1
|
Choi S, Choi H, Chung JW, Kim SH. Injectable Endoplasmin-Loaded Lipid Nanoparticles-Hydrogel Composite for Cartilage Regeneration. Tissue Eng Regen Med 2025:10.1007/s13770-024-00698-2. [PMID: 39992620 DOI: 10.1007/s13770-024-00698-2] [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: 10/21/2024] [Revised: 12/12/2024] [Accepted: 12/22/2024] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Endoplasmin (ENPL), a heat shock protein 90 family member, promotes chondrogenic differentiation of stem cells by inhibiting ERK1/2 phosphorylation and inducing endoplasmic reticulum stress. However, its large size limits cellular uptake and therapeutic potential. To overcome this challenge, a cationic lipid nanoparticle (C_LNP) system was designed to deliver ENPL intracellularly, enhancing its effects on human tonsil-derived mesenchymal stem cells (hTMSCs). METHODS ENPL-loaded cationic lipid nanoparticles (ENPL_C_LNP) were synthesized to facilitate intracellular ENPL delivery. The delivery efficiency and cytotoxicity were assessed in vitro using hTMSCs. Additionally, ENPL_C_LNPs were incorporated into a hyaluronic acid and chondroitin sulfate-based injectable hydrogel and tested for chondrogenic differentiation potential in a mouse subcutaneous model. RESULTS ENPL_C_LNP achieved over 80% intracellular protein delivery efficiency with no cytotoxic effects. Co-cultured hTMSCs exhibited increased glycosaminoglycans (GAGs) and collagen expression over 21 days. In vivo, the hydrogel-embedded ENPL_C_LNP system enabled stable cartilage differentiation, evidenced by abundant cartilage-specific lacuna structures in regenerated tissue. CONCLUSION Combining ENPL_C_LNP with an injectable hydrogel scaffold supports chondrogenic differentiation and cartilage regeneration, offering a promising strategy for cartilage tissue engineering.
Collapse
Affiliation(s)
- Sumi Choi
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan, 49315, Republic of Korea
| | - Hyeongrok Choi
- Department of Biomedical Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Jin Woong Chung
- Department of Biomedical Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan, 49315, Republic of Korea.
| |
Collapse
|
2
|
Liao Z. Clinical research progress of umbilical cord blood mesenchymal stem cells in Knee articular cartilage repair: A review. Medicine (Baltimore) 2025; 104:e41402. [PMID: 39928895 PMCID: PMC11813059 DOI: 10.1097/md.0000000000041402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/27/2024] [Accepted: 01/13/2025] [Indexed: 02/12/2025] Open
Abstract
Umbilical cord blood mesenchymal stem cells (UCB-MSCs) are a type of adult stem cell with multipotent differentiation potential and immunoregulatory functions, primarily found in neonatal cord blood. Due to their noninvasive collection method, abundance, and ease of preservation, UCB-MSCs represent a promising biological material. This review examines the clinical research on UCB-MSCs in knee articular cartilage repair, highlighting their regenerative potential for treating knee joint cartilage defects. Our aim is to provide insights into current applications and propose directions for future research, focusing on optimizing clinical use and enhancing patient outcomes.
Collapse
Affiliation(s)
- ZhongKai Liao
- The Second Affiliated Hospital of Hainan Medical College, Master of Medicine, Haikou, China
| |
Collapse
|
3
|
Singer J, Knezic N, Gohring G, Fite O, Christiansen J, Huard J. Synovial mesenchymal stem cells. ORTHOBIOLOGICS 2025:141-154. [DOI: 10.1016/b978-0-12-822902-6.00005-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
4
|
Rauer SB, Stüwe L, Steinbeck L, de Toledo MAS, Fischer G, Wennemaring S, Marschick J, Koschmieder S, Wessling M, Linkhorst J. Cell Adhesion and Local Cytokine Control on Protein-Functionalized PNIPAM-co-AAc Hydrogel Microcarriers. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2404183. [PMID: 39535368 PMCID: PMC11735893 DOI: 10.1002/smll.202404183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Achieving adequate cell densities remains a major challenge in establishing economic biotechnological and biomedical processes. A possible remedy is microcarrier-based cultivation in stirred-tank bioreactors (STBR), which offers a high surface-to-volume ratio, appropriate process control, and scalability. However, despite their potential, commercial microcarriers are currently limited to material systems featuring unnatural mechanical properties and low adaptability. Because matrix stiffness and ligand presentation impact phenotypical attributes, differentiation potential, and genetic stability, biotechnological processes can significantly benefit from microcarrier systems tailorable toward cell-type specific requirements. This study introduces hydrogel particles co-polymerized from poly(N-isopropylacrylamide) (PNIPAM) and acrylic acid (AAc) as a platform technology for cell expansion. The resulting microcarriers exhibit an adjustable extracellular matrix-like softness, an adaptable gel charge, and functional carboxyl groups, allowing electrostatic and covalent coupling of cell adhesive and cell fate-modulating proteins. These features enable the attachment and growth of L929 mouse fibroblast cells in static microtiter plates and dynamic STBR cultivations while also providing vital growth factors, such as interleukin-3, to myeloblast-like 32D cells over 20 days of cultivation. The study explores the effects of different educt compositions on cell-particle interactions and reveals that PNIPAM-co-AAc microcarriers can provide both covalently coupled and diffusively released cytokine to adjacent cells.
Collapse
Affiliation(s)
- Sebastian Bernhard Rauer
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
- DWI ‐ Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052074AachenGermany
| | - Lucas Stüwe
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Lea Steinbeck
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Marcelo Augusto Szymanski de Toledo
- Department of HematologyOncology, Hemostaseology, and Stem Cell TransplantationFaculty of MedicineRWTH Aachen University52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD)52074AachenGermany
| | - Gereon Fischer
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Simon Wennemaring
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Jonas Marschick
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
| | - Steffen Koschmieder
- Department of HematologyOncology, Hemostaseology, and Stem Cell TransplantationFaculty of MedicineRWTH Aachen University52074AachenGermany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD)52074AachenGermany
| | - Matthias Wessling
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
- DWI ‐ Leibniz Institute for Interactive MaterialsForckenbeckstr. 5052074AachenGermany
| | - John Linkhorst
- Chemical Process EngineeringRWTH Aachen UniversityForckenbeckstr. 5152074AachenGermany
- Process Engineering of Electrochemical SystemsDepartment of Mechanical EngineeringTechnical University of DarmstadtOtto‐Berndt‐Str. 264287DarmstadtGermany
| |
Collapse
|
5
|
Yi J, Byun Y, Kang SS, Shim KM, Jang K, Lee JY. Enhanced Chondrogenic Differentiation of Electrically Primed Human Mesenchymal Stem Cells for the Regeneration of Osteochondral Defects. Biomater Res 2024; 28:0109. [PMID: 39697183 PMCID: PMC11654951 DOI: 10.34133/bmr.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 10/12/2024] [Accepted: 10/26/2024] [Indexed: 12/20/2024] Open
Abstract
Background: Mesenchymal stem cells (MSCs) offer a promising avenue for cartilage regeneration; however, their therapeutic efficacy requires substantial improvement. Cell priming using electrical stimulation (ES) is a promising approach to augmenting the therapeutic potential of MSCs and has shown potential for various regenerative applications. This study aimed to promote the ES-mediated chondrogenic differentiation of human MSCs and facilitate the repair of injured articular cartilage. Methods: MSCs were subjected to ES under various conditions (e.g., voltage, frequency, and number of repetitions) to enhance their capability of chondrogenesis and cartilage regeneration. Chondrogenic differentiation of electrically primed MSCs (epMSCs) was assessed based on gene expression and sulfated glycosaminoglycan production, and epMSCs with hyaluronic acid were transplanted into a rat osteochondral defect model. Transcriptomic analysis was performed to determine changes in gene expression by ES. Results: epMSCs exhibited significantly increased chondrogenic gene expression and sulfated glycosaminoglycan production compared with those in unstimulated controls. Macroscopic and histological results showed that in vivo epMSC transplantation considerably enhanced cartilage regeneration. Furthermore, ES markedly altered the expression of numerous genes of MSCs, including those associated with the extracellular matrix, the Wnt signaling pathway, and cartilage development. Conclusion: ES can effectively prime MSCs to improve articular cartilage repair, offering a promising strategy for enhancing the efficacy of various MSC-based therapies.
