1
|
Zheng Y, Ke Z, Hu G, Tong S. Hydrogel promotes bone regeneration through various mechanisms: a review. BIOMED ENG-BIOMED TE 2025; 70:103-114. [PMID: 39571066 DOI: 10.1515/bmt-2024-0391] [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: 08/16/2024] [Accepted: 11/05/2024] [Indexed: 04/05/2025]
Abstract
Large defects in bone tissue due to trauma, tumors, or developmental abnormalities usually require surgical treatment for repair. Numerous studies have shown that current bone repair and regeneration treatments have certain complications and limitations. With the in-depth understanding of bone regeneration mechanisms and biological tissue materials, a variety of materials with desirable physicochemical properties and biological functions have emerged in the field of bone regeneration in recent years. Among them, hydrogels have been widely used in bone regeneration research due to their biocompatibility, unique swelling properties, and ease of fabrication. In this paper, the development and classification of hydrogels were introduced, and the mechanism of hydrogels in promoting bone regeneration was described in detail, including the promotion of bone marrow mesenchymal stem cell differentiation, the promotion of angiogenesis, the enhancement of the activity of bone morphogenetic proteins, and the regulation of the microenvironment of bone regeneration tissues. In addition, the future research direction of hydrogel in bone tissue engineering was discussed.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, Ningbo, China
| | - Zengguang Ke
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, Ningbo, China
| | - Guofeng Hu
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, Ningbo, China
| | - Songlin Tong
- Department of Orthopaedic Surgery, Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, Ningbo, China
| |
Collapse
|
2
|
Abbas SEM, Maged G, Wang H, Lotfy A. Mesenchymal Stem/Stromal Cells Microencapsulation for Cell Therapy. Cells 2025; 14:149. [PMID: 39936941 PMCID: PMC11817150 DOI: 10.3390/cells14030149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 02/13/2025] Open
Abstract
Cell microencapsulation is one of the most studied strategies to overcome the challenges associated with the implementation of mesenchymal stem/stromal cells (MSCs) in vivo. This approach isolates/shields donor MSCs from the host immune system using a semipermeable membrane that allows for the diffusion of gases, nutrients, and therapeutics, but not host immune cells. As a result, microencapsulated MSCs survive and engraft better after infusion, and they can be delivered specifically to the targeted site. Additionally, microencapsulation enables the co-culture of MSCs with different types of cells in a three-dimensional (3D) environment, allowing for better cellular interaction. Alginate, collagen, and cellulose are the most popular materials, and air jet extrusion, microfluidics, and emulsion are the most used techniques for MSC cell encapsulation in the literature. These materials and techniques differ in the size range of the resultant microcapsules and their compatibility with the applied materials. This review discusses various materials and techniques used for the microencapsulation of MSCs. We also shed light on the recent findings in this field, the advantages and drawbacks of using encapsulated MSCs, and the in vivo translation of the microencapsulated MSCs in cell therapy.
Collapse
Affiliation(s)
| | - Ghada Maged
- Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria 21526, Egypt
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401, USA
| | - Ahmed Lotfy
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
3
|
Devernois E, Coradin T. Synthesis, Characterization and Biological Properties of Type I Collagen-Chitosan Mixed Hydrogels: A Review. Gels 2023; 9:518. [PMID: 37504397 PMCID: PMC10379456 DOI: 10.3390/gels9070518] [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: 05/31/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Type I collagen and chitosan are two of the main biological macromolecules used to design scaffolds for tissue engineering. The former has the benefits of being biocompatible and provides biochemical cues for cell adhesion, proliferation and differentiation. However, collagen hydrogels usually exhibit poor mechanical properties and are difficult to functionalize. Chitosan is also often biocompatible, but is much more versatile in terms of structure and chemistry. Although it does have important biological properties, it is not a good substrate for mammalian cells. Combining of these two biomacromolecules is therefore a strategy of choice for the preparation of interesting biomaterials. The aim of this review is to describe the different protocols available to prepare Type I collagen-chitosan hydrogels for the purpose of presenting their physical and chemical properties and highlighting the benefits of mixed hydrogels over single-macromolecule ones. A critical discussion of the literature is provided to point out the poor understanding of chitosan-type I collagen interactions, in particular due to the lack of systematic studies addressing the effect of chitosan characteristics.
Collapse
Affiliation(s)
- Enguerran Devernois
- Laboratoire de Chimie de la Matière Condensée de Paris, CNRS, Sorbonne Université, 4 Place Jussieu, 75005 Paris, France
| | - Thibaud Coradin
- Laboratoire de Chimie de la Matière Condensée de Paris, CNRS, Sorbonne Université, 4 Place Jussieu, 75005 Paris, France
| |
Collapse
|
4
|
Nguyen-Thanh T, Nguyen-Tran BS, Cruciani S, Nguyen-Thi TD, Dang-Cong T, Maioli M. Osteochondral Regeneration Ability of Uncultured Bone Marrow Mononuclear Cells and Platelet-Rich Fibrin Scaffold. Bioengineering (Basel) 2023; 10:661. [PMID: 37370592 DOI: 10.3390/bioengineering10060661] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/25/2023] [Accepted: 05/26/2023] [Indexed: 06/29/2023] Open
Abstract
OBJECTIVES Platelet-rich fibrin (PRF) and bone marrow mononuclear cells are potential scaffolds and cell sources for osteochondral regeneration. The main aim of this paper is to examine the effects of PRF scaffolds and autologous uncultured bone marrow mononuclear cells on osteochondral regeneration in rabbit knees. MATERIALS AND METHODS Three different types of PRF scaffolds were generated from peripheral blood (Ch-PRF and L-PRF) and bone marrow combined with uncultured bone marrow mononuclear cells (BMM-PRF). The histological characteristics of these scaffolds were assessed via hematoxylin-eosin staining, PicroSirius red staining, and immunohistochemical staining. Osteochondral defects with a diameter of 3 mm and depth of 3 mm were created on the trochlear groove of the rabbit's femur. Different PRF scaffolds were then applied to treat the defects. A group of rabbits with induced osteochondral defects that were not treated with any scaffold was used as a control. Osteochondral tissue regeneration was assessed after 2, 4, and 6 weeks by macroscopy (using the Internal Cartilage Repair Society score, X-ray) and microscopy (hematoxylin-eosin stain, safranin O stain, toluidine stain, and Wakitani histological scale, immunohistochemistry), in addition to gene expression analysis of osteochondral markers. RESULTS Ch-PRF had a heterogeneous fibrin network structure and cellular population; L-PRF and BMM-PRF had a homogeneous structure with a uniform distribution of the fibrin network. Ch-PRF and L-PRF contained a population of CD45-positive leukocytes embedded in the fibrin network, while mononuclear cells in the BMM-PRF scaffold were positive for the pluripotent stem cell-specific antibody Oct-4. In comparison to the untreated group, the rabbits that were given the autologous graft displayed significantly improved healing of the articular cartilage tissue and of the subchondral bone. Regeneration was gradually observed after 2, 4, and 6 weeks of PRF scaffold treatment, which was particularly evident in the BMM-PRF group. CONCLUSIONS The combination of biomaterials with autologous platelet-rich fibrin and uncultured bone marrow mononuclear cells promoted osteochondral regeneration in a rabbit model more than platelet-rich fibrin material alone. Our results indicate that autologous platelet-rich fibrin scaffolds combined with uncultured bone marrow mononuclear cells applied in healing osteochondral lesions may represent a suitable treatment in addition to stem cell and biomaterial therapy.
Collapse
Affiliation(s)
- Tung Nguyen-Thanh
- Faculty of Basic Science, Hue University of Medicine and Pharmacy, 6 Ngo Quyen Street, Hue 49000, Vietnam
- Institute of Biomedicine, Hue University of Medicine and Pharmacy, 6 Ngo Quyen Street, Hue 49000, Vietnam
| | - Bao-Song Nguyen-Tran
- Department of Histology, Embryology, Pathology and Forensic, Hue University of Medicine and Pharmacy, 6 Ngo Quyen Street, Hue 49000, Vietnam
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| | - Thuy-Duong Nguyen-Thi
- Faculty of Odonto-Stomatology, Hue University of Medicine and Pharmacy, 6 Ngo Quyen Street, Hue 49000, Vietnam
| | - Thuan Dang-Cong
- Department of Histology, Embryology, Pathology and Forensic, Hue University of Medicine and Pharmacy, 6 Ngo Quyen Street, Hue 49000, Vietnam
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| |
Collapse
|
5
|
Silva CM, Ornellas DS, Ornellas FM, Santos RS, Martini SV, Ferreira D, Muiler C, Cruz FF, Takiya CM, Rocco PRM, Morales MM, Silva PL. Early effects of bone marrow-derived mononuclear cells on lung and kidney in experimental sepsis. Respir Physiol Neurobiol 2023; 309:103999. [PMID: 36460253 DOI: 10.1016/j.resp.2022.103999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/11/2022] [Accepted: 11/20/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND In experimental sepsis, functional and morphological effects of bone marrow-derived mononuclear cell (BMDMC) administration in lung tissue have been evaluated 1 and 7 days after therapy. However, to date no study has evaluated the early effects of BMDMCs in both lung and kidney in experimental polymicrobial sepsis. MATERIAL AND METHODS Twenty-five female C57BL/6 mice were randomly divided into the following groups: 1) cecal ligation and puncture (CLP)-induced sepsis; and 2) Sham (surgical procedure without CLP). After 1 h, CLP animals received saline (NaCl 0.9%) (CLP-Saline) or 106 BMDMCs (CLP-Cell) via the jugular vein. At 6, 12, and 24 h after saline or BMDMC administration, lungs and kidneys were removed for histology and molecular biology analysis. RESULTS In lungs, CLP-Saline, compared to Sham, was associated with increased lung injury score (LIS) and keratinocyte chemoattractant (KC) mRNA expression at 6, 12, and 24 h. BMDMCs were associated with reduced LIS and KC mRNA expression regardless of the time point of analysis. Interleukin (IL)- 10 mRNA content was higher in CLP-Cell than CLP-Saline at 6 and 24 h. In kidney tissue, CLP-Saline, compared to Sham, was associated with tubular cell injury and increased neutrophil gelatinase-associated lipocalin (NGAL) levels, which were reduced after BMDMC therapy at all time points. Surface high-mobility-group-box (HMGB)- 1 levels were higher in CLP-Saline than Sham at 6, 12, and 24 h, whereas nuclear HMGB-1 levels were increased only at 24 h. BMDMCs were associated with decreased surface HMGB-1 and increased nuclear HMGB-1 levels. Kidney injury molecule (KIM)- 1 and IL-18 gene expressions were reduced in CLP-Cell compared to CLP-Saline at 12 and 24 h. CONCLUSION In the present experimental polymicrobial sepsis, early intravenous therapy with BMDMCs was able to reduce lung and kidney damage in a time-dependent manner. BMDMCs thus represent a potential therapy in well-known scenarios of sepsis induction. PURPOSE To evaluate early bone marrow-derived mononuclear cell (BMDMC) therapy on lung and kidney in experimental polymicrobial sepsis. METHODS Twenty-five female C57BL/6 mice were randomly divided into the following groups: cecal ligation and puncture (CLP)-induced sepsis; and sham (surgical procedure without CLP). After 1 h, CLP animals received saline (CLP-saline) or 106 BMDMCs (CLP-cell) via the jugular vein. Lungs and kidneys were evaluated for histology and molecular biology after 6, 12, and 24 h. RESULTS In lungs, BMDMCs reduced the lung injury score and keratinocyte chemoattractant mRNA expression regardless of the time point of analysis; interleukin-10 mRNA content was higher in CLP-cell than CLP-saline at 6 and 24 h. In kidneys, BMDMCs reduced neutrophil gelatinase-associated lipocalin levels at all time points. BMDMCs decreased surface high mobility group box (HMGB)- 1 but increased nuclear HMGB-1 levels. CONCLUSION Early BMDMC therapy reduced lung and kidney damage in a time-dependent manner.
