repaired augmented with a canine bone marrow stromal cell seeded fibrin interposition patch using either 5 mg/ml fibrinogen and 25 U/ml thrombin (physiological as a control), 40 mg/ml fibrinogen and 250 U/ml thrombin (low adhesive), or 80 mg/ml fibrinogen and 250 U/ml thrombin (high adhesive). The sutured tendons were cultured for two or four weeks. RESULTS Failure load was not significantly different among the groups. Cell-labeling staining showed that the stromal cells migrated across the gap in the control and low adhesive groups, but there was no cell migration in the high adhesive group at two weeks. CONCLUSION A high fibrinogen concentration in a fibrin patch or glue may impede early cell migration. LEVEL OF EVIDENCE Not applicable because this study was a laboratory study.
Collapse
Affiliation(s)
- Kosuke Uehara
- Orthopaedic Biomechanics and Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Chunfeng Zhao
- Orthopaedic Biomechanics and Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Anne Gingery
- Orthopaedic Biomechanics and Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Andrew R Thoreson
- Orthopaedic Biomechanics and Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Kai-Nan An
- Orthopaedic Biomechanics and Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Peter C Amadio
- Orthopaedic Biomechanics and Tendon and Soft Tissue Biology Laboratories, Division of Orthopedic Research, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
26
|
FITC-Dextran Release from Cell-Embedded Fibrin Hydrogels. Biomolecules 2021; 11:biom11020337. [PMID: 33672379 PMCID: PMC7926394 DOI: 10.3390/biom11020337] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
Fibrin hydrogel is a central biological material in tissue engineering and drug delivery applications. As such, fibrin is typically combined with cells and biomolecules targeted to the regenerated tissue. Previous studies have analyzed the release of different molecules from fibrin hydrogels; however, the effect of embedded cells on the release profile has yet to be quantitatively explored. This study focused on the release of Fluorescein isothiocyanate (FITC)-dextran (FD) 250 kDa from fibrin hydrogels, populated with different concentrations of fibroblast or endothelial cells, during a 48-h observation period. The addition of cells to fibrin gels decreased the overall release by a small percentage (by 7-15% for fibroblasts and 6-8% for endothelial cells) relative to acellular gels. The release profile was shown to be modulated by various cellular activities, including gel degradation and physical obstruction to diffusion. Cell-generated forces and matrix deformation (i.e., densification and fiber alignment) were not found to significantly influence the release profiles. This knowledge is expected to improve fibrin integration in tissue engineering and drug delivery applications by enabling predictions and ways to modulate the release profiles of various biomolecules.
Collapse
|
27
|
Jang CH, Cho GW, Song AJ. Effect of Bone Powder/Mesenchymal Stem Cell/BMP2/Fibrin Glue on Osteogenesis in a Mastoid Obliteration Model. In Vivo 2021; 34:1103-1110. [PMID: 32354898 DOI: 10.21873/invivo.11881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/02/2020] [Accepted: 03/05/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM This study aimed to prospectively compare the osteogenesis of bone powder (BP) substances with and without mesenchymal stem cells (MSCs) and evaluate the synergistic effect of topically applied recombinant human bone morphogenic protein-2 (BMP2) on MSC-loaded BP using fibrin glue in a mastoid obliteration model. MATERIALS AND METHODS To determine the expression of osteocyte-specific genes, total RNA was isolated from three MSC groups: Untreated MSCs, MSCs cultured with BP, and MSCs cultured with BP and BMP2. Real-time polymerase chain reaction was carried out with specific primers of osteogenesis-related genes runt-related transcription factor 2, osteocalcin, osteoprotegerin, osterix, alkaline phosphatase, transforming growth factor beta, and type I collagen. Live/dead staining was also performed. To observe the adhesion of MSCs to the BP, MSCs were treated with BP for 2 days and the surface was observed by scanning electron microscopy (SEM). Under general anesthesia, mastoid obliteration was performed in rats using three groups: treated with BP alone, BP/MSCs, and BP/MSC/BMP2. Before decapitation at 8 weeks post operation, in vivo micro computed tomography (micro CT) was performed. The bullae were dissected, fixed, and decalcified. followed by dehydration, paraffin embedding, and staining by hematoxylin and eosin and Masson's trichrome. RESULTS SEM showed the MSCs to be well-attached to the superficial area of the BP. The expression of osteocyte-specific genes was the highest in the MSCs cultured with BP and BMP2, followed by cultured with BP only, and untreated MSCs. The BP/MSC/BMP2 group showed the highest radiodensity of bullae in microCT analysis. The microCT findings revealed that the BP/MSC/BMP2 group showed the most enhanced osteogenesis of the scaffold compared to the other two groups. No significant difference was found in osteoconductive osteogenesis between the control and BP/MSC groups. However, the BP/MSC/BMP2 group showed significantly enhanced osteoconductive osteogenesis and osteoinductive change of the BP as shown by hematoxylin and eosin staining. Histomorphometry of osteogenesis revealed that the difference between the BP/MSC/BMP2 group and the other two groups was statistically significant. CONCLUSION A small amount of BMP2 is necessary during MSC loading to enhance the osteogenesis of BP and avoid complications associated with high doses of BMP2. These results may be applicable to mastoid obliteration in clinical practice.