Collapse
Affiliation(s)
- Jongdarm Yi
- School of Materials Science and Engineering,
Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| | - Yujin Byun
- Department of Veterinary Surgery, College of Veterinary Medicine and BK21 FOUR Program,
Chonnam National University, Gwangju 61186, Republic of Korea
- Biomaterial R&BD Center,
Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seong Soo Kang
- Department of Veterinary Surgery, College of Veterinary Medicine and BK21 FOUR Program,
Chonnam National University, Gwangju 61186, Republic of Korea
- Biomaterial R&BD Center,
Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kyung Mi Shim
- Department of Veterinary Surgery, College of Veterinary Medicine and BK21 FOUR Program,
Chonnam National University, Gwangju 61186, Republic of Korea
- Biomaterial R&BD Center,
Chonnam National University, Gwangju 61186, Republic of Korea
| | - Kwangsik Jang
- Department of Veterinary Surgery, College of Veterinary Medicine and BK21 FOUR Program,
Chonnam National University, Gwangju 61186, Republic of Korea
- Biomaterial R&BD Center,
Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering,
Gwangju Institute of Science and Technology, Gwangju 61005, Republic of Korea
| |
Collapse
|
6
|
Jakutowicz T, Wasyłeczko M, Płończak M, Wojciechowski C, Chwojnowski A, Czubak J. Comparative Study of Autogenic and Allogenic Chondrocyte Transplants on Polyethersulfone Scaffolds for Cartilage Regeneration. Int J Mol Sci 2024; 25:9075. [PMID: 39201763 PMCID: PMC11354243 DOI: 10.3390/ijms25169075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/10/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
The aim of this study was to evaluate the chondrogenic potential of chondrocyte transplants cultured in vitro on polyethersulfone (PES) membranes. Forty-eight rabbits (96 knee joints) were used in the project. The synthetic, macro-porous PES membranes were used as scaffolds. Fragments of articular cartilage were harvested from non-weight-bearing areas of the joints of the animals. Chondrocytes were isolated and then cultivated on PES scaffolds for 3 weeks. The animals were divided into four groups. All the lesions in the articular cartilage were full thickness defects. In Group I, autogenic chondrocytes on PES membranes were transplanted into the defect area; in Group II, allogenic chondrocytes on PES membranes were transplanted into the defect area; in Group III, pure PES membranes were transplanted into the defect area; and in Group IV, lesions were left untreated. Half of the animals from each group were terminated after 8 weeks, and the remaining half were terminated 12 weeks postoperatively. The samples underwent macroscopic evaluation using the Brittberg scale and microscopic evaluation using the O'Driscoll scale. The best regeneration was observed in Groups II and I. In Group I, the results were achieved with two surgeries, while in Group II, only one operation was needed. This indicates that allogenic chondrocytes do not require two surgeries, highlighting the importance of further in vivo studies to better understand this advantage. The success of the study and the desired properties of PES scaffolds are attributed mainly to the presence of sulfonic groups in the structure of the material. These groups, similar to chondroitin sulfate, which naturally occurs in hyaline cartilage, likely enable mutual affinity between the scaffold and cells and promote scaffold colonization by the cells.
Collapse
Affiliation(s)
- Tomasz Jakutowicz
- Paediatric Orthopaedics and Traumatology Department, Children’s Hospital, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Monika Wasyłeczko
- Nałęcz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Maciej Płończak
- Department of Orthopedics, Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland
- Gruca Teaching Hospital, 05-400 Otwock, Poland
- Mazovia Regional Hospital in Siedlce, 08-110 Siedlce, Poland
| | - Cezary Wojciechowski
- Nałęcz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Andrzej Chwojnowski
- Nałęcz Institute of Biocybernetics and Biomedical Engineering Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Jarosław Czubak
- Department of Orthopedics, Medical Centre of Postgraduate Education, 01-813 Warsaw, Poland
- Gruca Teaching Hospital, 05-400 Otwock, Poland
| |
Collapse
|
7
|
Hwang S, Eom YW, Kang SH, Baik SK, Kim MY. IFN-β Overexpressing Adipose-Derived Mesenchymal Stem Cells Mitigate Alcohol-Induced Liver Damage and Gut Permeability. Int J Mol Sci 2024; 25:8509. [PMID: 39126076 PMCID: PMC11313321 DOI: 10.3390/ijms25158509] [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: 06/17/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Alcoholic liver disease (ALD) is a form of hepatic inflammation. ALD is mediated by gut leakiness. This study evaluates the anti-inflammatory effects of ASCs overexpressing interferon-beta (ASC-IFN-β) on binge alcohol-induced liver injury and intestinal permeability. In vitro, ASCs were transfected with a non-viral vector carrying the human IFN-β gene, which promoted hepatocyte growth factor (HGF) secretion in the cells. To assess the potential effects of ASC-IFN-β, C57BL/6 mice were treated with three oral doses of binge alcohol and were administered intraperitoneal injections of ASC-IFN-β. Mice treated with binge alcohol and administered ASC-IFN-β showed reduced liver injury and inflammation compared to those administered a control ASC. Analysis of intestinal tissue from ethanol-treated mice administered ASC-IFN-β also indicated decreased inflammation. Additionally, fecal albumin, blood endotoxin, and bacterial colony levels were reduced, indicating less gut leakiness in the binge alcohol-exposed mice. Treatment with HGF, but not IFN-β or TRAIL, mitigated the ethanol-induced down-regulation of cell death and permeability in Caco-2 cells. These results demonstrate that ASCs transfected with a non-viral vector to induce IFN-β overexpression have protective effects against binge alcohol-mediated liver injury and gut leakiness via HGF.
Collapse
Affiliation(s)
- Soonjae Hwang
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea;
- Regeneration Medicine Research Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Republic of Korea; (Y.W.E.); (S.K.B.)
- Cell Therapy and Tissue Engineering Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Republic of Korea
| | - Young Woo Eom
- Regeneration Medicine Research Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Republic of Korea; (Y.W.E.); (S.K.B.)
- Cell Therapy and Tissue Engineering Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Republic of Korea
| | - Seong Hee Kang
- Department of Internal Medicine, College of Medicine, Korea University, Seoul 02841, Republic of Korea;
| | - Soon Koo Baik
- Regeneration Medicine Research Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Republic of Korea; (Y.W.E.); (S.K.B.)
- Department of Internal Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Republic of Korea
| | - Moon Young Kim
- Regeneration Medicine Research Center, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Republic of Korea; (Y.W.E.); (S.K.B.)
- Department of Internal Medicine, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Republic of Korea
| |
Collapse
|
8
|
Reis IL, Lopes B, Sousa P, Sousa AC, Rêma A, Caseiro AR, Briote I, Rocha AM, Pereira JP, Mendonça CM, Santos JM, Lamas L, Atayde LM, Alvites RD, Maurício AC. Case report: Equine metacarpophalangeal joint partial and full thickness defects treated with allogenic equine synovial membrane mesenchymal stem/stromal cell combined with umbilical cord mesenchymal stem/stromal cell conditioned medium. Front Vet Sci 2024; 11:1403174. [PMID: 38840629 PMCID: PMC11150641 DOI: 10.3389/fvets.2024.1403174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/01/2024] [Indexed: 06/07/2024] Open
Abstract
Here, we describe a case of a 5-year-old show-jumping stallion presented with severe lameness, swelling, and pain on palpation of the left metacarpophalangeal joint (MCj). Diagnostic imaging revealed full and partial-thickness articular defects over the lateral condyle of the third metacarpus (MC3) and the dorsolateral aspect of the first phalanx (P1). After the lesion's arthroscopic curettage, the patient was subjected to an innovative regenerative treatment consisting of two intra-articular injections of equine synovial membrane mesenchymal stem/stromal cells (eSM-MSCs) combined with umbilical cord mesenchymal stem/stromal cells conditioned medium (UC-MSC CM), 15 days apart. A 12-week rehabilitation program was accomplished, and lameness, pain, and joint effusion were remarkably reduced; however, magnetic resonance imaging (MRI) and computed tomography (CT) scan presented incomplete healing of the MC3's lesion, prompting a second round of treatment. Subsequently, the horse achieved clinical soundness and returned to a higher level of athletic performance, and imaging exams revealed the absence of lesions at P1, fulfillment of the osteochondral lesion, and cartilage-like tissue formation at MC3's lesion site. The positive outcomes suggest the effectiveness of this combination for treating full and partial cartilage defects in horses. Multipotent mesenchymal stem/stromal cells (MSCs) and their bioactive factors compose a novel therapeutic approach for tissue regeneration and organ function restoration with anti-inflammatory and pro-regenerative impact through paracrine mechanisms.