Collapse
Affiliation(s)
- Carla M Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Debora S Ornellas
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Felipe M Ornellas
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Laboratory of Cellular, Genetic and Molecular Nephrology, Renal Division, Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Raquel S Santos
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Sabrina V Martini
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Debora Ferreira
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Caroline Muiler
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fernanda F Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Christina M Takiya
- Immunopathology Laboratory, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Patricia R M Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Marcelo M Morales
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
| | - Pedro L Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Boffa A, Perucca Orfei C, Sourugeon Y, Laver L, Magalon J, Sánchez M, Tischer T, de Girolamo L, Filardo G. Cell-based therapies have disease-modifying effects on osteoarthritis in animal models. A systematic review by the ESSKA Orthobiologic Initiative. Part 2: bone marrow-derived cell-based injectable therapies. Knee Surg Sports Traumatol Arthrosc 2023:10.1007/s00167-023-07320-3. [PMID: 36823238 DOI: 10.1007/s00167-023-07320-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/10/2023] [Indexed: 02/25/2023]
Abstract
PURPOSE Aim of this systematic review was to determine if bone marrow-derived cell-based injectable therapies induce disease-modifying effects in joints affected by osteoarthritis (OA) in animal models. METHODS A systematic review was performed on three electronic databases (PubMed, Web of Science, Embase) according to PRISMA guidelines. A synthesis of the results was performed investigating disease-modifying effects in preclinical animal studies comparing injectable bone marrow-derived products with OA controls or other products, different formulations or injection intervals, and the combination with other products. The risk of bias was assessed according to the SYRCLE's tool. RESULTS Fifty-three studies were included (1819 animals) with an increasing publication trend over time. Expanded cells were used in 48 studies, point-of-care products in 3 studies, and both approaches were investigated in 2 studies. Among the 47 studies presenting results on the disease-modifying effects, 40 studies (85%) reported better results with bone marrow-derived products compared to OA controls, with positive findings evident in 14 out of 20 studies (70%) in macroscopic assessment, in 30 out of 41 studies (73%) in histological assessment, and in 10 out of 13 studies (77%) in immunohistochemical evaluations. Clinical evaluations showed positive results in 7 studies out of 9 (78%), positive imaging results in 11 studies out of 17 (65%), and positive biomarker results in 5 studies out of 10 (50%). While 36 out of 46 studies (78%) reported positive results at the cartilage level, only 3 out of 10 studies (30%) could detect positive changes at the synovial level. The risk of bias was low in 42% of items, unclear in 50%, and high in 8%. CONCLUSION This systematic review of preclinical studies demonstrated that intra-articular injections of bone marrow-derived products can induce disease-modifying effects in the treatment of OA, slowing down the progression of cartilage damage with benefits at macroscopic, histological, and immunohistochemical levels. Positive results have been also observed in terms of clinical and imaging findings, as well as in the modulation of inflammatory and cartilage biomarkers, while poor effects have been described on the synovial membrane. These findings are important to understand the potential of bone marrow-derived products and to guide further research to optimise their use in the clinical practice. LEVEL OF EVIDENCE II.
Collapse
Affiliation(s)
- Angelo Boffa
- Applied and Translational Research Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Clinica Ortopedica e Traumatologica 2, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Carlotta Perucca Orfei
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Ospedale Galeazzi Sant'Ambrogio, Via Cristina Belgioioso 173, 20157, Milan, Italy.
| | | | - Lior Laver
- Department of Orthopaedics, Hillel Yaffe Medical Center (HYMC), Hadera, Israel
- Arthrosport Clinic, Tel‑Aviv, Israel
- Rappaport Faculty of Medicine, Technion University Hospital (Israel Institute of Technology), Haifa, Israel
| | - Jérémy Magalon
- Cell Therapy Laboratory, Hôpital De La Conception, AP-HM, Marseille, France
- INSERM, NRA, C2VN, Aix Marseille Univ, Marseille, France
- SAS Remedex, Marseille, France
| | - Mikel Sánchez
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, Vitoria‑Gasteiz, Spain
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, Vitoria‑Gasteiz, Spain
| | - Thomas Tischer
- Department of Orthopaedic Surgery, University of Rostock, Rostock, Germany
- Department of Orthopaedic and Trauma Surgery, Malteser Waldkrankenhaus St. Marien, Erlangen, Germany
| | - Laura de Girolamo
- Laboratorio di Biotecnologie Applicate all'Ortopedia, IRCCS Ospedale Galeazzi Sant'Ambrogio, Via Cristina Belgioioso 173, 20157, Milan, Italy
| | - Giuseppe Filardo
- Applied and Translational Research Center, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
7
|
Tavelli L, Barootchi S, Rasperini G, Giannobile WV. Clinical and patient-reported outcomes of tissue engineering strategies for periodontal and peri-implant reconstruction. Periodontol 2000 2023; 91:217-269. [PMID: 36166659 PMCID: PMC10040478 DOI: 10.1111/prd.12446] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/25/2022] [Accepted: 06/05/2022] [Indexed: 11/28/2022]
Abstract
Scientific advancements in biomaterials, cellular therapies, and growth factors have brought new therapeutic options for periodontal and peri-implant reconstructive procedures. These tissue engineering strategies involve the enrichment of scaffolds with living cells or signaling molecules and aim at mimicking the cascades of wound healing events and the clinical outcomes of conventional autogenous grafts, without the need for donor tissue. Several tissue engineering strategies have been explored over the years for a variety of clinical scenarios, including periodontal regeneration, treatment of gingival recessions/mucogingival conditions, alveolar ridge preservation, bone augmentation procedures, sinus floor elevation, and peri-implant bone regeneration therapies. The goal of this article was to review the tissue engineering strategies that have been performed for periodontal and peri-implant reconstruction and implant site development, and to evaluate their safety, invasiveness, efficacy, and patient-reported outcomes. A detailed systematic search was conducted to identify eligible randomized controlled trials reporting the outcomes of tissue engineering strategies utilized for the aforementioned indications. A total of 128 trials were ultimately included in this review for a detailed qualitative analysis. Commonly performed tissue engineering strategies involved scaffolds enriched with mesenchymal or somatic cells (cell-based tissue engineering strategies), or more often scaffolds loaded with signaling molecules/growth factors (signaling molecule-based tissue engineering strategies). These approaches were found to be safe when utilized for periodontal and peri-implant reconstruction therapies and implant site development. Tissue engineering strategies demonstrated either similar or superior clinical outcomes than conventional approaches for the treatment of infrabony and furcation defects, alveolar ridge preservation, and sinus floor augmentation. Tissue engineering strategies can promote higher root coverage, keratinized tissue width, and gingival thickness gain than scaffolds alone can, and they can often obtain similar mean root coverage compared with autogenous grafts. There is some evidence suggesting that tissue engineering strategies can have a positive effect on patient morbidity, their preference, esthetics, and quality of life when utilized for the treatment of mucogingival deformities. Similarly, tissue engineering strategies can reduce the invasiveness and complications of autogenous graft-based staged bone augmentation. More studies incorporating patient-reported outcomes are needed to understand the cost-benefits of tissue engineering strategies compared with traditional treatments.
Collapse
Affiliation(s)
- Lorenzo Tavelli
- Division of Periodontology, Department of Oral Medicine, Infection, and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts, USA
- Center for Clinical Research and Evidence Synthesis in Oral Tissue Regeneration (CRITERION), Boston, Massachusetts, USA
| | - Shayan Barootchi
- Center for Clinical Research and Evidence Synthesis in Oral Tissue Regeneration (CRITERION), Boston, Massachusetts, USA
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Giulio Rasperini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Milan, Italy
- IRCCS Foundation Polyclinic Ca’ Granda, University of Milan, Milan, Italy
| | | |
Collapse
|
8
|
Qi P, Ning Z, Zhang X. Synergistic effects of 3D chitosan-based hybrid scaffolds and mesenchymal stem cells in orthopaedic tissue engineering. IET Nanobiotechnol 2023; 17:41-48. [PMID: 36708277 PMCID: PMC10116017 DOI: 10.1049/nbt2.12103] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/06/2022] [Accepted: 10/20/2022] [Indexed: 01/29/2023] Open
Abstract
Restoration of damaged bone and cartilage tissue with biomaterial scaffolds is an area of interest in orthopaedics. Chitosan is among the low-cost biomaterials used as scaffolds with considerable biocompability to almost every human tissue. Considerable osteoconductivity, porosity, and appropriate pore size distribution have made chitosan an appropriate scaffold for loading of stem cells and a good homing place for differentiation of stem cells to bone tissue. Moreover, the similarity of chitosan to glycosaminoglycans and its potential to be used as soft gels, which could be lasting more than 1 week in mobile chondral defects, has made chitosan a polymer of interest in repairing bone and cartilage defects. Different types of scaffolds using chitosan in combination with mesenchymal stem cells (MSCs) are discussed. MSCs are widely used in regenerative medicine because of their regenerative ability, and recent line evidence reviewed demonstrated that the combination of MSCs with a combination of chitosan with different materials, including collagen type 1, hyaluronic acid, Poly(L-lacticacid)/gelatin/β-tricalcium phosphate, gamma-poly[glutamic acid] polyelectrolyte/titanium alloy, modified Poly(L-Lactide-co-Epsilon-Caprolactone), calcium phosphate, β-glycerophosphate hydrogel/calcium phosphate cement (CPC), and CPC-Chitosan-RGD, can increase the efficacy of using MSCs, and chitosan-based stem cell delivery can be a promising method in restoration of damaged bone and cartilage tissue.