Collapse
Affiliation(s)
- Chul Ho Jang
- Department of Otolaryngology, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Gwang Won Cho
- Department of Biology, College of Natural Science, Chosun University, Gwangju, Republic of Korea.,Department of Life Science, BK-21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, Republic of Korea
| | - An-Ji Song
- Department of Biology, College of Natural Science, Chosun University, Gwangju, Republic of Korea.,Department of Life Science, BK-21-Plus Research Team for Bioactive Control Technology, Chosun University, Gwangju, Republic of Korea
| |
Collapse
|
28
|
Hashemzadeh MR, Taghavizadeh Yazdi ME, Amiri MS, Mousavi SH. Stem cell therapy in the heart: Biomaterials as a key route. Tissue Cell 2021; 71:101504. [PMID: 33607524 DOI: 10.1016/j.tice.2021.101504] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/29/2021] [Accepted: 01/31/2021] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases are one of the main concerns, nowadays causing a high rate of mortality in the world. The majority of conventional treatment protects the heart from failure progression. As a novel therapeutic way, Regenerative medicine in the heart includes cellular and noncellular approaches. Despite the irrefutable privileges of noncellular aspects such as administration of exosomes, utilizing of miRNAs, and growth factors, they cannot reverse necrotic or ischemic myocardium, hence recruiting of stem cells to help regenerative therapy in the heart seems indispensable. Stem cell lineages are varied and divided into two main groups namely pluripotent and adult stem cells. Not only has each of which own regenerative capacity, benefits, and drawbacks, but their turnover also close correlates with the target organ and/or tissue as well as the stage and level of failure. In addition to inefficient tissue integration due to the defects in delivering methods and poor retention of transplanted cells, the complexity of the heart and its movement also make more rigorous the repair process. Hence, utilizing biomaterials can make a key route to tackle such obstacles. In this review, we evaluate some natural products which can help stem cells in regenerative medicine of the cardiovascular system.
Collapse
Affiliation(s)
- Mohammad Reza Hashemzadeh
- Department of Stem Cells and Regenerative Medicine, Royesh Stem Cell Biotechnology Institute, Mashhad, Iran; Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | | | - Seyed Hadi Mousavi
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
29
|
Mansoorifar A, Gordon R, Bergan R, Bertassoni LE. Bone-on-a-chip: microfluidic technologies and microphysiologic models of bone tissue. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2006796. [PMID: 35422682 PMCID: PMC9007546 DOI: 10.1002/adfm.202006796] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Indexed: 05/07/2023]
Abstract
Bone is an active organ that continuously undergoes an orchestrated process of remodeling throughout life. Bone tissue is uniquely capable of adapting to loading, hormonal, and other changes happening in the body, as well as repairing bone that becomes damaged to maintain tissue integrity. On the other hand, diseases such as osteoporosis and metastatic cancers disrupt normal bone homeostasis leading to compromised function. Historically, our ability to investigate processes related to either physiologic or diseased bone tissue has been limited by traditional models that fail to emulate the complexity of native bone. Organ-on-a-chip models are based on technological advances in tissue engineering and microfluidics, enabling the reproduction of key features specific to tissue microenvironments within a microfabricated device. Compared to conventional in-vitro and in-vivo bone models, microfluidic models, and especially organs-on-a-chip platforms, provide more biomimetic tissue culture conditions, with increased predictive power for clinical assays. In this review, we will report microfluidic and organ-on-a-chip technologies designed for understanding the biology of bone as well as bone-related diseases and treatments. Finally, we discuss the limitations of the current models and point toward future directions for microfluidics and organ-on-a-chip technologies in bone research.
Collapse
Affiliation(s)
- Amin Mansoorifar
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
| | - Ryan Gordon
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Raymond Bergan
- Division of Hematology/Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Luiz E. Bertassoni
- Department of Restorative Dentistry, School of Dentistry, Oregon Health & Science University, Portland, OR, USA
- Center for Regenerative Medicine, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Portland, OR, USA
| |
Collapse
|
30
|
Wang K, Albert K, Mosser G, Haye B, Percot A, Paris C, Peccate C, Trichet L, Coradin T. Self-assembly/condensation interplay in nano-to-microfibrillar silicified fibrin hydrogels. Int J Biol Macromol 2020; 164:1422-1431. [DOI: 10.1016/j.ijbiomac.2020.07.220] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/10/2020] [Accepted: 07/25/2020] [Indexed: 12/15/2022]
|
31
|
Coradin T, Wang K, Law T, Trichet L. Type I Collagen-Fibrin Mixed Hydrogels: Preparation, Properties and Biomedical Applications. Gels 2020; 6:E36. [PMID: 33092154 PMCID: PMC7709698 DOI: 10.3390/gels6040036] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/13/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022] Open
Abstract
Type I collagen and fibrin are two essential proteins in tissue regeneration and have been widely used for the design of biomaterials. While they both form hydrogels via fibrillogenesis, they have distinct biochemical features, structural properties and biological functions which make their combination of high interest. A number of protocols to obtain such mixed gels have been described in the literature that differ in the sequence of mixing/addition of the various reagents. Experimental and modelling studies have suggested that such co-gels consist of an interpenetrated structure where the two proteins networks have local interactions only. Evidences have been accumulated that immobilized cells respond not only to the overall structure of the co-gels but can also exhibit responses specific to each of the proteins. Among the many biomedical applications of such type I collagen-fibrin mixed gels, those requiring the co-culture of two cell types with distinct affinity for these proteins, such as vascularization of tissue engineering constructs, appear particularly promising.