Collapse
Affiliation(s)
- I. L. Reis
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Avenida Central de Gandra, Gandra, Portugal
| | - B. Lopes
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - P. Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - A. C. Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - A. Rêma
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - A. R. Caseiro
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Departamento de Ciências Veterinárias, Escola Universitária Vasco da Gama (EUVG), Coimbra, Portugal
- Centro de Investigação Vasco da Gama (CIVG), Escola Universitária Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, Coimbra, Portugal
| | - I. Briote
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Vairão, Portugal
| | - A. M. Rocha
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Vairão, Portugal
| | - J. P. Pereira
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Vairão, Portugal
| | - C. M. Mendonça
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Vairão, Portugal
| | - J. M. Santos
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
| | - L. Lamas
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisboa, Portugal
- CIISA—Centro Interdisciplinar-Investigação em Saúde Animal, Faculdade de Medicina Veterinária, Av. Universidade Técnica de Lisboa, Lisboa, Portugal
| | - L. M. Atayde
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Vairão, Portugal
| | - R. D. Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Avenida Central de Gandra, Gandra, Portugal
| | - A. C. Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Porto, Portugal
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Vairão, Portugal
| |
Collapse
|
9
|
Reis IL, Lopes B, Sousa P, Sousa AC, Caseiro AR, Mendonça CM, Santos JM, Atayde LM, Alvites RD, Maurício AC. Equine Musculoskeletal Pathologies: Clinical Approaches and Therapeutical Perspectives-A Review. Vet Sci 2024; 11:190. [PMID: 38787162 PMCID: PMC11126110 DOI: 10.3390/vetsci11050190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 04/12/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Musculoskeletal injuries such as equine osteoarthritis, osteoarticular defects, tendonitis/desmitis, and muscular disorders are prevalent among sport horses, with a fair prognosis for returning to exercise or previous performance levels. The field of equine medicine has witnessed rapid and fruitful development, resulting in a diverse range of therapeutic options for musculoskeletal problems. Staying abreast of these advancements can be challenging, prompting the need for a comprehensive review of commonly used and recent treatments. The aim is to compile current therapeutic options for managing these injuries, spanning from simple to complex physiotherapy techniques, conservative treatments including steroidal and non-steroidal anti-inflammatory drugs, hyaluronic acid, polysulfated glycosaminoglycans, pentosan polysulfate, and polyacrylamides, to promising regenerative therapies such as hemoderivatives and stem cell-based therapies. Each therapeutic modality is scrutinized for its benefits, limitations, and potential synergistic actions to facilitate their most effective application for the intended healing/regeneration of the injured tissue/organ and subsequent patient recovery. While stem cell-based therapies have emerged as particularly promising for equine musculoskeletal injuries, a multidisciplinary approach is underscored throughout the discussion, emphasizing the importance of considering various therapeutic modalities in tandem.
Collapse
Affiliation(s)
- Inês L. Reis
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Departamento de Ciências Veterinárias, Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Bruna Lopes
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Patrícia Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Ana C. Sousa
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Ana R. Caseiro
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Veterinary Sciences Department, University School Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, Lordemão, 3020-210 Coimbra, Portugal
- Vasco da Gama Research Center (CIVG), University School Vasco da Gama (EUVG), Avenida José R. Sousa Fernandes, Lordemão, 3020-210 Coimbra, Portugal
| | - Carla M. Mendonça
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| | - Jorge M. Santos
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
| | - Luís M. Atayde
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| | - Rui D. Alvites
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Departamento de Ciências Veterinárias, Cooperativa de Ensino Superior Politécnico e Universitário (CESPU), Instituto Universitário de Ciências da Saúde (IUCS), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
| | - Ana C. Maurício
- Departamento de Clínicas Veterinárias, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Rua de Jorge Viterbo Ferreira, n° 228, 4050-313 Porto, Portugal; (I.L.R.); (B.L.); (P.S.); (A.C.S.); (C.M.M.); (J.M.S.); (L.M.A.); (R.D.A.)
- Centro de Estudos de Ciência Animal (CECA), Instituto de Ciências, Tecnologias e Agroambiente da Universidade do Porto (ICETA), Rua D. Manuel II, Apartado 55142, 4051-401 Porto, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), 1300-477 Lisboa, Portugal
- Campus Agrário de Vairão, Centro Clínico de Equinos de Vairão (CCEV), Rua da Braziela n° 100, 4485-144 Vairão, Portugal
| |
Collapse
|
10
|
Kaonis S, Aboellail Z, Forman J, Ghosh S. High-Throughput Multiparametric Quantification of Mechanics Driven Heterogeneity in Mesenchymal Stromal Cell Population. Adv Biol (Weinh) 2024; 8:e2300318. [PMID: 37840408 DOI: 10.1002/adbi.202300318] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Indexed: 10/17/2023]
Abstract
Mesenchymal stromal or stem cells (MSCs) are one of the most promising candidates for a myriad of cell therapy applications. Despite showing promise in numerous preclinical and clinical studies, MSC-based therapy is not yet a reality for regenerative medicine due to its suboptimal outcome at the clinical endpoint. The mechanical environment is a critical determinant of MSC gene expression and function. This study reports that MSC population becomes phenotypically heterogenous and commits to an unwanted osteoprogenitor pathway when it experiences an abnormal mechanically stiff environment, compared to its native softer environment. A method is developed to measure the heterogeneity using nuclear shape, chromatin state, and CD73 marker. Heterogeneity is shown to be associated with a larger spread in the nuclear shape parameters and a smaller spread in the chromatin openness. Subsequently, intervention strategies are investigated to create a more homogeneous MSC population. Culturing MSCs on soft surfaces or inhibiting actomyosin on stiff surfaces can make them more homogeneous, while inhibiting YAP, Runx2, and actin polymerization helps maintain but does not fully homogenize them. This study offers insights for cell and tissue engineers, aiding in the design of optimal conditions and materials for MSC culture, ultimately enhancing their therapeutic potential.
Collapse
Affiliation(s)
- Samantha Kaonis
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
| | - Zack Aboellail
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
| | - Jack Forman
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Chemical and Biological Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
| | - Soham Ghosh
- School of Biomedical Engineering, Colorado State University, 700 Meridian Ave, Fort Collins, CO, 80523, USA
- Translational Medicine Institute, Colorado State University, 2350 Gillette Dr, Fort Collins, CO, 80523, USA
- Department of Mechanical Engineering, Colorado State University, 400 Isotope Dr, Fort Collins, CO, 80521, USA
- Cell and Molecular Biology, Colorado State University, 1050 Libbie Coy Way, Fort Collins, CO, 80524, USA
| |
Collapse
|
11
|
Johnbosco C, Karbaat L, Korthagen NM, Warmink K, Koerselman M, Coeleveld K, Becker M, van Loo B, Zoetebier B, Both S, Weinans H, Karperien M, Leijten J. Microencapsulated stem cells reduce cartilage damage in a material dependent manner following minimally invasive intra-articular injection in an OA rat model. Mater Today Bio 2023; 22:100791. [PMID: 37731960 PMCID: PMC10507156 DOI: 10.1016/j.mtbio.2023.100791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/05/2023] [Accepted: 09/04/2023] [Indexed: 09/22/2023] Open
Abstract
Osteoarthritis (OA) is a degenerative disease of the joints for which no curative treatment exists. Intra-articular injection of stem cells is explored as a regenerative approach, but rapid clearance of cells from the injection site limits the therapeutic outcome. Microencapsulation of mesenchymal stem cells (MSCs) can extend the retention time of MSCs, but the outcomes of the few studies currently performed are conflicting. We hypothesize that the composition of the micromaterial's shell plays a deciding factor in the treatment outcome of intra-articular MSC injection. To this end, we microencapsulate MSCs using droplet microfluidic generators in flow-focus mode using various polymers and polymer concentrations. We demonstrate that polymer composition and concentration potently alter the metabolic activity as well as the secretome of MSCs. Moreover, while microencapsulation consistently prolongs the retention time of MSC injected in rat joints, distinct biodistribution within the joint is demonstrated for the various microgel formulations. Furthermore, intra-articular injections of pristine and microencapsulated MSC in OA rat joints show a strong material-dependent effect on the reduction of cartilage degradation and matrix loss. Collectively, this study highlights that micromaterial composition and concentration are key deciding factors for the therapeutic outcome of intra-articular injections of microencapsulated stem cells to treat degenerative joint diseases.