Collapse
Affiliation(s)
- Ping Qi
- Department of General Practice, Linyi People's Hospital, Linyi, Shandong, China
| | - Zhaohui Ning
- Department of Traditional Chinese Medicine, Taian Central Hospital, Taian, Shandong, China
| | - Xiuju Zhang
- Department of General Practice, Linyi People's Hospital, Linyi, Shandong, China
| |
Collapse
|
9
|
Shin DS, Touani FK, Aboud DG, Kietzig AM, Lerouge S, Hoesli CA. Mammalian cell encapsulation in monodisperse chitosan beads using microchannel emulsification. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.130807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
10
|
Liu J, Jin L, Zhou Q, Huang W, Wang S, Huang X, Zhao X. Collagen Nanofilm-Coated Partially Deproteinized Bone Combined With Bone Mesenchymal Stem Cells for Rat Femoral Defect Repair by Bone Tissue Engineering. Ann Plast Surg 2021; 87:580-588. [PMID: 34139739 DOI: 10.1097/sap.0000000000002905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The advantages of good biocompatibility, low degradation and low antigenicity of collagen, and the osteogenic differentiation characteristics of bone mesenchymal stem cells (BMSCs) were used to promote the recovery of bone defects using partially deproteinized bone (PDPB) by bone tissue engineering (BTE). METHODS The BMSCs were identified by examining their potential for osteogenic, lipogenic, and chondrogenic differentiation. The prepared pure PDPB was ground into bone blocks 4 × 2 × 2 mm in size, which were divided into the following groups: PDPB group, PDPB + collagen group, PDPB + collagen + BMSC group, PDPB with a composite collagen nanofilm, and BMSCs injected into the tail vein. At 2, 4, 6, and 8 weeks after surgery, the effects of the implants in the different groups on bone defect repair were continuously and dynamically observed through x-ray examination, gross specimen observation, histological evaluation, and microvascularization detection. RESULTS Postoperative x-ray examination and gross specimen observation revealed that, after 4 to 8 weeks, the external contour of the graft was gradually weakened, and the transverse comparison showed that the absorption of the graft and fusion of the defect were more obvious in PDPB + collagen + BMSC group than in PDPB group and PDPB + collagen group, and the healing was better (P < 0.05). Hematoxylin and eosin staining of histological sections showed very active proliferation of trabecular hematopoietic cells in groups PDPB + collagen + BMSC and PDPB + collagen. Masson's trichrome staining for evaluation of bone defect repair showed that the mean percent area of collagen fibers was greater in PDPB + collagen + BMSC group than in the PDPB group, with degradation of the scaffold material and the completion of repair. Immunofluorescence staining showed significantly enhanced expression of the vascular marker CD31 in group C (P < 0.05). CONCLUSIONS The proposed hybrid structure of the collagen matrix and PDPB provides an ideal 3-dimensional microenvironment for patient-specific BTE and cell therapy applications. The results showed that collagen appeared to regulate MSC-mediated osteogenesis and increase the migration and invasion of BMSCs. The combination of collagen nanofilm and biological bone transplantation with BMSC transplantation enhanced the proliferation and potential of the BMSCs for bone regeneration, successfully promoting bone repair after implantation at the defect site. This method may provide a new idea for treating clinical bone defects through BTE.
Collapse
Affiliation(s)
- Jiajie Liu
- From the Department of Plastic Surgery, Kunming Medical University
| | - Liang Jin
- From the Department of Plastic Surgery, Kunming Medical University
| | - Qingzhu Zhou
- From the Department of Plastic Surgery, Kunming Medical University
| | - Wenli Huang
- From the Department of Plastic Surgery, Kunming Medical University
| | - Songmei Wang
- From the Department of Plastic Surgery, Kunming Medical University
| | - Xinwei Huang
- Key Laboratory of The Second Affiliated Hospital of Kuming Medical College, Kunming, China
| | - Xian Zhao
- From the Department of Plastic Surgery, Kunming Medical University
| |
Collapse
|
11
|
Maleas G, Mageed M. Effectiveness of Platelet-Rich Plasma and Bone Marrow Aspirate Concentrate as Treatments for Chronic Hindlimb Proximal Suspensory Desmopathy. Front Vet Sci 2021; 8:678453. [PMID: 34222402 PMCID: PMC8253571 DOI: 10.3389/fvets.2021.678453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/24/2021] [Indexed: 11/13/2022] Open
Abstract
This retrospective study aimed to evaluate the clinical effect of bone marrow aspirate concentrate (BMAC) and leukocyte rich PRP (LR-PRP) compared to horses undergoing controlled exercise alone in horses with >3 months proximal suspensory desmopathy in hindlimbs (HPSD). Nighty-three horses were divided into three groups according to the treatment: a control (n = 22), LR-PRP (n = 46), and BMAC (n = 25) group. Lameness and ultrasound scores were recorded before treatment (T0) and at 6 months (T1) post-treatment. Records horses considered sound at evaluation and level of performance were additionally registered at 12 months (T2) and 18 months (T3) after treatment. The BMAC cytology profiles from 22 horses were also analysed and compared to clinical outcomes. The results at T1 showed that 9% (2/22) of the horses in the control group were sound compared to 59% (25/46) and 84% (21/25) in the LR-PRP and BMAC groups, respectively. Additionally, ultrasound scores at T1 in the BMAC and LR-PRP groups were improved in comparison with the control group (p = 0.02). At T2, 68% of the horses in the BMAC group and 39% of the horses in the LR-PRP group had returned to the previous performance level. At T3, a significantly higher percentage of horses in the LR-PRP (43%) and BMAC (72%) group were sound when compared to the control (4.6%) group (p = 0.02). Similarly, at T3, significantly more horses of the BMAC (16/25) and of the LR-PRP (15/46) group had returned to the previous or a higher performance level compared to the control (1/22) group (p = 0.01). No correlation was found between long-term clinical outcome and cytology profiles in the BMAC group. In conclusion, long-term outcomes of treatment with LR-PRP or BMAC are significantly better than conventional treatment of the hindlimb chronic PSD in horses. Additionally, BMAC yielded better lameness scores than LR-PRP at short- and long-term follow-up.
Collapse
|
12
|
Pires JLDS, de Carvalho JJ, Pereira MJDS, Brum IDS, Nascimento ALR, dos Santos PGP, Frigo L, Fischer RG. Repair of Critical Size Bone Defects Using Synthetic Hydroxyapatite or Xenograft with or without the Bone Marrow Mononuclear Fraction: A Histomorphometric and Immunohistochemical Study in Rat Calvaria. MATERIALS 2021; 14:ma14112854. [PMID: 34073482 PMCID: PMC8199028 DOI: 10.3390/ma14112854] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/23/2021] [Accepted: 05/19/2021] [Indexed: 11/16/2022]
Abstract
Bone defects are a challenging clinical situation, and the development of hydroxyapatite-based biomaterials is a prolific research field that, in addition, can be joined by stem cells and growth factors in order to deal with the problem. This study compares the use of synthetic hydroxyapatite and xenograft, used pure or enriched with bone marrow mononuclear fraction for the regeneration of critical size bone defects in rat calvaria through histomorphometric (Masson's staining) and immunohistochemical (anti-VEGF, anti-osteopontin) analysis. Forty young adult male rats were divided into five groups (n = 8). Animals were submitted to critical size bone defects (Ø = 8 mm) in the temporoparietal region. In the control group, there was no biomaterial placement in the critical bone defects; in group 1, it was filled with synthetic hydroxyapatite; in group 2, it was filled with xenograft; in group 3, it was filled with synthetic hydroxyapatite, enriched with bone marrow mononuclear fraction (BMMF), and in group 4 it was filled with xenograft, enriched with BMMF. After eight weeks, all groups were euthanized, and histological section images were captured and analyzed. Data analysis showed that in groups 1, 2, 3 and 4 (received biomaterials and biomaterials plus BMMF), a significant enhancement in new bone matrix formation was observed in relation to the control group. However, BMMF-enriched groups did not differ from hydroxyapatite-based biomaterials-only groups. Therefore, in this experimental model, BMMF did not enhance hydroxyapatite-based biomaterials' potential to induce bone matrix and related mediators.
Collapse
Affiliation(s)
- Jorge Luís da Silva Pires
- Department of Periodontology, PhD Candidate in Periodontology, School of Dentistry, State University of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil
- Correspondence: ; Tel.: +55-21-986794126
| | - Jorge José de Carvalho
- Laboratory of Cell Ultrastructure and Tissue Biology, Department of Histology and Embryology, State University of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil; (J.J.d.C.); (M.J.d.S.P.); (I.d.S.B.); (A.L.R.N.)
| | - Mario José dos Santos Pereira
- Laboratory of Cell Ultrastructure and Tissue Biology, Department of Histology and Embryology, State University of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil; (J.J.d.C.); (M.J.d.S.P.); (I.d.S.B.); (A.L.R.N.)
| | - Igor da Silva Brum
- Laboratory of Cell Ultrastructure and Tissue Biology, Department of Histology and Embryology, State University of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil; (J.J.d.C.); (M.J.d.S.P.); (I.d.S.B.); (A.L.R.N.)
| | - Ana Lucia Rosa Nascimento
- Laboratory of Cell Ultrastructure and Tissue Biology, Department of Histology and Embryology, State University of Rio de Janeiro, Rio de Janeiro 20550-900, Brazil; (J.J.d.C.); (M.J.d.S.P.); (I.d.S.B.); (A.L.R.N.)
| | - Paulo Gonçalo Pinto dos Santos
- Department of Periodontology, School of Dentistry, State University of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (P.G.P.d.S.); (R.G.F.)
| | - Lucio Frigo
- Department of Periodontology, School of Dentistry, Universidade Guarulhos, Guarulhos 07023-070, Brazil;
| | - Ricardo Guimaraes Fischer
- Department of Periodontology, School of Dentistry, State University of Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (P.G.P.d.S.); (R.G.F.)