Collapse
Affiliation(s)
- Thibaud Coradin
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 4 Place Jussieu, 75005 Paris, France; (K.W.); (T.L.); (L.T.)
| | | | | | | |
Collapse
|
32
|
Bédard P, Gauvin S, Ferland K, Caneparo C, Pellerin È, Chabaud S, Bolduc S. Innovative Human Three-Dimensional Tissue-Engineered Models as an Alternative to Animal Testing. Bioengineering (Basel) 2020; 7:E115. [PMID: 32957528 PMCID: PMC7552665 DOI: 10.3390/bioengineering7030115] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Animal testing has long been used in science to study complex biological phenomena that cannot be investigated using two-dimensional cell cultures in plastic dishes. With time, it appeared that more differences could exist between animal models and even more when translated to human patients. Innovative models became essential to develop more accurate knowledge. Tissue engineering provides some of those models, but it mostly relies on the use of prefabricated scaffolds on which cells are seeded. The self-assembly protocol has recently produced organ-specific human-derived three-dimensional models without the need for exogenous material. This strategy will help to achieve the 3R principles.
Collapse
Affiliation(s)
- Patrick Bédard
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Sara Gauvin
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Karel Ferland
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Christophe Caneparo
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Ève Pellerin
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Stéphane Chabaud
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Stéphane Bolduc
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
33
|
Yue H, Pathak JL, Zou R, Qin L, Liao T, Hu Y, Kuang W, Zhou L. Fabrication of chondrocytes/chondrocyte-microtissues laden fibrin gel auricular scaffold for microtia reconstruction. J Biomater Appl 2020; 35:838-848. [PMID: 32875937 DOI: 10.1177/0885328220954415] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fibrin gel-based scaffolds have promising potential for microtia reconstruction. Autologous chondrocytes and chondrocyte cell sheets are frequently used seed cell sources for cartilage tissue engineering. However, the aesthetic outcome of chondrocyte-based microtia reconstruction is still not satisfactory. In this study, we aimed to fabricate the chondrocytes/chondrocyte-microtissues laden fibrin gel auricular scaffold for microtia reconstruction. We designed a unique auricular mold that could fabricate a fibrin gel scaffold resembling human auricle anatomy. Primary chondrocytes were harvested from rabbit auricular cartilage, and chondrocyte cell sheets were developed. Chondrocyte-microtissues were prepared from the cell sheets. The mixture of chondrocytes/chondrocyte-microtissues was laden in fibrin gel during the auricular scaffold fabrication. The protrusions and recessed structure in the auricular scaffold surface were still clearly distinguishable. After a one-week in vitro culture, the 3 D structure and auricular anatomy of the scaffold were retained. And followed by eight-week subcutaneous implantation, cartilaginous tissue was regenerated in the artificial auricular structure as indicated by the results of H&E, Toluidine blue, Safranin O, and type II collagen (immunohistochemistry) staining. Protrusions and depressions of the auricular scaffold were slightly deformed, but the overall auricular anatomy was maintained after 8-week in vivo implantation. Extracellular matrix components content were similar in artificial auricular cartilage and rabbit native auricular cartilage. In conclusion, the mixture of chondrocytes/chondrocyte-microtissues laden fibrin gel auricular scaffold showed a promising potential for cartilaginous tissue regeneration, suggesting this as an effective approach for autologous chondrocyte-based microtia reconstruction.
Collapse
Affiliation(s)
- Haiqiong Yue
- Department of oral and Maxillofacial surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Janak L Pathak
- Department of oral and Maxillofacial surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Rui Zou
- Department of oral and Maxillofacial surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Lei Qin
- Department of oral and Maxillofacial surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Ting Liao
- Department of oral and Maxillofacial surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Yongxin Hu
- Department of oral and Maxillofacial surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Wei Kuang
- Department of oral and Maxillofacial surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China
| | - Libin Zhou
- Department of oral and Maxillofacial surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, China.,Department of Otolaryngology Head and Neck surgery, The 306th Hospital of PLA, Beijing, China *The first two authors contributed equally to this work
| |
Collapse
|
34
|
Panebianco C, DiStefano T, Mui B, Hom W, Iatridis J. Crosslinker concentration controls TGFβ-3 release and annulus fibrosus cell apoptosis in genipin-crosslinked fibrin hydrogels. Eur Cell Mater 2020; 39:211-226. [PMID: 32396210 PMCID: PMC7372750 DOI: 10.22203/ecm.v039a14] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Back pain is a leading cause of global disability associated with intervertebral disc (IVD) pathologies. Discectomy alleviates disabling pain caused by IVD herniation without repairing annulus fibrosus (AF) defects, which can cause accelerated degeneration and recurrent pain. Biological therapies show promise for IVD repair but developing high-modulus biomaterials capable of providing biomechanical stabilisation and delivering biologics remains an unmet challenge. The present study identified critical factors and developed an optimal formulation to enhance the delivery of AF cells and transforming growth beta-3 (TGFβ-3) in genipin-crosslinked fibrin (FibGen) hydrogels. Part 1 showed that AF cells encapsulated in TGFβ-3-supplemented high-modulus FibGen synthesised little extracellular matrix (ECM) but could release TGFβ-3 at physiologically relevant levels. Part 2 showed that AF cells underwent apoptosis when encapsulated in FibGen, even after reducing fibrin concentration from 70 to 5 mg/mL. Mechanistic experiments, modifying genipin concentration and integrin binding site presence demonstrated that genipin crosslinking caused AF cell apoptosis by inhibiting cell-biomaterial binding. Adding integrin binding sites with fibronectin partially rescued apoptosis, indicating genipin also caused acute cytotoxicity. Part 3 showed that FibGen formulations with 1 mg/mL genipin had enhanced ECM synthesis when supplemented with fibronectin and TGFβ-3. In conclusion, FibGen could be used for delivering biologically active compounds and AF cells, provided that formulations supplied additional sites for cell-biomaterial binding and genipin concentrations were low. Results also highlighted a need for developing strategies that protect cells against acute crosslinker cytotoxicity to overcome challenges of engineering high-modulus cell carriers for musculoskeletal tissues that experience high mechanical demands.