Collapse
Affiliation(s)
- Castro Johnbosco
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Lisanne Karbaat
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Nicoline M. Korthagen
- Faculty of Veterinary Sciences Department of equine sciences, University of Utrecht, the Netherlands
- Department of Orthopaedics, University Medical Centre Utrecht, the Netherlands
| | - Kelly Warmink
- Department of Orthopaedics, University Medical Centre Utrecht, the Netherlands
| | - Michelle Koerselman
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Katja Coeleveld
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, the Netherlands
| | - Malin Becker
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Bas van Loo
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Bram Zoetebier
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Sanne Both
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Harrie Weinans
- Department of Orthopaedics, University Medical Centre Utrecht, the Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| | - Jeroen Leijten
- Department of Developmental BioEngineering, TechMed Centre, University of Twente, the Netherlands
| |
Collapse
|
12
|
Martier AT, Maurice YV, Conrad KM, Mauvais-Jarvis F, Mondrinos MJ. Sex-specific actions of estradiol and testosterone on human fibroblast and endothelial cell proliferation, bioenergetics, and vasculogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.23.550236. [PMID: 37546849 PMCID: PMC10402012 DOI: 10.1101/2023.07.23.550236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Progress toward the development of sex-specific tissue engineered systems has been hampered by the lack of research efforts to define the effects of sex-specific hormone concentrations on relevant human cell types. Here, we investigated the effects of defined concentrations of estradiol (E2) and dihydrotestosterone (DHT) on primary human dermal and lung fibroblasts (HDF and HLF), and human umbilical vein endothelial cells (HUVEC) from female (XX) and male (XY) donors in both 2D expansion cultures and 3D stromal vascular tissues. Sex-matched E2 and DHT stimulation in 2D expansion cultures significantly increased the proliferation index, mitochondrial membrane potential, and the expression of genes associated with bioenergetics (Na+/K+ ATPase, somatic cytochrome C) and beneficial stress responses (chaperonin) in all cell types tested. Notably, cross sex hormone stimulation, i.e., DHT treatment of XX cells in the absence of E2 and E2 stimulation of XY cells in the absence of DHT, decreased bioenergetic capacity and inhibited cell proliferation. We used a microengineered 3D vasculogenesis assay to assess hormone effects on tissue scale morphogenesis. E2 increased metrics of vascular network complexity compared to vehicle in XX tissues. Conversely, and in line with results from 2D expansion cultures, E2 potently inhibited vasculogenesis compared to vehicle in XY tissues. DHT did not significantly alter vasculogenesis in XX or XY tissues but increased the number of non-participating endothelial cells in both sexes. This study establishes a scientific rationale and adaptable methods for using sex hormone stimulation to develop sex-specific culture systems.
Collapse
Affiliation(s)
- Ashley T. Martier
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
| | - Yasmin V. Maurice
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
| | - K. Michael Conrad
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
| | - Franck Mauvais-Jarvis
- Tulane Center for Excellence in Sex-based Biology and Medicine, New Orleans, LA, USA
- Section of Endocrinology, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Southeast Louisiana VA Medical Center, New Orleans, LA, USA
| | - Mark J. Mondrinos
- Department of Biomedical Engineering, Tulane University School of Science & Engineering, New Orleans, LA, USA
- Tulane Center for Excellence in Sex-based Biology and Medicine, New Orleans, LA, USA
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
13
|
Liang J, Liu P, Yang X, Liu L, Zhang Y, Wang Q, Zhao H. Biomaterial-based scaffolds in promotion of cartilage regeneration: Recent advances and emerging applications. J Orthop Translat 2023; 41:54-62. [PMID: 37691640 PMCID: PMC10485599 DOI: 10.1016/j.jot.2023.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/07/2023] [Accepted: 08/05/2023] [Indexed: 09/12/2023] Open
Abstract
Osteoarthritis (OA) poses a significant burden for countless individuals, inflicting relentless pain and impairing their quality of life. Although traditional treatments for OA focus on pain management and surgical interventions, they often fall short of addressing the underlying cause of the disease. Fortunately, emerging biomaterial-based scaffolds offer hope for OA therapy, providing immense promise for cartilage regeneration in OA. These innovative scaffolds are ingeniously designed to provide support and mimic the intricate structure of the natural extracellular matrix, thus stimulating the regeneration of damaged cartilage. In this comprehensive review, we summarize and discuss current landscape of biomaterial-based scaffolds for cartilage regeneration in OA. Furthermore, we delve into the diverse range of biomaterials employed in their construction and explore the cutting-edge techniques utilized in their fabrication. By examining both preclinical and clinical studies, we aim to illuminate the remarkable versatility and untapped potential of biomaterial-based scaffolds in the context of OA. Thetranslational potential of this article By thoroughly examining the current state of research and clinical studies, this review provides valuable insights that bridge the gap between scientific knowledge and practical application. This knowledge is crucial for clinicians and researchers who strive to develop innovative treatments that go beyond symptom management and directly target the underlying cause of OA. Through the comprehensive analysis and multidisciplinary approach, the review paves the way for the translation of scientific knowledge into practical applications, ultimately improving the lives of individuals suffering from OA and shaping the future of orthopedic medicine.
Collapse
Affiliation(s)
| | | | - Xinquan Yang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Liu
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qiong Wang
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hongmou Zhao
- Department of Foot and Ankle Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14
|
Płończak M, Wasyłeczko M, Jakutowicz T, Chwojnowski A, Czubak J. Intraarticular Implantation of Autologous Chondrocytes Placed on Collagen or Polyethersulfone Scaffolds: An Experimental Study in Rabbits. Polymers (Basel) 2023; 15:polym15102360. [PMID: 37242936 DOI: 10.3390/polym15102360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Hyaline cartilage has very limited repair capability and cannot be rebuilt predictably using conventional treatments. This study presents Autologous Chondrocyte Implantation (ACI) on two different scaffolds for the treatment of lesions in hyaline cartilage in rabbits. The first one is a commercially available scaffold (Chondro-Gide) made of collagen type I/III and the second one is a polyethersulfone (PES) synthetic membrane, manufactured by phase inversion. The revolutionary idea in the present study is the fact that we used PES membranes, which have unique features and benefits that are desirable for the 3D cultivation of chondrocytes. Sixty-four White New Zealand rabbits were used in this research. Defects penetrating into the subchondral bone were filled with or without the placement of chondrocytes on collagen or PES membranes after two weeks of culture. The expression of the gene encoding type II procollagen, a molecular marker of chondrocytes, was evaluated. Elemental analysis was performed to estimate the weight of tissue grown on the PES membrane. The reparative tissue was analyzed macroscopically and histologically after surgery at 12, 25, and 52 weeks. RT-PCR analysis of the mRNA isolated from cells detached from the polysulphonic membrane revealed the expression of type II procollagen. The elementary analysis of polysulphonic membrane slices after 2 weeks of culture with chondrocytes revealed a concentration of 0.23 mg of tissue on one part of the membrane. Macroscopic and microscopic evaluation indicated that the quality of regenerated tissue was similar after the transplantation of cells placed on polysulphonic or collagen membranes. The established method for the culture and transplantation of chondrocytes placed on polysulphonic membranes resulted in the growth of the regenerated tissue, revealing the morphology of hyaline-like cartilage to be of similar quality to collagen membranes.