- Department of Periodontology, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro 22451-900, Brazil
| |
Collapse
|
13
|
Du F, Wang Q, Ouyang L, Wu H, Yang Z, Fu X, Liu X, Yan L, Cao Y, Xiao R. Comparison of concentrated fresh mononuclear cells and cultured mesenchymal stem cells from bone marrow for bone regeneration. Stem Cells Transl Med 2020; 10:598-609. [PMID: 33341102 PMCID: PMC7980203 DOI: 10.1002/sctm.20-0234] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/20/2020] [Accepted: 10/25/2020] [Indexed: 12/21/2022] Open
Abstract
Autologous bone marrow mononuclear cell (BMMNC) transplantation has been widely studied in recent years. The fresh cell cocktail in BMMNCs, without going through the in vitro culture process, helps to establish a stable microenvironment for osteogenesis, and each cell type may play a unique role in bone regeneration. Our study compared the efficacy of concentrated fresh BMMNCs and cultured bone marrow‐derived mesenchymal stem cells (BMSCs) in Beagle dogs for the first time. Fifteen‐millimeter segmental bone defects were created in the animals' tibia bones. In BMMNCs group, the defects were repaired with concentrated fresh BMMNCs combined with β‐TCP (n = 5); in cultured BMSC group, with in vitro cultured and osteo‐induced BMSCs combined with β‐TCP (n = 5); in scaffold‐only group, with a β‐TCP graft alone (n = 5); and in blank group, nothing was grafted (n = 3). The healing process was monitored by X‐rays and single photon emission computed tomography. The animals were sacrificed 12 months after surgery and their tibias were harvested and analyzed by microcomputed tomography and hard tissue histology. Moreover, the microstructure, chemical components, and microbiomechanical properties of the regenerated bone tissue were explored by multiphoton microscopy, Raman spectroscopy and nanoindentation. The results showed that BMMNCs group promoted much more bone regeneration than cultured BMSC group. The grafts in BMMNCs group were better mineralized, and they had collagen arrangement and microbiomechanical properties similar to the contralateral native tibia bone. These results indicate that concentrated fresh bone marrow mononuclear cells may be superior to in vitro expanded stem cells in segmental bone defect repair.
Collapse
Affiliation(s)
- Fengzhou Du
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China.,Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Qian Wang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Long Ouyang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Huanhuan Wu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Zhigang Yang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Xia Liu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Li Yan
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Yilin Cao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
14
|
Uchikawa E, Yoshizawa M, Li X, Matsumura N, Li N, Chen K, Kagami H. Tooth transplantation with a β-tricalcium phosphate scaffold accelerates bone formation and periodontal tissue regeneration. Oral Dis 2020; 27:1226-1237. [PMID: 32881188 DOI: 10.1111/odi.13634] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 08/17/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Although tooth transplantation is a useful treatment option as a substitute for a missing tooth, transplantation to a narrow alveolar ridge is not feasible. In this study, we tested a tissue engineering approach simultaneously with tooth transplantation using a scaffold or a combination with cells to accelerate bone formation and periodontal tissue regeneration. MATERIALS AND METHODS Bone marrow mononuclear cells (BM-MNCs) were harvested from C57BL/6J mice. The upper first or the second molar of 3-week-old C57BL/6J mice and a β-tricalcium phosphate (β-TCP) scaffold were transplanted with BM-MNCs (MNC group) or without BM-MNCs (β-TCP group) into the thigh muscle of syngeneic mice. The tooth alone was also transplanted (control group). After 4 weeks, the transplants were harvested and analyzed. RESULTS Bone volume was significantly larger in the MNC and the β-TCP groups than that in the control group, and the newly formed bone was observed on the lateral wall of the root. Compared with the control group, the MNC group showed a larger trabecular thickness and fractal dimension. CONCLUSION This study showed accelerated bone formation and periodontal tissue regeneration when tooth transplantation was performed with a β-TCP scaffold. BM-MNCs may accelerate bone maturation, while the effect on bone formation was limited.
Collapse
Affiliation(s)
- Eri Uchikawa
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Matsumoto Dental University, Shiojiri, Japan.,Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University, Shiojiri, Japan
| | - Michiko Yoshizawa
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Matsumoto Dental University, Shiojiri, Japan.,Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University, Shiojiri, Japan
| | - Xianqi Li
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Matsumoto Dental University, Shiojiri, Japan.,Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University, Shiojiri, Japan
| | - Nahomi Matsumura
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Matsumoto Dental University, Shiojiri, Japan.,Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University, Shiojiri, Japan
| | - Ni Li
- Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University, Shiojiri, Japan
| | - Kai Chen
- Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University, Shiojiri, Japan
| | - Hideaki Kagami
- Department of Hard Tissue Research, Graduate School of Oral Medicine, Matsumoto Dental University, Shiojiri, Japan.,Division of Hard Tissue Research, Institute of Oral Science, Matsumoto Dental University, Shiojiri, Japan.,Department of General Medicine, IMSUT Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
15
|
Bai H, Kyu-Cheol N, Wang Z, Cui Y, Liu H, Liu H, Feng Y, Zhao Y, Lin Q, Li Z. Regulation of inflammatory microenvironment using a self-healing hydrogel loaded with BM-MSCs for advanced wound healing in rat diabetic foot ulcers. J Tissue Eng 2020; 11:2041731420947242. [PMID: 32913623 PMCID: PMC7444096 DOI: 10.1177/2041731420947242] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023] Open
Abstract
A diabetic foot ulcer (DFUs) is a state of prolonged chronic inflammation, which can result in amputation. Different from normal skin wounds, various commercially available dressings have not sufficiently improved the healing of DFUs. In this study, a novel self-healing hydrogel was prepared by in situ crosslinking of N-carboxyethyl chitosan (N-chitosan) and adipic acid dihydrazide (ADH) with hyaluronic acid-aldehyde (HA-ALD), to provide a moist and inflammatory relief environment to promote stem cell proliferation or secretion of growth factors, thus accelerating wound healing. The results demonstrated that this injectable and self-healing hydrogel has excellent swelling properties, stability, and mechanical properties. This biocompatible hydrogel stimulated secretion of growth factors from bone marrow mesenchymal stem cells (BM-MSCs) and regulated the inflammatory environment by inhibiting the expression of M1 macrophages and promoting the expression of M2 macrophages, resulting in granulation tissue formation, collagen deposition, nucleated cell proliferation, neovascularization, and enhanced diabetic wound healing. This study showed that N-chitosan/HA-ALD hydrogel could be used as a multifunctional injectable wound dressing to regulate chronic inflammation and provide an optimal environment for BM-MSCs to promote diabetic wound healing.
Collapse
Affiliation(s)
- Haotian Bai
- Department of Orthopedics, Hallym University, Chuncheon, Gangwon-do, Korea
| | - Noh Kyu-Cheol
- Department of Orthopedics, Hallym University, Chuncheon, Gangwon-do, Korea
| | - Zhonghan Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Yutao Cui
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - He Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Hou Liu
- State Key Lab of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, P. R. China
| | - Yubin Feng
- State Key Lab of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, P. R. China
| | - Yue Zhao
- State Key Lab of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, P. R. China
| | - Quan Lin
- State Key Lab of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun, Jilin, P. R. China
| | - Zuhao Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
- Department of Pain, Renji Hospital, South Campus, Shanghai Jiaotong University, Shanghai, P. R. China
| |
Collapse
|
16
|
Hypoxia influences the effects of magnesium degradation products on the interactions between endothelial and mesenchymal stem cells. Acta Biomater 2020; 101:624-636. [PMID: 31622779 DOI: 10.1016/j.actbio.2019.10.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 10/07/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023]
Abstract
Biodegradable materials like well-documented Magnesium (Mg) are promising for their biocompatibility and tissue regeneration. Since Mg degradation is reported to be oxygen related, the effects of Mg were hypothesised to be influenced by oxygen. As two vital components of bone marrow, endothelial cells (EC) and mesenchymal stem cells (MSC), their interactions represent high scientific interest for tissue engineering and biodegradable Mg application. Human umbilical cord perivascular (HUCPV) and umbilical vein endothelial cell (HUVEC) were selected as sources of MSC and EC, respectively. Two types of coculture models were established to represent different phases of MSC-EC interaction: (i) where cells were physically separated thanks to a transwell and (ii) where cells were allowed to have heterotypic cellular contacts. Cell migration, gene, cytokines, and proliferation were investigated in HUCPV-HUVEC coculture using DNA, flow cytometry, wound healing assay, semi-quantitative real-time polymerase chain reaction (qRT-PCR), and enzyme-linked immunosorbent assay (ELISA). Mg degradation products increased HUCPV migration in transwell under hypoxia. Oxygen tension changed the gene regulation of migratory, angiogenetic or osteogenic regulators. Under contacting coculture and hypoxia, Mg degradation products remarkably increased cytokines (e.g., c-c motif chemokine ligand 2 and vascular endothelial growth factor) and MSC mineralisation. Mg degradation products decreased and increased the MSC proliferation in transwell and in heterotypic-contact coculture, respectively. In summary, this study indicates the roles of low oxygen and heterotypic contact to effects of Mg materials facilitating HUVEC and HUCPV. STATEMENT OF SIGNIFICANCE.
Collapse
|
17
|
Alinejad Y, Bitar CME, Martinez Villegas K, Perignon S, Hoesli CA, Lerouge S. Chitosan Microbeads Produced by One-Step Scalable Stirred Emulsification: A Promising Process for Cell Therapy Applications. ACS Biomater Sci Eng 2019; 6:288-297. [PMID: 33463194 DOI: 10.1021/acsbiomaterials.9b01638] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Cell microencapsulation is a promising approach to improve cell therapy outcomes by protecting injected cells from rapid dispersion and allowing bidirectional diffusion of nutrients, oxygen, and waste that promote cell survival in the target tissues. Here, we describe a simple and scalable emulsification method to encapsulate animal cells in chitosan microbeads using thermosensitive gel formulations without any chemical modification and cross-linker. The process consists of a water-in-oil emulsion where the aqueous phase droplets contain cells (L929 fibroblasts or human mesenchymal stromal cells), chitosan acidic solution and gelling agents (sodium hydrogen carbonate and phosphate buffer or beta-glycerophosphate). The oil temperature is maintained at 37 °C, allowing rapid physical gelation of the microbeads. Alginate beads prepared with the same method were used as a control. Microbeads with a diameter of 300-450 μm were successfully produced. Chitosan and alginate (2% w/v) microbeads presented similar rigidity in compression, but chitosan microbeads endured >80% strain without rupture, while alginate microbeads presented fragile breakage at <50% strain. High cell viability and metabolic activity were observed after up to 7 days in culture for encapsulated cells. Mesenchymal stromal cells encapsulated in chitosan microbeads released higher amounts of the vascular endothelial growth factor after 24 h compared to the cells encapsulated in manually cast macrogels. Moreover, microbeads were injectable through 23G needles without significant deformation or rupture. The emulsion-generated chitosan microbeads are a promising delivery vehicle for therapeutic cells because of their cytocompatibility, biodegradation, mechanical strength, and injectability. Clinical-scale encapsulation of therapeutic cells such as mesenchymal stromal cells in chitosan microbeads can readily be achieved using this simple and scalable emulsion-based process.