Collapse
Affiliation(s)
| | | | | | | | - J.C. Iatridis
- Address for correspondence: James C. Iatridis, PhD, 1468 Madison Avenue-Annenberg Building, floor 20, Room A20-086, New York, 10029 NY, USA., Telephone number: +1 2122411517
| |
Collapse
|
35
|
Wang S, Li Y, Li S, Yang J, Tang R, Li X, Li L, Fei J. Platelet-rich plasma loaded with antibiotics as an affiliated treatment for infected bone defect by combining wound healing property and antibacterial activity. Platelets 2020; 32:479-491. [PMID: 32396493 DOI: 10.1080/09537104.2020.1759792] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
To be faced with an infected bone defect and the need to accelerate bone union while controlling infection is a welcome challenge for orthopedists. Platelet-rich plasma (PRP) has been applied in tissue defects given their composition of growth factors however the weak antibacterial effects have limited the use of PRP in the clinical setting. Therefore, the aim of this study was to explore the feasibility of using PRP in a local antibiotic delivery system (PADS) with the characteristics of promoting wound healing of bone infection. PADS was prepared with the addition of antibiotics or no antibiotics as control after PRP was prepared by a two-step centrifugation procedure. Antibacterial tests showed zones of inhibition produced by antibiotics were not significantly different with antibiotics combined with PRP. HPLC analysis demonstrated that about 60% of the total vancomycin (VAN) and ceftazidime (CAZ) dose were released within 10 min, then the release rate gradually decreased. However, 90% clindamycin was released within 10 min. Interestingly, above 10 times the minimum inhibitory concentration was presented after 72 h. Additionally, ELISA and morphology studies of PADS indicated that loaded antibiotics could reduce the PRP-released growth factor concentration and disturb the structure of platelet-fibrin beams and fibrin network in a dose-dependent manner. Fortunately, the lower dose of antibiotics maintained their anti-microbial effect, meanwhile growth factors released from PADS, the structure of platelet-fibrin beams, fibrin network remained unaffected. In addition, a patient experiencing infected bone defect receiving this PADS treatment achieved union within the 15-month follow-up. Therefore, this novel PADS approach might represent a potential therapy for patients who have sustained infected bone defects.
Collapse
Affiliation(s)
- Shaochuan Wang
- Department of Emergency Medicine of Daping Hospital, Army Medical University, Chongqing, China
| | - Youbin Li
- Department of Emergency Medicine of Daping Hospital, Army Medical University, Chongqing, China
| | - Shidan Li
- Department of Emergency Medicine of Daping Hospital, Army Medical University, Chongqing, China
| | - Jing Yang
- Department of Emergency Medicine of Daping Hospital, Army Medical University, Chongqing, China
| | - Ruohui Tang
- Department of Emergency Medicine of Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaoming Li
- Department 4, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Lei Li
- Department of Immunity and Infection, Research Institute of Surgery, Army Medical University, Chongqing, China
| | - Jun Fei
- Department of Emergency Medicine of Daping Hospital, Army Medical University, Chongqing, China.,State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, China
| |
Collapse
|
36
|
Madhusudanan P, Raju G, Shankarappa S. Hydrogel systems and their role in neural tissue engineering. J R Soc Interface 2020; 17:20190505. [PMID: 31910776 PMCID: PMC7014813 DOI: 10.1098/rsif.2019.0505] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 12/02/2019] [Indexed: 12/27/2022] Open
Abstract
Neural tissue engineering (NTE) is a rapidly progressing field that promises to address several serious neurological conditions that are currently difficult to treat. Selecting the right scaffolding material to promote neural and non-neural cell differentiation as well as axonal growth is essential for the overall design strategy for NTE. Among the varieties of scaffolds, hydrogels have proved to be excellent candidates for culturing and differentiating cells of neural origin. Considering the intrinsic resistance of the nervous system against regeneration, hydrogels have been abundantly used in applications that involve the release of neurotrophic factors, antagonists of neural growth inhibitors and other neural growth-promoting agents. Recent developments in the field include the utilization of encapsulating hydrogels in neural cell therapy for providing localized trophic support and shielding neural cells from immune activity. In this review, we categorize and discuss the various hydrogel-based strategies that have been examined for neural-specific applications and also highlight their strengths and weaknesses. We also discuss future prospects and challenges ahead for the utilization of hydrogels in NTE.