Collapse
Affiliation(s)
- Maciej Płończak
- Mazovia Regional Hospital John Paul II, 08-110 Siedlce, Poland
| | - Monika Wasyłeczko
- Nałęcz Institute of Biocybernetic and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Tomasz Jakutowicz
- Department of Neurosurgery and Children Traumatology, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Andrzej Chwojnowski
- Nałęcz Institute of Biocybernetic and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Jarosław Czubak
- Department of Orthopedics, Pediatric Orthopedics and Traumatology, Centre of Postgraduate Medical Education, Gruca Orthopaedic and Trauma Teaching Hospital, 05-402 Otwock, Poland
| |
Collapse
|
15
|
Liu Y, Graves DT, Wang S. Development and clinical application of human mesenchymal stem cell drugs. Sci Bull (Beijing) 2023; 68:860-863. [PMID: 37045665 PMCID: PMC10784989 DOI: 10.1016/j.scib.2023.03.050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Affiliation(s)
- Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing 100050, China; Beijing Laboratory of Oral Health, Capital Medical University, Beijing 100069, China
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia PA 19104, USA
| | - Songlin Wang
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing 100069, China; Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
16
|
Jankovic MG, Stojkovic M, Bojic S, Jovicic N, Kovacevic MM, Ivosevic Z, Juskovic A, Kovacevic V, Ljujic B. Scaling up human mesenchymal stem cell manufacturing using bioreactors for clinical uses. Curr Res Transl Med 2023; 71:103393. [PMID: 37163885 DOI: 10.1016/j.retram.2023.103393] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/13/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023]
Abstract
Human mesenchymal stem cells (hMSCs) are multipotent cells and an attractive therapeutic agent in regenerative medicine and intensive clinical research. Despite the great potential, the limitation that needs to be overcome is the necessity of ex vivo expansion because of insufficient number of hMSCs presented within adult organs and the high doses required for a transplantation. As a result, numerous research studies aim to provide novel expansion methods in order to achieve appropriate numbers of cells with preserved therapeutic quality. Bioreactor-based cell expansion provide high-level production of hMSCs in accordance with good manufacturing practice (GMP) and quality standards. This review summarizes current knowledge about the hMSCs manufacturing platforms with a main focus to the application of bioreactors for large-scale production of GMP-grade hMSCs.
Collapse
Affiliation(s)
- Marina Gazdic Jankovic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Genetics, Serbia.
| | | | - Sanja Bojic
- Newcastle University, School of Computing, Newcastle upon Tyne, UK
| | - Nemanja Jovicic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Histology and Embryology, Serbia
| | - Marina Miletic Kovacevic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Histology and Embryology, Serbia
| | - Zeljko Ivosevic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Genetics, Serbia
| | - Aleksandar Juskovic
- Department of Orthopaedic Surgery, Clinical Centre of Montenegro, 81110 Podgorica, Montenegro
| | - Vojin Kovacevic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Surgery, Serbia
| | - Biljana Ljujic
- University of Kragujevac, Serbia, Faculty of Medical Sciences, Department of Genetics, Serbia
| |
Collapse
|
17
|
Theodoridis K, Aggelidou E, Manthou ME, Kritis A. Hypoxia Promotes Cartilage Regeneration in Cell-Seeded 3D-Printed Bioscaffolds Cultured with a Bespoke 3D Culture Device. Int J Mol Sci 2023; 24:ijms24076040. [PMID: 37047021 PMCID: PMC10094683 DOI: 10.3390/ijms24076040] [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: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
In this study, we investigated the effect of oxygen tension on the expansion of ADMSCs and on their differentiation toward their chondrocytic phenotype, regenerating a lab-based cartilaginous tissue with superior characteristics. Controversial results with reference to MSCs that were cultured under different hypoxic levels, mainly in 2D culturing settings combined with or without other biochemical stimulus factors, prompted our team to study the role of hypoxia on MSCs chondrogenic differentiation within an absolute 3D environment. Specifically, we used 3D-printed honeycomb-like PCL matrices seeded with ADMSCs in the presence or absence of TGF and cultured with a prototype 3D cell culture device, which was previously shown to favor nutrient/oxygen supply, cell adhesion, and infiltration within scaffolds. These conditions resulted in high-quality hyaline cartilage that was distributed uniformly within scaffolds. The presence of the TGF medium was necessary to successfully produce cartilaginous tissues with superior molecular and increased biomechanical properties. Despite hypoxia's beneficial effect, it was overall not enough to fully differentiate ADMSCs or even promote cell expansion within 3D scaffolds alone.
Collapse
Affiliation(s)
- Konstantinos Theodoridis
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
- CGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
| | - Eleni Aggelidou
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
- CGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
- Basic and Translational Research Unit (BTRU) of Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
| | - Maria-Eleni Manthou
- Laboratory of Histology, Embryology and Anthropology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
| | - Aristeidis Kritis
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
- CGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
- Basic and Translational Research Unit (BTRU) of Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki (A.U.Th), 54124 Thessaloniki, Greece
| |
Collapse
|
18
|
Smith CA, Humphreys PA, Naven MA, Woods S, Mancini FE, O’Flaherty J, Meng QJ, Kimber SJ. Directed differentiation of hPSCs through a simplified lateral plate mesoderm protocol for generation of articular cartilage progenitors. PLoS One 2023; 18:e0280024. [PMID: 36706111 PMCID: PMC9882893 DOI: 10.1371/journal.pone.0280024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 12/20/2022] [Indexed: 01/28/2023] Open
Abstract
Developmentally, the articular joints are derived from lateral plate (LP) mesoderm. However, no study has produced both LP derived prechondrocytes and preosteoblasts from human pluripotent stem cells (hPSC) through a common progenitor in a chemically defined manner. Differentiation of hPSCs through the authentic route, via an LP-osteochondral progenitor (OCP), may aid understanding of human cartilage development and the generation of effective cell therapies for osteoarthritis. We refined our existing chondrogenic protocol, incorporating knowledge from development and other studies to produce a LP-OCP from which prechondrocyte- and preosteoblast-like cells can be generated. Results show the formation of an OCP, which can be further driven to prechondrocytes and preosteoblasts. Prechondrocytes cultured in pellets produced cartilage like matrix with lacunae and superficial flattened cells expressing lubricin. Additionally, preosteoblasts were able to generate a mineralised structure. This protocol can therefore be used to investigate further cartilage development and in the development of joint cartilage for potential treatments.
Collapse
Affiliation(s)
- Christopher A. Smith
- Faculty of Biology, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Paul A. Humphreys
- Faculty of Biology, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Mark A. Naven
- Faculty of Biology, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Steven Woods
- Faculty of Biology, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Fabrizio E. Mancini
- Faculty of Biology, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Julieta O’Flaherty
- Faculty of Biology, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Qing-Jun Meng
- Faculty of Biology, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Susan J. Kimber
- Faculty of Biology, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Medicine and Health, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
19
|
Bono N, Saroglia G, Marcuzzo S, Giagnorio E, Lauria G, Rosini E, De Nardo L, Athanassiou A, Candiani G, Perotto G. Silk fibroin microgels as a platform for cell microencapsulation. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2022; 34:3. [PMID: 36586059 PMCID: PMC9805413 DOI: 10.1007/s10856-022-06706-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/27/2022] [Indexed: 06/17/2023]
Abstract
Cell microencapsulation has been utilized for years as a means of cell shielding from the external environment while facilitating the transport of gases, general metabolites, and secretory bioactive molecules at once. In this light, hydrogels may support the structural integrity and functionality of encapsulated biologics whereas ensuring cell viability and function and releasing potential therapeutic factors once in situ. In this work, we describe a straightforward strategy to fabricate silk fibroin (SF) microgels (µgels) and encapsulate cells into them. SF µgels (size ≈ 200 µm) were obtained through ultrasonication-induced gelation of SF in a water-oil emulsion phase. A thorough physicochemical (SEM analysis, and FT-IR) and mechanical (microindentation tests) characterization of SF µgels were carried out to assess their nanostructure, porosity, and stiffness. SF µgels were used to encapsulate and culture L929 and primary myoblasts. Interestingly, SF µgels showed a selective release of relatively small proteins (e.g., VEGF, molecular weight, MW = 40 kDa) by the encapsulated primary myoblasts, while bigger (macro)molecules (MW = 160 kDa) were hampered to diffusing through the µgels. This article provided the groundwork to expand the use of SF hydrogels into a versatile platform for encapsulating relevant cells able to release paracrine factors potentially regulating tissue and/or organ functions, thus promoting their regeneration.