Collapse
Affiliation(s)
- Yasaman Alinejad
- Laboratory of Endovascular Biomaterials (LBeV), Centre de recherche du CHUM (CRCHUM), 900 Saint-Denis Street, Montreal, Quebec H2X 0A9, Canada.,Department of Mechanical Engineering, École de technologie supérieure (ETS), 1100 Notre-Dame West, Montreal, Quebec H3C 1K3, Canada
| | - Christina M E Bitar
- Department of Chemical Engineering, McGill University, Wong Building, 3610 University Street #3060, Montreal, Quebec H3A 0C5, Canada
| | - Karina Martinez Villegas
- Laboratory of Endovascular Biomaterials (LBeV), Centre de recherche du CHUM (CRCHUM), 900 Saint-Denis Street, Montreal, Quebec H2X 0A9, Canada.,Department of Mechanical Engineering, École de technologie supérieure (ETS), 1100 Notre-Dame West, Montreal, Quebec H3C 1K3, Canada
| | - Sarah Perignon
- Laboratory of Endovascular Biomaterials (LBeV), Centre de recherche du CHUM (CRCHUM), 900 Saint-Denis Street, Montreal, Quebec H2X 0A9, Canada.,Department of Mechanical Engineering, École de technologie supérieure (ETS), 1100 Notre-Dame West, Montreal, Quebec H3C 1K3, Canada
| | - Corinne A Hoesli
- Department of Chemical Engineering, McGill University, Wong Building, 3610 University Street #3060, Montreal, Quebec H3A 0C5, Canada
| | - Sophie Lerouge
- Laboratory of Endovascular Biomaterials (LBeV), Centre de recherche du CHUM (CRCHUM), 900 Saint-Denis Street, Montreal, Quebec H2X 0A9, Canada.,Department of Mechanical Engineering, École de technologie supérieure (ETS), 1100 Notre-Dame West, Montreal, Quebec H3C 1K3, Canada
| |
Collapse
|
18
|
Bayat F, Karimi AR. Design of photodynamic chitosan hydrogels bearing phthalocyanine-colistin conjugate as an antibacterial agent. Int J Biol Macromol 2019; 129:927-935. [DOI: 10.1016/j.ijbiomac.2019.02.081] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 02/05/2019] [Accepted: 02/13/2019] [Indexed: 01/15/2023]
|
19
|
Daley EL, Kuttig J, Stegemann JP. Development of Modular, Dual-Perfused Osteochondral Constructs for Cartilage Repair. Tissue Eng Part C Methods 2019; 25:127-136. [PMID: 30724134 PMCID: PMC6457327 DOI: 10.1089/ten.tec.2018.0356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/25/2019] [Indexed: 11/13/2022] Open
Abstract
IMPACT STATEMENT This study describes methods for fabricating, culturing, and characterizing modular microbeads containing progenitor cells that can be used to create osteochondral tissue constructs. Such biphasic engineered tissues were cultured in a low flow rate perfusion bioreactor chamber to maintain tissue-specific differentiation while allowing development of the osteochondral interface.
Collapse
Affiliation(s)
- Ethan L.H. Daley
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Jochen Kuttig
- Faculty of Medicine, University of Tübingen, Tübingen, Germany
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
20
|
Al-Ahmady HH, Abd Elazeem AF, Bellah Ahmed NEM, Shawkat WM, Elmasry M, Abdelrahman MA, Abderazik MA. Combining autologous bone marrow mononuclear cells seeded on collagen sponge with Nano Hydroxyapatite, and platelet-rich fibrin: Reporting a novel strategy for alveolar cleft bone regeneration. J Craniomaxillofac Surg 2018; 46:1593-1600. [DOI: 10.1016/j.jcms.2018.05.049] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/03/2018] [Accepted: 05/25/2018] [Indexed: 01/08/2023] Open
|
21
|
Gansau J, Kelly L, Buckley CT. Influence of key processing parameters and seeding density effects of microencapsulated chondrocytes fabricated using electrohydrodynamic spraying. Biofabrication 2018; 10:035011. [PMID: 29888707 DOI: 10.1088/1758-5090/aacb95] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell delivery and leakage during injection remains a challenge for cell-based intervertebral disc regeneration strategies. Cellular microencapsulation may offer a promising approach to overcome these limitations by providing a protective niche during intradiscal injection. Electrohydrodynamic spraying (EHDS) is a versatile one-step approach for microencapsulation of cells using a high voltage electric field. The primary objective of this work was to characterise key processing parameters such as applied voltage (0, 5, 10 or 15 kV), emitter needle gauge (21, 26 or 30 G), alginate concentration (1%, 2% or 3%) and flow rate (50, 100, 250 or 500 μl min-1) to regulate the size and morphology of alginate microcapsules as well as subsequent cell viability when altering these parameters. The effect of initial cell seeding density (5, 10 and 20 × 106 cells ml-1) on subsequent matrix accumulation of microencapsulated articular chondrocytes was also evaluated. Results showed that increasing alginate concentration and thus viscosity increased overall microcapsule size but also affected the geometry towards ellipsoidal-shaped gels. Altering the electric field strength and needle diameter regulated microcapsule size towards a smaller diameter with increasing voltage and smaller needle diameter. Needle size did not appear to affect cell viability when operating with lower alginate concentrations (1% and 2%), although higher concentrations (3%) and thus higher viscosity hydrogels resulted in diminished viability with decreasing needle diameter. Increasing cell density resulted in decreased cell viability and a concomitant decrease in DNA content, perhaps due to competing nutrient demands as a result of more closely packed cells. However, higher cell densities resulted in increased levels of extracellular matrix accumulated. Overall, this work highlights the potential of EHDS as a controllable and versatile approach to fabricate microcapsules for injectable delivery which can be used in a variety of applications such as drug development or cell therapies.
Collapse
Affiliation(s)
- Jennifer Gansau
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, Dublin, Ireland. School of Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland
| | | | | |
Collapse
|
22
|
Liu H, Wang C, Li C, Qin Y, Wang Z, Yang F, Li Z, Wang J. A functional chitosan-based hydrogel as a wound dressing and drug delivery system in the treatment of wound healing. RSC Adv 2018; 8:7533-7549. [PMID: 35539132 PMCID: PMC9078458 DOI: 10.1039/c7ra13510f] [Citation(s) in RCA: 513] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/12/2018] [Indexed: 12/18/2022] Open
Abstract
Functional active wound dressings are expected to provide a moist wound environment, offer protection from secondary infections, remove wound exudate and accelerate tissue regeneration, as well as to improve the efficiency of wound healing. Chitosan-based hydrogels are considered as ideal materials for enhancing wound healing owing to their biodegradable, biocompatible, non-toxic, antimicrobial, biologically adhesive, biological activity and hemostatic effects. Chitosan-based hydrogels have been demonstrated to promote wound healing at different wound healing stages, and also can alleviate the factors against wound healing (such as excessive inflammatory and chronic wound infection). The unique biological properties of a chitosan-based hydrogel enable it to serve as both a wound dressing and as a drug delivery system (DDS) to deliver antibacterial agents, growth factors, stem cells and so on, which could further accelerate wound healing. For various kinds of wounds, chitosan-based hydrogels are able to promote the effectiveness of wound healing by modifying or combining with other polymers, and carrying different types of active substances. In this review, we will take a close look at the application of chitosan-based hydrogels in wound dressings and DDS to enhance wound healing.
Collapse
Affiliation(s)
- He Liu
- Orthopaedic Medical Center, The Second Hospital of Jilin University Changchun 130041 P. R. China
| | - Chenyu Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University Changchun 130041 P. R. China
- Hallym University 1Hallymdaehak-gil Chuncheon Gangwon-do 200-702 Korea
| | - Chen Li
- Orthopaedic Medical Center, The Second Hospital of Jilin University Changchun 130041 P. R. China
| | - Yanguo Qin
- Orthopaedic Medical Center, The Second Hospital of Jilin University Changchun 130041 P. R. China
| | - Zhonghan Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University Changchun 130041 P. R. China
| | - Fan Yang
- Orthopaedic Medical Center, The Second Hospital of Jilin University Changchun 130041 P. R. China
| | - Zuhao Li
- Orthopaedic Medical Center, The Second Hospital of Jilin University Changchun 130041 P. R. China
| | - Jincheng Wang
- Orthopaedic Medical Center, The Second Hospital of Jilin University Changchun 130041 P. R. China
| |
Collapse
|
23
|
An Injectable Oxygen Release System to Augment Cell Survival and Promote Cardiac Repair Following Myocardial Infarction. Sci Rep 2018; 8:1371. [PMID: 29358595 PMCID: PMC5778078 DOI: 10.1038/s41598-018-19906-w] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/10/2018] [Indexed: 01/15/2023] Open
Abstract
Oxygen deficiency after myocardial infarction (MI) leads to massive cardiac cell death. Protection of cardiac cells and promotion of cardiac repair are key therapeutic goals. These goals may be achieved by re-introducing oxygen into the infarcted area. Yet current systemic oxygen delivery approaches cannot efficiently diffuse oxygen into the infarcted area that has extremely low blood flow. In this work, we developed a new oxygen delivery system that can be delivered specifically to the infarcted tissue, and continuously release oxygen to protect the cardiac cells. The system was based on a thermosensitive, injectable and fast gelation hydrogel, and oxygen releasing microspheres. The fast gelation hydrogel was used to increase microsphere retention in the heart tissue. The system was able to continuously release oxygen for 4 weeks. The released oxygen significantly increased survival of cardiac cells under the hypoxic condition (1% O2) mimicking that of the infarcted hearts. It also reduced myofibroblast formation under hypoxic condition (1% O2). After implanting into infarcted hearts for 4 weeks, the released oxygen significantly augmented cell survival, decreased macrophage density, reduced collagen deposition and myofibroblast density, and stimulated tissue angiogenesis, leading to a significant increase in cardiac function.