Collapse
Affiliation(s)
| | | | - Sahadev Shankarappa
- Centre for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi 682041, India
| |
Collapse
|
37
|
Wang K, Trichet L, Rieu C, Peccate C, Pembouong G, Bouteiller L, Coradin T. Interactions of Organosilanes with Fibrinogen and Their Influence on Muscle Cell Proliferation in 3D Fibrin Hydrogels. Biomacromolecules 2019; 20:3684-3695. [DOI: 10.1021/acs.biomac.9b00686] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Kun Wang
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 75005 Paris, France
| | - Léa Trichet
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 75005 Paris, France
| | - Clément Rieu
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 75005 Paris, France
| | - Cécile Peccate
- Sorbonne Université, Inserm UMRS974, Association Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Gaëlle Pembouong
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, 75005 Paris, France
| | - Laurent Bouteiller
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, 75005 Paris, France
| | - Thibaud Coradin
- Sorbonne Université, CNRS, Laboratoire de Chimie de la Matière Condensée de Paris, 75005 Paris, France
| |
Collapse
|
38
|
Wong C, Yoganarasimha S, Carrico C, Madurantakam P. Incorporation of Fibrin Matrix into Electrospun Membranes for Periodontal Wound Healing. Bioengineering (Basel) 2019; 6:bioengineering6030057. [PMID: 31262021 PMCID: PMC6784027 DOI: 10.3390/bioengineering6030057] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/22/2019] [Accepted: 06/25/2019] [Indexed: 11/23/2022] Open
Abstract
Guided tissue regeneration (GTR) aims to regenerate the lost attachment apparatus caused by periodontal disease through the use of a membrane. The goal of this study is to create and characterize a novel hybrid membrane that contains biologically active fibrin matrix within a synthetic polycaprolactone (PCL) electrospun membrane. Three-dimensional fibrin matrices and fibrin-incorporated electrospun membrane were created from fresh frozen plasma by centrifugation in glass vials under three different conditions: 400 g for 12 min, 1450 g for 15 min and 3000 g for 60 min. Half the membranes were crosslinked with 1% genipin. Degradation against trypsin indicated biologic stability while uniaxial tensile testing characterized mechanical properties. Continuous data was analyzed by ANOVA to detect differences between groups (p = 0.05). Fibrin-incorporated electrospun membranes showed statistically significant increase in mechanical properties (elastic modulus, strain at break and energy to break) compared to fibrin matrices. While crosslinking had marginal effects on mechanical properties, it did significantly increase biologic stability against trypsin (p < 0.0001). Lastly, membranes generated at 400 g and 1450 g were superior in mechanical properties and biologic stability compared to those generated at 3000 g. Fibrin-incorporated, crosslinked electrospun PCL membranes generated at lower centrifugation forces offers a novel strategy to generate a potentially superior membrane for GTR procedures.
Collapse
Affiliation(s)
- Choyi Wong
- Department of Periodontics, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Suyog Yoganarasimha
- Department of Biomedical Engineering, School of Engineering, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Caroline Carrico
- Department of Oral Health Promotion and Community Outreach, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Parthasarathy Madurantakam
- Department of General Practice, School of Dentistry, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
39
|
Hu P, Fang D, Shen L, Zhou H, Shao R, Chen M, Yao C, Shi Y, Chen Q. Fibrin matrix containing high-dose calcitriol promotes the apoptosis of gastric cancer cells by sustainably releasing calcitrol and D-dimer. J Biomater Appl 2019; 34:509-522. [PMID: 31195918 DOI: 10.1177/0885328219856248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Pingping Hu
- 1 Department of Pathology, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Affiliated to Jiangsu University, Zhenjiang, China
| | - Dong Fang
- 2 Department of Oncology, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Affiliated to Jiangsu University, Zhenjiang, China
| | - Liping Shen
- 2 Department of Oncology, Zhenjiang Hospital of Chinese Traditional and Western Medicine, Affiliated to Jiangsu University, Zhenjiang, China
| | - Huangao Zhou
- 3 Intensive Care Unit, Jiangyin People's Hospital, Jiangyin, China
| | - Rui Shao
- 4 Department of Pathology, Zhenjiang People's Hospital, Zhenjiang, China
| | - Miao Chen
- 4 Department of Pathology, Zhenjiang People's Hospital, Zhenjiang, China
| | - Chenghu Yao
- 4 Department of Pathology, Zhenjiang People's Hospital, Zhenjiang, China
| | - Yang Shi
- 4 Department of Pathology, Zhenjiang People's Hospital, Zhenjiang, China
| | - Qian Chen
- 5 School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
40
|
Fibrin glue as a local drug-delivery system for bacteriophage PA5. Sci Rep 2019; 9:2091. [PMID: 30765740 PMCID: PMC6376040 DOI: 10.1038/s41598-018-38318-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/06/2018] [Indexed: 01/25/2023] Open
Abstract
Fibrin glue has been used clinically for decades in a wide variety of surgical specialties and is now being investigated as a medium for local, prolonged drug delivery. Effective local delivery of antibacterial substances is important perioperatively in patients with implanted medical devices or postoperatively for deep wounds. However, prolonged local application of antibiotics is often not possible or simply inadequate. Biofilm formation and antibiotic resistance are also major obstacles to antibacterial therapy. In this paper we test the biocompatibility of bacteriophages incorporated within fibrin glue, track the release of bacteriophages from fibrin scaffolds, and measure the antibacterial activity of released bacteriophages. Fibrin glue polymerized in the presence of the PA5 bacteriophage released high titers of bacteriophages during 11 days of incubation in liquid medium. Released PA5 bacteriophages were effective in killing Pseudomonas aeruginosa PA01. Overall, our results show that fibrin glue can be used for sustained delivery of bacteriophages and this strategy holds promise for many antibacterial applications.