Collapse
Affiliation(s)
- Nina Bono
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Via Mancinelli 7, 20131, Milan, Italy.
| | - Giulio Saroglia
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Via Mancinelli 7, 20131, Milan, Italy
- Smart Materials, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genova, Italy
| | - Stefania Marcuzzo
- Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133, Milan, Italy
| | - Eleonora Giagnorio
- Neurology IV-Neuroimmunology and Neuromuscular Diseases Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133, Milan, Italy
| | - Giuseppe Lauria
- Department of Clinical Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, Via Celoria 11, 20133, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Via Vanvitelli 32, 20133, Milan, Italy
| | - Elena Rosini
- The Protein Factory 2.0, Department of Biotechnology and Life Sciences, University of Insubria, Via J.H. Dunant 3, 21100, Varese, Italy
| | - Luigi De Nardo
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Via Mancinelli 7, 20131, Milan, Italy
| | | | - Gabriele Candiani
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Via Mancinelli 7, 20131, Milan, Italy
| | - Giovanni Perotto
- Smart Materials, Istituto Italiano di Tecnologia, Via Morego 30, 16163, Genova, Italy.
| |
Collapse
|
20
|
O'Connell CD, Duchi S, Onofrillo C, Caballero‐Aguilar LM, Trengove A, Doyle SE, Zywicki WJ, Pirogova E, Di Bella C. Within or Without You? A Perspective Comparing In Situ and Ex Situ Tissue Engineering Strategies for Articular Cartilage Repair. Adv Healthc Mater 2022; 11:e2201305. [PMID: 36541723 PMCID: PMC11468013 DOI: 10.1002/adhm.202201305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/21/2022] [Indexed: 11/23/2022]
Abstract
Human articular cartilage has a poor ability to self-repair, meaning small injuries often lead to osteoarthritis, a painful and debilitating condition which is a major contributor to the global burden of disease. Existing clinical strategies generally do not regenerate hyaline type cartilage, motivating research toward tissue engineering solutions. Prospective cartilage tissue engineering therapies can be placed into two broad categories: i) Ex situ strategies, where cartilage tissue constructs are engineered in the lab prior to implantation and ii) in situ strategies, where cells and/or a bioscaffold are delivered to the defect site to stimulate chondral repair directly. While commonalities exist between these two approaches, the core point of distinction-whether chondrogenesis primarily occurs "within" or "without" (outside) the body-can dictate many aspects of the treatment. This difference influences decisions around cell selection, the biomaterials formulation and the surgical implantation procedure, the processes of tissue integration and maturation, as well as, the prospects for regulatory clearance and clinical translation. Here, ex situ and in situ cartilage engineering strategies are compared: Highlighting their respective challenges, opportunities, and prospects on their translational pathways toward long term human cartilage repair.
Collapse
Affiliation(s)
- Cathal D. O'Connell
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Serena Duchi
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Carmine Onofrillo
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
| | - Lilith M. Caballero‐Aguilar
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- School of ScienceComputing and Engineering TechnologiesSwinburne University of TechnologyMelbourneVictoria3122Australia
| | - Anna Trengove
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Stephanie E. Doyle
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| | - Wiktor J. Zywicki
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of Biomedical EngineeringUniversity of MelbourneMelbourneVictoria3010Australia
| | - Elena Pirogova
- Discipline of Electrical and Biomedical EngineeringRMIT UniversityMelbourneVictoria3000Australia
| | - Claudia Di Bella
- Aikenhead Centre for Medical Discovery (ACMD)St Vincent's Hospital MelbourneFitzroyVictoria3065Australia
- Department of SurgerySt Vincent's HospitalUniversity of MelbourneFitzroyVictoria3065Australia
- Department of MedicineSt Vincent's Hospital MelbourneFitzroyVictoria3065Australia
| |
Collapse
|
21
|
Sadri B, Tamimi A, Nouraein S, Bagheri Fard A, Mohammadi J, Mohammadpour M, Hassanzadeh M, Bajouri A, Madani H, Barekat M, Karimi Torshizi S, Malek M, Ghorbani Liastani M, Beheshti Maal A, Niknejadi M, Vosough M. Clinical and laboratory findings following transplantation of allogeneic adipose-derived mesenchymal stromal cells in knee osteoarthritis, a brief report. Connect Tissue Res 2022; 63:663-674. [PMID: 35856397 DOI: 10.1080/03008207.2022.2074841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) injection has been proposed as an innovative treatment for knee osteoarthritis (KOA). Since, allogeneic MSCs can be available as off-the-shelf products, they are preferable in regenerative medicine. Among different sources for MSCs, adipose-derived MSCs (AD-MSCs) appear to be more available. METHODS Three patients with KOA were enrolled in this study. A total number of 100 × 106 AD-MSCs was injected intra-articularly, per affected knee. They were followed up for 6 months by the assessment of clinical outcomes, magnetic resonance imaging (MRI), and serum inflammatory biomarkers. RESULTS The primary outcome of this study was safety and feasibility of allogeneic AD-MSCs injection during the 6 months follow-up. Fortunately, no serious adverse events (SAEs) were reported. Assessment of secondary outcomes of visual analogue scale (VAS), Western Ontario and McMaster Universities Osteoarthritis Index (WOMAC), and knee osteoarthritis outcome score (KOOS) indicated improvement in all patients. Comparison between baseline and endpoint findings of MRI demonstrated a slight improvement in two patients. In addition, decrease in serum cartilage oligomeric matrix protein (COMP) and hyaluronic acid (HA) indicated the possibility of reduced cartilage degeneration. Moreover, quantification of serum interleukin-10 (IL-10) and interleukin-6 (IL-6) levels indicated that the host immune system immunomodulated after infusion of AD-MSCs. CONCLUSION Intra-articular injection of AD-MSCs is safe and could be effective in cartilage regeneration in KOA. Preliminary assessment after six-month follow-up suggests the potential efficacy of this intervention which would need to be confirmed in randomized controlled trials on a larger population. TRIAL REGISTRATION This study was registered in the Iranian registry of clinical trials (https://en.irct.ir/trial/46) in 24 April 2018 with identifier IRCT20080728001031N23.
Collapse
Affiliation(s)
- Bahareh Sadri
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Atena Tamimi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shirin Nouraein
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Abolfazl Bagheri Fard
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Mohammadi
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mehdi Mohammadpour
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hassanzadeh
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amir Bajouri
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hoda Madani
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Barekat
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahedeh Karimi Torshizi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahrooz Malek
- Department of Radiology, Medical Imaging Center, Advanced Diagnostic and Interventional Radiology Research Center (ADIR), Tehran University of Medical Sciences, Imam Khomeini Hospital, Tehran, Iran
| | - Maede Ghorbani Liastani
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Alireza Beheshti Maal
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Maryam Niknejadi
- Department of Reproductive Imaging, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
22
|
Yoon DS, Lee KM, Choi Y, Ko EA, Lee NH, Cho S, Park KH, Lee JH, Kim HW, Lee JW. TLR4 downregulation by the RNA-binding protein PUM1 alleviates cellular aging and osteoarthritis. Cell Death Differ 2022; 29:1364-1378. [PMID: 35034101 DOI: 10.1038/s41418-021-00925-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/04/2021] [Accepted: 12/07/2021] [Indexed: 12/27/2022] Open
Abstract
Dysfunction of mRNA or RNA-binding proteins (RBPs) causes cellular aging and age-related degenerative diseases; however, information regarding the mechanism through which RBP-mediated posttranscriptional regulation affects cellular aging and related disease processes is limited. In this study, PUM1 was found to be associated with the self-renewal capacity and aging process of human mesenchymal stem cells (MSC). PUM1 interacted with the 3'-untranslated region of Toll-like receptor 4 (TLR4) to suppress TLR4 mRNA translation and regulate the activity of nuclear factor-κB (NF-κB), a master regulator of the aging process in MSCs. PUM1 overexpression protected MSCs against H2O2-induced cellular senescence by suppressing TLR4-mediated NF-κB activity. TLR4-mediated NF-κB activation is a key regulator in osteoarthritis (OA) pathogenesis. PUM1 overexpression enhanced the chondrogenic potential of MSCs even under the influence of inflammation-inducing factors, such as lipopolysaccharide (LPS) or interleukin-1β (IL-1β), whereas the chondrogenic potential was reduced following the PUM1 knockdown-mediated TLR4 activation. PUM1 levels decreased under inflammatory conditions in vitro and during OA progression in human and mouse disease models. PUM1 knockdown in human chondrocytes promoted chondrogenic phenotype loss, whereas PUM1 overexpression protected the cells from inflammation-mediated disruption of the chondrogenic phenotype. Gene therapy using a lentiviral vector encoding mouse PUM1 showed promise in preserving articular cartilage integrity in OA mouse models. In conclusion, PUM1 is a novel suppressor of MSC aging, and the PUM1-TLR4 regulatory axis represents a potential therapeutic target for OA.