Collapse
|
24
|
Magnetic Targeted Delivery of Induced Pluripotent Stem Cells Promotes Articular Cartilage Repair. Stem Cells Int 2017; 2017:9514719. [PMID: 29441091 PMCID: PMC5758849 DOI: 10.1155/2017/9514719] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 11/20/2017] [Indexed: 12/24/2022] Open
Abstract
Cartilage regeneration treatments using stem cells are associated with problems due to the cell source and the difficulty of delivering the cells to the cartilage defect. We consider labeled induced pluripotent stem (iPS) cells to be an ideal source of cells for tissue regeneration, and if iPS cells could be delivered only into cartilage defects, it would be possible to repair articular cartilage. Consequently, we investigated the effect of magnetically labeled iPS (m-iPS) cells delivered into an osteochondral defect by magnetic field on the repair of articular cartilage. iPS cells were labeled magnetically and assessed for maintenance of pluripotency by their ability to form embryoid bodies in vitro and to form teratomas when injected subcutaneously into nude rats. These cells were delivered specifically into cartilage defects in nude rats using a magnetic field. The samples were graded according to the histologic grading score for cartilage regeneration. m-iPS cells differentiated into three embryonic germ layers and formed teratomas in the subcutaneous tissue. The histologic grading score was significantly better in the treatment group compared to the control group. m-iPS cells maintained pluripotency, and the magnetic delivery system proved useful and safe for cartilage repair using iPS cells.
Collapse
|
25
|
Jiang T, Xu G, Wang Q, Yang L, Zheng L, Zhao J, Zhang X. In vitro expansion impaired the stemness of early passage mesenchymal stem cells for treatment of cartilage defects. Cell Death Dis 2017; 8:e2851. [PMID: 28569773 PMCID: PMC5520885 DOI: 10.1038/cddis.2017.215] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 02/08/2023]
Abstract
In vitro cultured autologous mesenchymal stem cells (MSCs) within passage 5 have been approved for clinical application in stem cell-based treatment of cartilage defects. However, their chondrogenic potential has not yet been questioned or verified. In this study, the chondrogenic potential of bone marrow MSCs at passage 3 (P3 BMSCs) was investigated both in cartilage repair and in vitro, with freshly isolated bone marrow mononuclear cells (BMMNCs) as controls. The results showed that P3 BMSCs were inferior to BMMNCs not only in their chondrogenic differentiation ability but also as candidates for long-term repair of cartilage defects. Compared with BMMNCs, P3 BMSCs presented a decay in telomerase activity and a change in chromosomal morphology with potential anomalous karyotypes, indicating senescence. In addition, interindividual variability in P3 BMSCs is much higher than in BMMNCs, demonstrating genomic instability. Interestingly, remarkable downregulation in cell cycle, DNA replication and mismatch repair (MMR) pathways as well as in multiple genes associated with telomerase activity and chromosomal stability were found in P3 BMSCs. This result indicates that telomerase and chromosome anomalies might originate from expansion, leading to impaired stemness and pluripotency of stem cells. In vitro culture and expansion are not recommended for cell-based therapy, and fresh BMMNCs are the first choice.
Collapse
Affiliation(s)
- Tongmeng Jiang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Guojie Xu
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning 530021, China
| | - Lihui Yang
- School of Nursing, Guangxi Medical University, Nanning 530021, China
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Center of Guangxi Biological Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Collaborative Innovation Center of Guangxi Biological Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
- Guangxi Key Laboratory of Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, China
| |
Collapse
|
26
|
Rioja AY, Daley ELH, Habif JC, Putnam AJ, Stegemann JP. Distributed vasculogenesis from modular agarose-hydroxyapatite-fibrinogen microbeads. Acta Biomater 2017; 55:144-152. [PMID: 28365482 DOI: 10.1016/j.actbio.2017.03.050] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 03/03/2017] [Accepted: 03/28/2017] [Indexed: 11/29/2022]
Abstract
Critical limb ischemia impairs circulation to the extremities, causing pain, disrupted wound healing, and potential tissue necrosis. Therapeutic angiogenesis seeks to repair the damaged microvasculature directly to restore blood flow. In this study, we developed modular, micro-scale constructs designed to possess robust handling qualities, allow in vitro pre-culture, and promote microvasculature formation. The microbead matrix consisted of an agarose (AG) base to prevent aggregation, combined with cell-adhesive components of fibrinogen (FGN) and/or hydroxyapatite (HA). Microbeads encapsulating a co-culture of human umbilical vein endothelial cells (HUVEC) and fibroblasts were prepared and characterized. Microbeads were generally 80-100µm in diameter, and the size increased with the addition of FGN and HA. Addition of HA increased the yield of microbeads, as well as the homogeneity of distribution of FGN within the matrix. Cell viability was high in all microbead types. When cell-seeded microbeads were embedded in fibrin hydrogels, HUVEC sprouting and inosculation between neighboring microbeads were observed over seven days. Pre-culture of microbeads for an additional seven days prior to embedding in fibrin resulted in significantly greater HUVEC network length in AG+HA+FGN microbeads, as compared to AG, AG+HA or AG+FGN microbeads. Importantly, composite microbeads resulted in more even and widespread endothelial network formation, relative to control microbeads consisting of pure fibrin. These results demonstrate that AG+HA+FGN microbeads support HUVEC sprouting both within and between adjacent microbeads, and can promote distributed vascularization of an external matrix. Such modular microtissues may have utility in treating ischemic tissue by rapidly re-establishing a microvascular network. STATEMENT OF SIGNIFICANCE Critical limb ischemia (CLI) is a chronic disease that can lead to tissue necrosis, amputation, and death. Cell-based therapies are being explored to restore blood flow and prevent the complications of CLI. In this study, we developed small, non-aggregating agarose-hydroxyapatite-fibrinogen microbeads that contained endothelial cells and fibroblasts. Microbeads were easy to handle and culture, and endothelial sprouts formed within and between microbeads. Our data demonstrates that the composition of the microbead matrix altered the degree of endothelial sprouting, and that the addition of hydroxyapatite and fibrinogen resulted in more distributed sprouting compared to pure fibrin microbeads. The microbead format and control of the matrix formulation may therefore be useful in developing revascularization strategies for the treatment of ischemic disease.
Collapse
Affiliation(s)
- Ana Y Rioja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Ethan L H Daley
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Julia C Habif
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Andrew J Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
27
|
Wise JK, Alford AI, Goldstein SA, Stegemann JP. Synergistic enhancement of ectopic bone formation by supplementation of freshly isolated marrow cells with purified MSC in collagen-chitosan hydrogel microbeads. Connect Tissue Res 2016; 57:516-525. [PMID: 26337827 PMCID: PMC4864208 DOI: 10.3109/03008207.2015.1072519] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Bone marrow-derived mesenchymal stem cells (MSC) can differentiate osteogenic lineages, but their tissue regeneration ability is inconsistent. The bone marrow mononuclear cell (BMMC) fraction of adult bone marrow contains a variety of progenitor cells that may potentiate tissue regeneration. This study examined the utility of BMMC, both alone and in combination with purified MSC, as a cell source for bone regeneration. METHODS Fresh BMMC, culture-expanded MSC, and a combination of BMMC and MSC were encapsulated in collagen-chitosan hydrogel microbeads for pre-culture and minimally invasive delivery. Microbeads were cultured in growth medium for 3 days, and then in either growth or osteogenic medium for 17 days prior to subcutaneous injection in the rat dorsum. RESULTS MSC remained viable in microbeads over 17 days in pre-culture, while some of the BMMC fraction were nonviable. After 5 weeks of implantation, microCT and histology showed that supplementation of BMMC with MSC produced a strong synergistic effect on the volume of ectopic bone formation, compared to either cell source alone. Microbeads containing only fresh BMMC or only cultured MSC maintained in osteogenic medium resulted in more bone formation than their counterparts cultured in growth medium. Histological staining showed evidence of residual microbead matrix in undifferentiated samples and indications of more advanced tissue remodeling in differentiated samples. CONCLUSIONS These data suggest that components of the BMMC fraction can act synergistically with predifferentiated MSC to potentiate ectopic bone formation. The microbead system may have utility in delivering desired cell populations in bone regeneration applications.
Collapse
Affiliation(s)
- Joel K. Wise
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Andrea I. Alford
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Steven A. Goldstein
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA,Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Jan P. Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
28
|
Xie L, Mao M, Zhou L, Jiang B. Spheroid Mesenchymal Stem Cells and Mesenchymal Stem Cell-Derived Microvesicles: Two Potential Therapeutic Strategies. Stem Cells Dev 2016; 25:203-13. [PMID: 26575103 DOI: 10.1089/scd.2015.0278] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Lili Xie
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Mao Mao
- Departments of Ophthalmology and Anatomy, Institute for Human Genetics, UCSF School of Medicine, San Francisco, California
| | - Liang Zhou
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Bing Jiang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
29
|
Tiruvannamalai Annamalai R, Mertz DR, Daley ELH, Stegemann JP. Collagen Type II enhances chondrogenic differentiation in agarose-based modular microtissues. Cytotherapy 2016; 18:263-77. [PMID: 26794716 PMCID: PMC4724061 DOI: 10.1016/j.jcyt.2015.10.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 10/04/2015] [Accepted: 10/13/2015] [Indexed: 12/14/2022]
Abstract
BACKGROUND AIMS Cell-based therapies have made an impact on the treatment of osteoarthritis; however, the repair and regeneration of thick cartilage defects is an important and growing clinical problem. Next-generation therapies that combine cells with biomaterials may provide improved outcomes. We have developed modular microenvironments that mimic the composition of articular cartilage as a delivery system for consistently differentiated cells. METHODS Human bone marrow-derived mesenchymal stem cells (MSC) were embedded in modular microbeads consisting of agarose (AG) supplemented with 0%, 10% and 20% collagen Type II (COL-II) using a water-in-oil emulsion technique. AG and AG/COL-II microbeads were characterized in terms of their structural integrity, size distribution and protein content. The viability of embedded MSC and their ability to differentiate into osteogenic, adipogenic and chondrogenic lineages over 3 weeks in culture were also assessed. RESULTS Microbeads made with <20% COL-II were robust, generally spheroidal in shape and 80 ± 10 µm in diameter. MSC viability in microbeads was consistently high over a week in culture, whereas viability in corresponding bulk hydrogels decreased with increasing COL-II content. Osteogenic differentiation of MSC was modestly supported in both AG and AG/COL-II microbeads, whereas adipogenic differentiation was strongly inhibited in COL-II containing microbeads. Chondrogenic differentiation of MSC was clearly promoted in microbeads containing COL-II, compared with pure AG matrices. CONCLUSIONS Inclusion of collagen Type II in agarose matrices in microbead format can potentiate chondrogenic differentiation of human MSC. Such compositionally tailored microtissues may find utility for cell delivery in next-generation cartilage repair therapies.