Collapse
|
41
|
Abelseth E, Abelseth L, De la Vega L, Beyer ST, Wadsworth SJ, Willerth SM. 3D Printing of Neural Tissues Derived from Human Induced Pluripotent Stem Cells Using a Fibrin-Based Bioink. ACS Biomater Sci Eng 2018; 5:234-243. [DOI: 10.1021/acsbiomaterials.8b01235] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | | | | | - Simon T. Beyer
- Aspect Biosystems, 1781 W 75th Avenue, Vancouver, British Columbia V6P 6P2, Canada
| | - Samuel J. Wadsworth
- Aspect Biosystems, 1781 W 75th Avenue, Vancouver, British Columbia V6P 6P2, Canada
| | | |
Collapse
|
42
|
Wang O, Ismail A, Fabian FM, Lin H, Li Q, Elowsky C, Carlson MA, Burgess W, Velander WH, Kidambi S, Lei Y. A totally recombinant fibrin matrix for mesenchymal stem cell culture and delivery. J Biomed Mater Res A 2018; 106:3135-3142. [PMID: 30152030 DOI: 10.1002/jbm.a.36508] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 06/15/2018] [Accepted: 07/12/2018] [Indexed: 01/14/2023]
Abstract
Mesenchymal stem cells (MSCs) have been widely studied for tissue engineering and treating diseases in laboratories, clinical trials, and clinics. Fibrin matrices are often used to culture MSCs or increase the retention of MSCs at the injection site. However, fibrins made with the human plasma derived fibrinogen have high cost and risk of human pathogen transmission. In this article, we studied if fibrin matrices made with recombinant human fibrinogen, recombinant human thrombin, and recombinant human factor XIII could be used to culture and deliver MSCs. We systematically investigated the relationships between the fibrin matrix formulation, its nanostructure, and the behaviors of the cells in the matrix including the cell morphology, viability, and growth. We found that the fibrinogen concentration significantly affected the matrix structure and cell behaviors. We then used an optimized fibrin matrix to deliver human MSCs into mice subcutaneously. We found that the matrix could significantly enhance the retention of MSCs at the injection site. To our best knowledge, this is the first study on using fibrin matrices made with entirely recombinant proteins for culturing and delivering MSCs. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 3135-3142, 2018.
Collapse
Affiliation(s)
- Ou Wang
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska.,Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska
| | - Ayman Ismail
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Frank Marco Fabian
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Haishuang Lin
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Qiang Li
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Christian Elowsky
- Department of Agronomy and Horticulture, University of Nebraska, Lincoln, Nebraska
| | - Mark A Carlson
- Department of Surgery, University of Nebraska Medical Center and the Omaha VA Medical Center, Omaha, Nebraska
| | - Wilson Burgess
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - William H Velander
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska
| | - Srivatsan Kidambi
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska.,Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska.,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Yuguo Lei
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Nebraska.,Biomedical Engineering Program, University of Nebraska, Lincoln, Nebraska.,Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, Nebraska.,Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
43
|
Gessmann J, Seybold D, Ayami F, Peter E, Baecker H, Schildhauer TA, Köller M. Peripheral Blood Plasma Clot as a Local Antimicrobial Drug Delivery Matrix. Tissue Eng Part A 2018; 24:809-818. [DOI: 10.1089/ten.tea.2017.0319] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jan Gessmann
- Department of Trauma Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
- Department of Surgical Research, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Dominik Seybold
- Department of Trauma Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
- Department of Surgical Research, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Fahim Ayami
- Department of Surgical Research, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Elvira Peter
- Department of Surgical Research, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Hinnerk Baecker
- Department of Trauma Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Thomas Armin Schildhauer
- Department of Trauma Surgery, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Manfred Köller
- Department of Surgical Research, BG University Hospital Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
44
|
Heher P, Mühleder S, Mittermayr R, Redl H, Slezak P. Fibrin-based delivery strategies for acute and chronic wound healing. Adv Drug Deliv Rev 2018; 129:134-147. [PMID: 29247766 DOI: 10.1016/j.addr.2017.12.007] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/24/2017] [Accepted: 12/09/2017] [Indexed: 12/17/2022]
Abstract
Fibrin, a natural hydrogel, is the end product of the physiological blood coagulation cascade and naturally involved in wound healing. Beyond its role in hemostasis, it acts as a local reservoir for growth factors and as a provisional matrix for invading cells that drive the regenerative process. Its unique intrinsic features do not only promote wound healing directly via modulation of cell behavior but it can also be fine-tuned to evolve into a delivery system for sustained release of therapeutic biomolecules, cells and gene vectors. To further augment tissue regeneration potential, current strategies exploit and modify the chemical and physical characteristics of fibrin to employ combined incorporation of several factors and their timed release. In this work we show advanced therapeutic approaches employing fibrin matrices in wound healing and cover the many possibilities fibrin offers to the field of regenerative medicine.