Collapse
Affiliation(s)
- Dong Suk Yoon
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Yoorim Choi
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Eun Ae Ko
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Na-Hyun Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, South Korea.,Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, South Korea
| | - Sehee Cho
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea.,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Kwang Hwan Park
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, South Korea.,Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, South Korea.,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, South Korea.,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, South Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, South Korea. .,Department of Nanobiomedical Science and BK21 PLUS NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, South Korea. .,Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan, 31116, South Korea. .,UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, South Korea.
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, 03722, South Korea. .,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, 03722, South Korea. .,Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
23
|
Fayyazpour P, Alizadeh E, Hosseini V, Kalantary-Charvadeh A, Niafar M, Sadra V, Norouzi Z, Saebnazar A, Mehdizadeh A, Darabi M. Fatty acids of type 2 diabetic serum decrease the stemness properties of human adipose-derived mesenchymal stem cells. J Cell Biochem 2022; 123:1157-1170. [PMID: 35722966 DOI: 10.1002/jcb.30270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 03/26/2022] [Accepted: 04/20/2022] [Indexed: 11/11/2022]
Abstract
In type 2 diabetes, dyslipidemia and increased serum free fatty acids (FFAs) exacerbate the development of the disease through a negative effect on insulin secretion. Adipose-derived mesenchymal stem cells (AdMSCs) play a key role in regenerative medicine, and these cells can potentially be applied as novel therapeutic resources in the treatment of diabetes. In this study, AdMSCs were treated with diabetic or nondiabetic serum FFAs isolated from women of menopausal age. Serum FFAs were analyzed using gas-liquid chromatography. The expression level of the stemness markers CD49e and CD90 and the Wnt signaling target genes Axin-2 and c-Myc were evaluated using real-time PCR. The proliferation rate and colony formation were also assessed using a BrdU assay and crystal violet staining, respectively. The level of glutathione was assessed using cell fluorescence staining. Compared to nondiabetic serum, diabetic serum contained a higher percentage of oleate (1.5-fold, p < 0.01). In comparison with nondiabetic FFAs, diabetic FFAs demonstrated decreasing effects on the expression of CD90 (-51%, p < 0.001) and c-Myc (-48%, p < 0.05), and proliferation rate (-35%, p < 0.001), colony formation capacity (-50%, p < 0.01), and GSH levels (-62%, p < 0.05). The negative effect of the FFAs of diabetic serum on the stemness characteristics may impair the regenerative capabilities of AdMSCs.
Collapse
Affiliation(s)
- Parisa Fayyazpour
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Hosseini
- Molecular Medicine Research Center, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ashkan Kalantary-Charvadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mitra Niafar
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahideh Sadra
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Norouzi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysan Saebnazar
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Masoud Darabi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Internal Medicine IV, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
24
|
Kim HR, Choi H, Park SY, Song YC, Kim JH, Shim S, Jun W, Kim KJ, Han J, Chi SW, Leem SH, Chung JW. Endoplasmin regulates differentiation of tonsil-derived mesenchymal stem cells into chondrocytes through ERK signaling. BMB Rep 2022. [PMID: 35168699 PMCID: PMC9152576 DOI: 10.5483/bmbrep.2022.55.5.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is well-known that some species of lizard have an exceptional ability known as caudal autotomy (voluntary self-amputation of the tail) as an anti-predation mechanism. After amputation occurs, they can regenerate their new tails in a few days. The new tail section is generally shorter than the original one and is composed of cartilage rather than vertebrae bone. In addition, the skin of the regenerated tail distinctly differs from its original appearance. We performed a proteomics analysis for extracts derived from regenerating lizard tail tissues after amputation and found that endoplasmin (ENPL) was the main factor among proteins up-regulated in expression during regeneration. Thus, we performed further experiments to determine whether ENPL could induce chondrogenesis of tonsil-derived mesenchymal stem cells (T-MSCs). In this study, we found that chondrogenic differentiation was associated with an increase of ENPL expression by ER stress. We also found that ENPL was involved in chondrogenic differentiation of T-MSCs by suppressing extracellular signal-regulated kinase (ERK) phosphorylation.
Collapse
Affiliation(s)
- Hye Ryeong Kim
- Department of Biological Science, Dong-A University, Busan 49315, Korea
| | - Hyeongrok Choi
- Department of Biological Science, Dong-A University, Busan 49315, Korea
| | - Soon Yong Park
- Department of Research Center, Dongnam Institute of Radiological & Medical Sciences, Busan 46033, Korea
| | - Young-Chul Song
- Department of Physiology, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Jae-Ho Kim
- Department of Physiology, Pusan National University School of Medicine, Yangsan 50612, Korea
| | - Sangin Shim
- Department of Agronomy, Gyeongsang National University, Jinju 52828, Korea
| | - Woojin Jun
- Department of Food and Nutrition, Chonnam National University, Gwangju 61186, Korea
| | - Kyung-Jin Kim
- Department of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Jin Han
- Department of Physiology, College of Medicine, Inje University, Busan 47392, Korea
| | - Seung-Wook Chi
- Disease Target Structure Research Center, KRIBB, Daejeon 34141, Korea
| | - Sun-Hee Leem
- Department of Health Sciences, Dong-A University, Busan 49315, Korea
| | - Jin Woong Chung
- Department of Biological Science, Dong-A University, Busan 49315, Korea
| |
Collapse
|
25
|
Srinivasan A, Sathiyanathan P, Yin L, Liu TM, Lam A, Ravikumar M, Smith RAA, Loh HP, Zhang Y, Ling L, Ng SK, Yang YS, Lezhava A, Hui J, Oh S, Cool SM. Strategies to enhance immunomodulatory properties and reduce heterogeneity in mesenchymal stromal cells during ex vivo expansion. Cytotherapy 2022; 24:456-472. [PMID: 35227601 DOI: 10.1016/j.jcyt.2021.11.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 02/06/2023]
Abstract
Therapies using mesenchymal stromal cells (MSCs) to treat immune and inflammatory conditions are now at an exciting stage of development, with many MSC-based products progressing to phase II and III clinical trials. However, a major bottleneck in the clinical translation of allogeneic MSC therapies is the variable immunomodulatory properties of MSC products due to differences in their tissue source, donor heterogeneity and processes involved in manufacturing and banking. This variable functionality of MSC products likely contributes to the substantial inconsistency observed in the clinical outcomes of phase III trials of MSC therapies; several trials have failed to reach the primary efficacy endpoint. In this review, we discuss various strategies to consistently maintain or enhance the immunomodulatory potency of MSCs during ex vivo expansion, which will enable the manufacture of allogeneic MSC banks that have high potency and low variability. Biophysical and biochemical priming strategies, the use of culture additives such as heparan sulfates, and genetic modification can substantially enhance the immunomodulatory properties of MSCs during in vitro expansion. Furthermore, robust donor screening, the use of biomarkers to select for potent MSC subpopulations, and rigorous quality testing to improve the release criteria for MSC banks have the potential to reduce batch-to-batch heterogeneity and enhance the clinical efficacy of the final MSC product. Machine learning approaches to develop predictive models of individual patient response can enable personalized therapies and potentially establish correlations between in vitro potency measurements and clinical outcomes in human trials.