Collapse
Affiliation(s)
| | - David R Mertz
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Ethan L H Daley
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
30
|
Bornes TD, Jomha NM, Mulet-Sierra A, Adesida AB. Optimal Seeding Densities for In Vitro Chondrogenesis of Two- and Three-Dimensional-Isolated and -Expanded Bone Marrow-Derived Mesenchymal Stromal Stem Cells Within a Porous Collagen Scaffold. Tissue Eng Part C Methods 2016; 22:208-20. [PMID: 26651081 DOI: 10.1089/ten.tec.2015.0365] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal stem cells (BMSCs) are a promising cell source for treating articular cartilage defects. The objective of this study was to assess the impact of cell seeding density within a collagen I scaffold on in vitro BMSC chondrogenesis following isolation and expansion in two-dimensional (2D) and three-dimensional (3D) environments. It was hypothesized that both expansion protocols would produce BMSCs capable of hyaline-like chondrogenesis with an optimal seeding density of 10 × 10(6) cells/cm(3). Ovine BMSCs were isolated in a 2D environment by plastic adherence, expanded to passage two in flasks containing an expansion medium, and seeded within collagen I scaffolds at densities of 50, 10, 5, 1, and 0.5 × 10(6) BMSCs/cm(3). For 3D isolation and expansion, aspirates containing known quantities of mononucleated cells (bone marrow-derived mononucleated cells [BMNCs]) were seeded on scaffolds at 50, 10, 5, 1, and 0.5 × 10(6) BMNCs/cm(3) and cultured in the expansion medium for an equivalent duration to 2D expansion. Constructs were differentiated in vitro in the chondrogenic medium for 21 days and assessed with reverse-transcription quantitative polymerase chain reaction, safranin O staining, histological scoring using the Bern Score, collagen immunofluorescence, and glycosaminoglycan (GAG) quantification. Two-dimensional-expanded BMSCs seeded at all densities were capable of proteoglycan production and displayed increased expressions of aggrecan and collagen II messenger RNA (mRNA) relative to predifferentiation controls. Collagen II deposition was apparent in scaffolds seeded at 0.5-10 × 10(6) BMSCs/cm(3). Chondrogenesis of 2D-expanded BMSCs was most pronounced in scaffolds seeded at 5-10 × 10(6) BMSCs/cm(3) based on aggrecan and collagen II mRNA, safranin O staining, Bern Score, total GAG, and GAG/deoxyribonucleic acid (DNA). For 3D-expanded BMSC-seeded scaffolds, increased aggrecan and collagen II mRNA expressions relative to controls were noted with all densities. Proteoglycan deposition was present in scaffolds seeded at 0.5-50 × 10(6) BMNCs/cm(3), while collagen II deposition occurred in scaffolds seeded at 10-50 × 10(6) BMNCs/cm(3). The highest levels of aggrecan and collagen II mRNA, Bern Score, total GAG, and GAG/DNA occurred with seeding at 50 × 10(6) BMNCs/cm(3). Within a collagen I scaffold, 2D- and 3D-expanded BMSCs are capable of hyaline-like chondrogenesis with optimal cell seeding densities of 5-10 × 10(6) BMSCs/cm(3) and 50 × 10(6) BMNCs/cm(3), respectively.
Collapse
Affiliation(s)
- Troy D Bornes
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| | - Nadr M Jomha
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| | - Aillette Mulet-Sierra
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| | - Adetola B Adesida
- Divisions of Orthopaedic Surgery and Surgical Research, Department of Surgery, University of Alberta , Edmonton, Canada
| |
Collapse
|
31
|
Rioja AY, Tiruvannamalai Annamalai R, Paris S, Putnam AJ, Stegemann JP. Endothelial sprouting and network formation in collagen- and fibrin-based modular microbeads. Acta Biomater 2016; 29:33-41. [PMID: 26481042 PMCID: PMC4681647 DOI: 10.1016/j.actbio.2015.10.022] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Revised: 09/04/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022]
Abstract
A modular tissue engineering approach may have advantages over current therapies in providing rapid and sustained revascularization of ischemic tissue. In this study, modular protein microbeads were prepared from pure fibrin (FIB) and collagen-fibrin composites (COL-FIB) using a simple water-in-oil emulsification technique. Human endothelial cells and fibroblasts were embedded directly in the microbead matrix. The resulting microbeads were generally spheroidal with a diameter of 100-200μm. Cell viability was high (75-80% viable) in microbeads, but was marginally lower than in bulk hydrogels of corresponding composition (85-90% viable). Cell proliferation was significantly greater in COL-FIB microbeads after two weeks in culture, compared to pure FIB microbeads. Upon embedding of microbeads in a surrounding fibrin hydrogel, endothelial cell networks formed inside the microbead matrix and extended into the surrounding matrix. The number of vessel segments, average segment length, and number of branch points was higher in FIB samples, compared to COL-FIB samples, resulting in significantly longer total vessel networks. Anastomosis of vessel networks from adjacent microbeads was also observed. These studies demonstrate that primitive vessel networks can be formed by modular protein microbeads containing embedded endothelial cells and fibroblasts. Such microbeads may find utility as prevascularized tissue modules that can be delivered minimally invasively as a therapy to restore blood flow to ischemic tissues. STATEMENT OF SIGNIFICANCE Vascularization is critically important for tissue engineering and regenerative medicine, and materials that support and/or promote neovascularization are of value both for translational applications and for mechanistic studies and discovery-based research. Therefore, we fabricated small modular microbeads formulated from pure fibrin (FIB) and collagen-fibrin (COL-FIB) containing endothelial cells and supportive fibroblasts. We explored how cells encapsulated within these materials form microvessel-like networks both within and outside of the microbeads when embedded in larger 3D matrices. FIB microbeads were found to initiate more extensive sprouting into the surrounding ECM in vitro. These results represent an important step towards our goal of developing injectable biomaterial modules containing preformed vascular units that can rapidly restore vascularization to an ischemic tissue in vivo.
Collapse
Affiliation(s)
- Ana Y Rioja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States
| | | | - Spencer Paris
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States
| | - Andrew J Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States.
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, United States.
| |
Collapse
|
32
|
Daley EL, Coleman RM, Stegemann JP. Biomimetic microbeads containing a chondroitin sulfate/chitosan polyelectrolyte complex for cell-based cartilage therapy. J Mater Chem B 2015; 3:7920-7929. [PMID: 26693016 DOI: 10.1039/c5tb00934k] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Articular cartilage has a limited healing capacity that complicates the treatment of joint injuries and osteoarthritis. Newer repair strategies have focused on the use of cells and biomaterials to promote cartilage regeneration. In the present study, we developed and characterized bioinspired materials designed to mimic the composition of the cartilage extracellular matrix. Chondroitin sulfate (CS) and chitosan (CH) were used to form physically cross-linked macromolecular polyelectrolyte complexes (PEC) without the use of additional crosslinkers. A single-step water-in-oil emulsification process was used to either directly embed mesenchymal stem cells (MSC) in PEC particles created with a various concentrations of CS and CH, or to co-embed MSC with PEC in agarose-based microbeads. Direct embedding of MSC in PEC resulted in high cell viability but irregular and large particles. Co-embedding of PEC particles with MSC in agarose (Ag) resulted in uniform microbeads 80-90 μm in diameter that maintained high cell viability over three weeks in culture. Increased serum content resulted in more uniform PEC distribution within the microbead matrix, and both high and low CS:CH ratios resulted in more homogeneous microbeads than 1:1 formulations. Under chondrogenic conditions, expression of sulfated GAG and collagen type II was increased in 10:1 CS:CH PEC-Ag microbeads compared to pure Ag beads, indicating a chondrogenic influence of the PEC component. Such PEC-Ag microbeads may have utility in the directed differentiation and delivery of progenitor cell populations for cartilage repair.
Collapse
Affiliation(s)
- Ethan Lh Daley
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Rhima M Coleman
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jan P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
33
|
Uncultured bone marrow mononuclear cells delay the dedifferentiation of unexpanded chondrocytes in pellet culture. Cell Tissue Res 2015; 361:811-21. [DOI: 10.1007/s00441-015-2156-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 02/13/2015] [Indexed: 02/08/2023]
|
34
|
Wei B, Yao Q, Guo Y, Mao F, Liu S, Xu Y, Wang L. Three-dimensional polycaprolactone-hydroxyapatite scaffolds combined with bone marrow cells for cartilage tissue engineering. J Biomater Appl 2015; 30:160-70. [PMID: 25766036 DOI: 10.1177/0885328215575762] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The goal of this study was to investigate the chondrogenic potential of three-dimensional polycaprolactone-hydroxyapatite (PCL-HA) scaffolds loaded with bone marrow cells in vitro and the effect of PCL-HA scaffolds on osteochondral repair in vivo. Here, bone marrow was added to the prepared PCL-HA scaffolds and cultured in chondrogenic medium for 10 weeks. Osteochondral defects were created in the trochlear groove of 29 knees in 17 New Zealand white rabbits, which were then divided into four groups that underwent: implantation of PCL-HA scaffolds (left knee, n = 17; Group 1), microfracture (right knee, n = 6; Group 2), autologous osteochondral transplantation (right knee, n = 6; Group 3), and no treatment (right knee, n = 5; Control). Extracellular matrix produced by bone marrow cells covered the surface and filled the pores of PCL-HA scaffolds after 10 weeks in culture. Moreover, many cell-laden cartilage lacunae were observed, and cartilage matrix was concentrated in the PCL-HA scaffolds. After a 12-week repair period, Group 1 showed excellent vertical and lateral integration with host bone, but incomplete cartilage regeneration and matrix accumulation. An uneven surface of regenerated cartilage and reduced distribution of cartilage matrix were observed in Group 2. In addition, abnormal bone growth and unstable integration between repaired and host tissues were detected. For Group 3, the integration between transplanted and host cartilage was interrupted. Our findings indicate that the PCL-HA scaffolds loaded with bone marrow cells improved chondrogenesis in vitro and implantation of PCL-HA scaffolds for osteochondral repairenhanced integration with host bone. However, cartilage regeneration remained unsatisfactory. The addition of trophic factors or the use of precultured cell-PCL-HA constructs for accelerated osteochondral repair requires further investigation.