Collapse
|
45
|
Hoberman AR, Cirino C, McCarthy MB, Cote MP, Pauzenberger L, Beitzel K, Mazzocca AD, Dyrna F. Bone Marrow-Derived Mesenchymal Stromal Cells Enhanced by Platelet-Rich Plasma Maintain Adhesion to Scaffolds in Arthroscopic Simulation. Arthroscopy 2018; 34:872-881. [PMID: 29146168 DOI: 10.1016/j.arthro.2017.08.291] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 08/24/2017] [Accepted: 08/25/2017] [Indexed: 02/02/2023]
Abstract
PURPOSE To assess the response of bone marrow-derived mesenchymal stromal cells (bMSCs) enhanced by platelet-rich plasma (PRP) in the setting of a normal human tendon (NHT), a demineralized bone matrix (DBM), and a fibrin scaffold (FS) with simulated arthroscopic mechanical washout stress. METHODS Bone marrow was aspirated from the humeral head and concentrated. BMSCs were counted, plated, and grown to confluence. Cells were seeded onto 3 different scaffolds: (1) NHT, (2) DBM, and (3) FS. Each scaffold was treated with a combination of (+)/(-) PRP and (+)/(-) arthroscopic washout simulation. A period of 60 minutes was allotted before arthroscopic washout. Adhesion, proliferation, and differentiation assays were performed to assess cellular activity in each condition. RESULTS Significant differences were seen in mesenchymal stromal cell adhesion, proliferation, and differentiation among the scaffolds. DBM and FS showed superior results to NHT for cell adhesion, proliferation, and differentiation. PRP significantly enhanced cellular adhesion, proliferation, and differentiation. Arthroscopic simulation did not significantly decrease bMSC adhesion. CONCLUSIONS We found that the type of scaffold impacts bMSCs' behavior. Both scaffolds (DBM and FS) were superior to NHT. The use of an arthroscopic simulator did not significantly decrease the adhesion of bMSCs to the scaffolds nor did it decrease their biologic differentiation potential. In addition, PRP enhanced cellular adhesion, proliferation, and differentiation. CLINICAL RELEVANCE Improved healing after tendon repair can lead to better clinical outcomes. BMSCs are attractive for enhancing healing given their accessibility and regenerative potential. Application of bMSCs using scaffolds as cell carriers relies on arthroscopic feasibility.
Collapse
Affiliation(s)
- Alexander R Hoberman
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A..
| | - Carl Cirino
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Mary Beth McCarthy
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Mark P Cote
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Leo Pauzenberger
- Department of Orthopaedic Sports Medicine, Technical University, Munich, Germany
| | - Knut Beitzel
- Department of Orthopaedic Sports Medicine, Technical University, Munich, Germany
| | - Augustus D Mazzocca
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, Connecticut, U.S.A
| | - Felix Dyrna
- Department of Orthopaedic Sports Medicine, Technical University, Munich, Germany
| |
Collapse
|
46
|
Fibrin-Based Biomaterial Applications in Tissue Engineering and Regenerative Medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:253-261. [DOI: 10.1007/978-981-13-0445-3_16] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
47
|
Foidl BM, Ucar B, Schwarz A, Rebelo AL, Pandit A, Humpel C. Nerve growth factor released from collagen scaffolds protects axotomized cholinergic neurons of the basal nucleus of Meynert in organotypic brain slices. J Neurosci Methods 2017; 295:77-86. [PMID: 29221639 DOI: 10.1016/j.jneumeth.2017.12.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 12/04/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Alzheimeŕs disease is accompanied by cell death of cholinergic neurons, resulting in cognitive impairment and memory loss. Nerve growth factor (NGF) is the most potent protein to support survival of cholinergic neurons. NEW METHOD Organotypic brain slices of the basal nucleus of Meynert (nBM) are a valuable tool to study cell death of axotomized cholinergic neurons, as well as protective effects of NGF added into the medium. The aim of the present study is to use collagen scaffolds crosslinked with polyethyleneglycole and load with NGF to target delivery of NGF to organotypic nBM brain slices. RESULTS Collagen scaffolds (visualized by incorporating AlexaFluor 488 antibodies) slowly degraded when applied onto organotypic brain slices within 2 weeks in culture. GFAP reactive astrocytes and Iba1+ microglia became visible around the collagen scaffolds 7days after incubation, showing reactive gliosis. Cholinergic neurons of the nBM survived (201±21, n=8) when incubated with 100ng/ml NGF in the medium compared to NGF-free medium (69±12, n=7). Collagen scaffolds loaded with NGF (1ng/2μl scaffold) significantly rescued cholinergic cell death in the nBM brain slices (175±12, n=10), which was counteracted by an anti-NGF antibody (77±5, n=5). COMPARISON WITH EXISTING METHODS The combination of coronal brain slices with biomaterial is a novel and potent tool to selectively study neuroprotective effects. CONCLUSIONS Collagen scaffolds loaded with low amounts of a protein/drug of interest can be easily applied directly onto organotypic brain slices, allowing slow targeted release of a protective molecule. Such an approach is highly useful to optimize CollScaff for further in vivo applications.
Collapse
Affiliation(s)
- Bettina M Foidl
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Austria
| | - Buket Ucar
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Austria
| | - Alina Schwarz
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Austria
| | - Ana L Rebelo
- Centre for Research in Medical Devices, Biomedical Sciences National University of Ireland Galway, Galway, Ireland
| | - Abhay Pandit
- Centre for Research in Medical Devices, Biomedical Sciences National University of Ireland Galway, Galway, Ireland
| | - Christian Humpel
- Laboratory of Psychiatry and Experimental Alzheimer's Research, Medical University of Innsbruck, Austria.
| |
Collapse
|
48
|
Hasan ML, Taz M, Lee BT. Effects of platelet-rich plasma on biological activity and bone regeneration of brushite-based calcium phosphate cement. J Biomed Mater Res B Appl Biomater 2017; 106:2316-2326. [DOI: 10.1002/jbm.b.34036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 09/12/2017] [Accepted: 10/13/2017] [Indexed: 11/05/2022]
Affiliation(s)
- Md Lemon Hasan
- Department of Regenerative Medicine; College of Medicine, Soonchunhyang University, Cheonan-City; ChungCheongNam-Do South Korea
| | - Mirana Taz
- Department of Regenerative Medicine; College of Medicine, Soonchunhyang University, Cheonan-City; ChungCheongNam-Do South Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine; College of Medicine, Soonchunhyang University, Cheonan-City; ChungCheongNam-Do South Korea
- Institute of Tissue Regeneration, College of Medicine, Soonchunhyang University, Cheonan-City; ChungCheongNam-Do South Korea
| |
Collapse
|
49
|
Huang S, Wang C, Xu J, Ma L, Gao C. In situ assembly of fibrinogen/hyaluronic acid hydrogel via knob-hole interaction for 3D cellular engineering. Bioact Mater 2017; 2:253-259. [PMID: 29744434 PMCID: PMC5935512 DOI: 10.1016/j.bioactmat.2017.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 09/14/2017] [Accepted: 09/15/2017] [Indexed: 12/26/2022] Open
Abstract
Hyaluronic acid (HA)-based hydrogels have applied widely for biomedical applications due to its biocompatibility and biodegradability. However, the use of initiators or crosslinkers during the hydrogel formation may cause cytotoxicity and thereby impair the biocompatibility. Inspired by the crosslinking mechanism of fibrin gel, a novel HA-based hydrogel was developed via the in situ supramolecular assembly based on knob-hole interactions between fibrinogen and knob-grafted HA (knob-g-HA) in this study. The knob-grafted HA was synthesized by coupling knob peptides (GPRPAAC, a mimic peptide of fibrin knob A) to HA via Michael addition. Then the translucent fibrinogen/knob-g-HA hydrogels were prepared by simply mixing the solutions of knob-g-HA and fibrinogen at the knob/hole ratio of 1.2. The rheological behaviors of the fibrinogen/knob-g-HA hydrogels with the fibrinogen concentrations of 50, 100 and 200 mg/mL were evaluated, and it was found that the dynamic storage moduli (G') were higher than the loss moduli (G″) over the whole frequency range for all the groups. The SEM results showed that fibrinogen/knob-g-HA hydrogels presented the heterogeneous mesh-like structures which were different from the honeycomb-like structures of fibrinogen/MA-HA hydrogels. Correspondingly, a higher swelling ratio was obtained in the groups of fibrinogen/knob-g-HA hydrogel. Finally, the cytocompatibility of fibrinogen/knob-g-HA hydrogels was proved by live/dead stainings and MTT assays in the 293T cells encapsulation test. All these results highlight the biological potential of the fibrinogen/knob-g-HA hydrogels for 3D cellular engineering.
Collapse
Affiliation(s)
- Shengjie Huang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Chunfen Wang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jingwei Xu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Lie Ma
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| |
Collapse
|
50
|
Robinson M, Douglas S, Michelle Willerth S. Mechanically stable fibrin scaffolds promote viability and induce neurite outgrowth in neural aggregates derived from human induced pluripotent stem cells. Sci Rep 2017; 7:6250. [PMID: 28740258 PMCID: PMC5524903 DOI: 10.1038/s41598-017-06570-9] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 06/14/2017] [Indexed: 01/08/2023] Open
Abstract
Recent work demonstrated that 3D fibrin scaffolds function as an effective substrate for engineering tissues from pluripotent stem cells. However, the rapid degradation rate of fibrin remains a major limitation when differentiating human pluripotent stem cells for tissue engineering applications. The addition of crosslinking agents, such as genipin, during the polymerization process increases scaffold stability while decreasing the degradation rate of fibrin. Genipin crosslinking alters the physical characteristics of the fibrin scaffolds, which influences the behaviour of the differentiating cells seeded inside. It also possesses neuritogenic and neuroprotective properties, making it particularly attractive for engineering neural tissue from pluripotent stem cells. Here we show that genipin enhances neuronal differentiation of neural progenitors derived from human induced pluripotent stem cells (hiPSCs) in 2D culture and genipin concentration influences the morphological and mechanical properties of 3D fibrin scaffolds. These mechanically stable genipin-crosslinked fibrin scaffolds support hiPSC-derived neural aggregates and induce neurite outgrowth while remaining intact for 2 weeks as opposed to 5 days for unmodified fibrin scaffolds.
Collapse
Affiliation(s)
- Meghan Robinson
- Biomedical Engineering Program, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8W 2Y2, Canada
| | - Sarah Douglas
- Biomedical Engineering Program, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8W 2Y2, Canada
| | - Stephanie Michelle Willerth
- Department of Mechanical Engineering, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8W 2Y2, Canada.
- Division of Medical Sciences, University of Victoria, 3800 Finnerty Road, Victoria, BC, V8W 2Y2, Canada.
- International Collaboration on Repair Discoveries, University of British Columbia, 818 West 10th Avenue, Vancouver, BC, V5Z 1M9, Canada.
| |
Collapse
|