Collapse
Affiliation(s)
- Bo Wei
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Qingqiang Yao
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yang Guo
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Fengyong Mao
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Shuai Liu
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yan Xu
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Liming Wang
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, China Cartilage Regeneration Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China China-Korea United Cell Therapy Center, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
35
|
Taylor DL, Thevarajah JJ, Narayan DK, Murphy P, Mangala MM, Lim S, Wuhrer R, Lefay C, O’Connor MD, Gaborieau M, Castignolles P. Real-time monitoring of peptide grafting onto chitosan films using capillary electrophoresis. Anal Bioanal Chem 2015; 407:2543-55. [DOI: 10.1007/s00216-015-8483-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 12/22/2014] [Accepted: 01/12/2015] [Indexed: 01/17/2023]
|
36
|
Peterson AW, Caldwell DJ, Rioja AY, Rao RR, Putnam AJ, Stegemann JP. Vasculogenesis and Angiogenesis in Modular Collagen-Fibrin Microtissues. Biomater Sci 2014; 2:1497-1508. [PMID: 25177487 PMCID: PMC4145346 DOI: 10.1039/c4bm00141a] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The process of new blood vessel formation is critical in tissue development, remodeling and regeneration. Modular tissue engineering approaches have been developed to enable the bottom-up assembly of more complex tissues, including vascular networks. In this study, collagen-fibrin composite microbeads (100-300 μm in diameter) were fabricated using a water-in-oil emulsion technique. Human endothelial cells and human fibroblasts were embedded directly in the microbead matrix at the time of fabrication. Microbead populations were characterized and cultured for 14 days either as free-floating populations or embedded in a surrounding fibrin gel. The collagen-fibrin matrix efficiently entrapped cells and supported their viability and spreading. By 7 days in culture, endothelial cell networks were evident within microbeads, and these structures became more prominent by day 14. Fibroblasts co-localized with endothelial cells, suggesting a pericyte-like function, and laminin deposition indicated maturation of the vessel networks over time. Microbeads embedded in a fibrin gel immediately after fabrication showed the emergence of cells and the coalescence of vessel structures in the surrounding matrix by day 7. By day 14, inosculation of neighboring cords and prominent vessel structures were observed. Microbeads pre-cultured for 7 days prior to embedding in fibrin gave rise to vessel networks that emanated radially from the microbead by day 7, and developed into connected networks by day 14. Lumen formation in endothelial cell networks was confirmed using confocal sectioning. These data show that collagen-fibrin composite microbeads support vascular network formation. Microbeads embedded directly after fabrication emulated the process of vasculogenesis, while the branching and joining of vessels from pre-cultured microbeads resembled angiogenesis. This modular microtissue system has utility in studying the processes involved in new vessel formation, and may be developed into a therapy for the treatment of ischemic conditions.
Collapse
Affiliation(s)
- A W Peterson
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - D J Caldwell
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - A Y Rioja
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - R R Rao
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - A J Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - J P Stegemann
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
37
|
Thompson EM, Matsiko A, Farrell E, Kelly DJ, O'Brien FJ. Recapitulating endochondral ossification: a promising route toin vivobone regeneration. J Tissue Eng Regen Med 2014; 9:889-902. [DOI: 10.1002/term.1918] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 02/14/2014] [Accepted: 04/24/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Emmet M. Thompson
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
| | - Amos Matsiko
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
| | - Eric Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC; University Medical Centre Rotterdam; The Netherlands
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of Engineering; Trinity College Dublin; Ireland
| | - Fergal J. O'Brien
- Tissue Engineering Research Group, Department of Anatomy; Royal College of Surgeons in Ireland; Dublin Ireland
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute; Trinity College Dublin; Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre; Dublin Ireland
| |
Collapse
|
38
|
Abstract
Bone defects requiring grafts to promote healing are frequently occurring and costly problems in health care. Chitosan, a biodegradable, naturally occurring polymer, has drawn considerable attention in recent years as scaffolding material in tissue engineering and regenerative medicine. Chitosan is especially attractive as a bone scaffold material because it supports the attachment and proliferation of osteoblast cells as well as formation of mineralized bone matrix. In this review, we discuss the fundamentals of bone tissue engineering and the unique properties of chitosan as a scaffolding material to treat bone defects for hard tissue regeneration. We present the common methods for fabrication and characterization of chitosan scaffolds, and discuss the influence of material preparation and addition of polymeric or ceramic components or biomolecules on chitosan scaffold properties such as mechanical strength, structural integrity, and functional bone regeneration. Finally, we highlight recent advances in development of chitosan-based scaffolds with enhanced bone regeneration capability.
Collapse
Affiliation(s)
- Sheeny Lan Levengood
- Department of Materials Science & Engineering, University of Washington, Seattle, WA 98195 USA
| | - Miqin Zhang
- Department of Materials Science & Engineering, University of Washington, Seattle, WA 98195 USA
| |
Collapse
|
39
|
Song F, Tang J, Geng R, Hu H, Zhu C, Cui W, Fan W. Comparison of the efficacy of bone marrow mononuclear cells and bone mesenchymal stem cells in the treatment of osteoarthritis in a sheep model. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:1415-1426. [PMID: 24817937 PMCID: PMC4014221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 02/04/2014] [Indexed: 06/03/2023]
Abstract
OBJECTIVE To evaluate the therapeutic efficacy of uncultured bone marrow mononuclear cells (BMMCs) and bone mesenchymal stem cells in an osteoarthritis (OA) model of sheep. METHODS Induction of sheep OA was performed surgically through anterior cruciate ligament transection and medial meniscectomy. After 12 weeks, concentrated BMMCs obtained from autologous bone marrow harvested from anterior iliac crest or a single dose of 10 million autologous bone mesenchymal stem cells (BMSCs) suspended in phosphate-buffered saline (PBS) was delivered to the injured knee via direct intra-articular injection. Animals of the PBS group received vehicle alone. The contra-lateral joints were selected randomly as the control group. Knees of the four groups were compared macroscopically and histologically, and glycosaminoglycan (GAG) contents normalized to cartilage wet weight were measured at lesions of cartilage from medial condyle of the femur head. Gene expression levels of type II collagen (Col2A1), Aggrecan and matrix metalloproteinase-13 (MMP-13) in cartilage were measured based on RT-PCR and prostaglandin E2 (PGE2), Tumor Necrosis Factor-α (TNF-α) and Transforming Growth Factor beta (TGF-β) concentrations in synovial fluid were determined with ELISA assays at 8 weeks after injection. RESULTS At 8 weeks post cell transplantation, partial cartilage repair was observed in the cell therapy, but not the PBS group (P<0.05). The BMSCs group showed higher regeneration of cartilage and lower proteoglycan loss than the BMMCs group (P<0.05). Concentrated BMMCs injection led to a weaker treatment effect, but also inhibited PGE2, TNF-α and TGF-β levels in synovial fluid and promoted higher levels of Aggrecan and Col2A1 and downregulation of MMP-13 in sheep chondrocytes in a similar manner to BMSCs, compared with the PBS group. CONCLUSIONS Bone marrow cells showed therapeutic efficacy in a sheep model of OA. Despite similar therapeutic potential, the easier and faster process of collection and isolation of BMMCs supports their utility as an effective alternative for OA treatment in the clinic.
Collapse
Affiliation(s)
- Fanglong Song
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing, 210029, China
| | - Jilei Tang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing, 210029, China
| | - Rui Geng
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing, 210029, China
| | - Hansheng Hu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing, 210029, China
| | - Chunhui Zhu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing, 210029, China
| | - Weiding Cui
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing, 210029, China
| | - Weimin Fan
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University Nanjing, 210029, China
| |
Collapse
|
40
|
Boyette LB, Creasey OA, Guzik L, Lozito T, Tuan RS. Human bone marrow-derived mesenchymal stem cells display enhanced clonogenicity but impaired differentiation with hypoxic preconditioning. Stem Cells Transl Med 2014; 3:241-54. [PMID: 24436440 DOI: 10.5966/sctm.2013-0079] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Stem cells are promising candidate cells for regenerative applications because they possess high proliferative capacity and the potential to differentiate into other cell types. Mesenchymal stem cells (MSCs) are easily sourced but do not retain their proliferative and multilineage differentiative capabilities after prolonged ex vivo propagation. We investigated the use of hypoxia as a preconditioning agent and in differentiating cultures to enhance MSC function. Culture in 5% ambient O(2) consistently enhanced clonogenic potential of primary MSCs from all donors tested. We determined that enhanced clonogenicity was attributable to increased proliferation, increased vascular endothelial growth factor secretion, and increased matrix turnover. Hypoxia did not impact the incidence of cell death. Application of hypoxia to osteogenic cultures resulted in enhanced total mineral deposition, although this effect was detected only in MSCs preconditioned in normoxic conditions. Osteogenesis-associated genes were upregulated in hypoxia, and alkaline phosphatase activity was enhanced. Adipogenic differentiation was inhibited by exposure to hypoxia during differentiation. Chondrogenesis in three-dimensional pellet cultures was inhibited by preconditioning with hypoxia. However, in cultures expanded under normoxia, hypoxia applied during subsequent pellet culture enhanced chondrogenesis. Whereas hypoxic preconditioning appears to be an excellent way to expand a highly clonogenic progenitor pool, our findings suggest that it may blunt the differentiation potential of MSCs, compromising their utility for regenerative tissue engineering. Exposure to hypoxia during differentiation (post-normoxic expansion), however, appears to result in a greater quantity of functional osteoblasts and chondrocytes and ultimately a larger quantity of high-quality differentiated tissue.
Collapse
Affiliation(s)
- Lisa B Boyette
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, USA; Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, McGowan Institute for Regenerative Medicine, and Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | | | | |
Collapse
|