Collapse
Affiliation(s)
- Akshaya Srinivasan
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Lu Yin
- Bioprocessing Technology Institute, A*STAR, Singapore
| | - Tong Ming Liu
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Alan Lam
- Bioprocessing Technology Institute, A*STAR, Singapore
| | - Maanasa Ravikumar
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, A*STAR, Singapore
| | | | - Han Ping Loh
- Bioprocessing Technology Institute, A*STAR, Singapore
| | - Ying Zhang
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Ling Ling
- Institute of Molecular and Cell Biology, A*STAR, Singapore
| | - Say Kong Ng
- Bioprocessing Technology Institute, A*STAR, Singapore
| | | | - Alexander Lezhava
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore
| | - James Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Steve Oh
- Bioprocessing Technology Institute, A*STAR, Singapore.
| | - Simon M Cool
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute of Molecular and Cell Biology, A*STAR, Singapore.
| |
Collapse
|
26
|
Aladal M, You W, Huang R, Huang J, Deng Z, Duan L, Wang D, Li W, Sun W. Insights into the implementation of Fibronectin 1 in the cartilage tissue engineering. Biomed Pharmacother 2022; 148:112782. [PMID: 35248846 DOI: 10.1016/j.biopha.2022.112782] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 11/02/2022] Open
Abstract
Recently, cartilage tissue engineering has become a cornerstone to treat cartilage degeneration and osteoarthritis (OA). Fibronectin1 (FN1) is described as multiple functional glycoproteins that play an essential role in chondrogenic and osteogenic differentiation. Few studies reported the potential of FN1 to enhance tissue engineering and reduce the death of chondrocytes in OA. Further, FN1 possesses multiple binding domains including collagen, integrin, and heparin that can interact with heparan sulfate proteoglycans at the surface of chondrocyte leading to promote cell signaling and differentiation. Recent studies suggested that FN1 can promote chondrocyte differentiation by upregulating TGF-β/PI3K/Akt pathways. Further, FN1 can inhibit the apoptosis of chondrocytes by preventing the release of metalloproteinases through lowering the expression of p-PI3K/PI3K and p-AKT/AKT pathways. However, the use of FN1 in cartilage repair studies using animal models or clinical trials was rarely reported. Therefore, this article provides new insights into the importance of FN1 in cartilage tissue engineering to encourage more studies concerning FN1 in cartilage repair studies. Further, we provided new suggestions for advanced applications of FN1 to treat OA and cartilage degeneration.
Collapse
Affiliation(s)
- Murad Aladal
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518000, China; Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center), Shenzhen 518035, China
| | - Wei You
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518000, China
| | - Rongxiang Huang
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518000, China
| | - Jianghong Huang
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518000, China
| | - Zhiqin Deng
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518000, China
| | - Li Duan
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518000, China
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Laboratory of Digital Orthopedic Engineering, Guangdong Provincial Research Center for Artificial Intelligence and Digital Orthopedic Technology, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University, Health Science Center), Shenzhen 518035, China; Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, China.
| | - Wencui Li
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518000, China.
| | - Wei Sun
- Hand and Foot Surgery Department, Shenzhen Second People's Hospital (The First Hospital Affiliated to Shenzhen University), Shenzhen 518000, China.
| |
Collapse
|
27
|
Gaobotse G, Mbunge E, Batani J, Muchemwa B. The future of smart implants towards personalized and pervasive healthcare in Sub-Saharan Africa: Opportunities, barriers and policy recommendations. SENSORS INTERNATIONAL 2022. [DOI: 10.1016/j.sintl.2022.100173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
28
|
Three-dimensional scaffolds for tissue bioengineering cartilages. Biocybern Biomed Eng 2022. [DOI: 10.1016/j.bbe.2022.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
29
|
Gupta A, Lee J, Ghosh T, Nguyen VQ, Dey A, Yoon B, Um W, Park JH. Polymeric Hydrogels for Controlled Drug Delivery to Treat Arthritis. Pharmaceutics 2022; 14:540. [PMID: 35335915 PMCID: PMC8948938 DOI: 10.3390/pharmaceutics14030540] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 12/31/2022] Open
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are disabling musculoskeletal disorders that affect joints and cartilage and may lead to bone degeneration. Conventional delivery of anti-arthritic agents is limited due to short intra-articular half-life and toxicities. Innovations in polymer chemistry have led to advancements in hydrogel technology, offering a versatile drug delivery platform exhibiting tissue-like properties with tunable drug loading and high residence time properties This review discusses the advantages and drawbacks of polymeric materials along with their modifications as well as their applications for fabricating hydrogels loaded with therapeutic agents (small molecule drugs, immunotherapeutic agents, and cells). Emphasis is given to the biological potentialities of hydrogel hybrid systems/micro-and nanotechnology-integrated hydrogels as promising tools. Applications for facile tuning of therapeutic drug loading, maintaining long-term release, and consequently improving therapeutic outcome and patient compliance in arthritis are detailed. This review also suggests the advantages, challenges, and future perspectives of hydrogels loaded with anti-arthritic agents with high therapeutic potential that may alter the landscape of currently available arthritis treatment modalities.
Collapse
Affiliation(s)
- Anuradha Gupta
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Jungmi Lee
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Torsha Ghosh
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Van Quy Nguyen
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Anup Dey
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Been Yoon
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Wooram Um
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University, Suwon 16419, Korea; (A.G.); (J.L.); (T.G.); (V.Q.N.); (A.D.); (B.Y.); (W.U.)
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
30
|
Herger N, Bermudez-Lekerika P, Farshad M, Albers CE, Distler O, Gantenbein B, Dudli S. Should Degenerated Intervertebral Discs of Patients with Modic Type 1 Changes Be Treated with Mesenchymal Stem Cells? Int J Mol Sci 2022; 23:ijms23052721. [PMID: 35269863 PMCID: PMC8910866 DOI: 10.3390/ijms23052721] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 12/16/2022] Open
Abstract
Low back pain (LBP) has been among the leading causes of disability for the past 30 years. This highlights the need for improvement in LBP management. Many clinical trials focus on developing treatments against degenerative disc disease (DDD). The multifactorial etiology of DDD and associated risk factors lead to a heterogeneous patient population. It comes as no surprise that the outcomes of clinical trials on intradiscal mesenchymal stem cell (MSC) injections for patients with DDD are inconsistent. Intradiscal MSC injections have demonstrated substantial pain relief and significant disability-related improvements, yet they have failed to regenerate the intervertebral disc (IVD). Increasing evidence suggests that the positive outcomes in clinical trials might be attributed to the immunomodulatory potential of MSCs rather than to their regenerative properties. Therefore, patient stratification for inflammatory DDD phenotypes may (i) better serve the mechanisms of action of MSCs and (ii) increase the treatment effect. Modic type 1 changes—pathologic inflammatory, fibrotic changes in the vertebral bone marrow—are frequently observed adjacent to degenerated IVDs in chronic LBP patients and represent a clinically distinct subpopulation of patients with DDD. This review discusses whether degenerated IVDs of patients with Modic type 1 changes should be treated with an intradiscal MSC injection.
Collapse
Affiliation(s)
- Nick Herger
- Center of Experimental Rheumatology, University Hospital Zurich and Balgrist University Hospital, University of Zurich, CH-8008 Zurich, Switzerland; (N.H.); (O.D.)
| | - Paola Bermudez-Lekerika
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, CH-3008 Bern, Switzerland; (P.B.-L.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Medical Faculty, University of Bern, CH-3010 Bern, Switzerland;
| | - Mazda Farshad
- Department of Orthopaedics, Balgrist University Hospital, CH-8008 Zurich, Switzerland;
| | - Christoph E. Albers
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Medical Faculty, University of Bern, CH-3010 Bern, Switzerland;
| | - Oliver Distler
- Center of Experimental Rheumatology, University Hospital Zurich and Balgrist University Hospital, University of Zurich, CH-8008 Zurich, Switzerland; (N.H.); (O.D.)
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, CH-3008 Bern, Switzerland; (P.B.-L.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, Medical Faculty, University of Bern, CH-3010 Bern, Switzerland;
| | - Stefan Dudli
- Center of Experimental Rheumatology, University Hospital Zurich and Balgrist University Hospital, University of Zurich, CH-8008 Zurich, Switzerland; (N.H.); (O.D.)
- Correspondence: ; Tel.: +41-4451-07511
| |
Collapse
|
31
|
Jeyaraman M, Shivaraj B, Bingi SK, Ranjan R, Muthu S, Khanna M. Does vehicle-based delivery of mesenchymal stromal cells give superior results in knee osteoarthritis? Meta-analysis of randomized controlled trials. J Clin Orthop Trauma 2022; 25:101772. [PMID: 35127439 PMCID: PMC8803619 DOI: 10.1016/j.jcot.2022.101772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/06/2022] [Accepted: 01/13/2022] [Indexed: 02/08/2023] Open
Abstract
STUDY DESIGN Meta-analysis. OBJECTIVES We aim to analyze and compare the efficacy and safety of vehicle-based delivery of Mesenchymal Stromal Cells (MSCs) in the management of osteoarthritis of the knee from Randomized Controlled Trials (RCTs) available in the literature. MATERIALS AND METHODS We conducted independent and duplicate electronic database searches including PubMed, Embase, Web of Science, and Cochrane Library till August 2021 for RCTs analyzing the efficacy and safety of vehicle-based delivery of MSCs in the management of knee osteoarthritis. Visual Analog Score (VAS) for Pain, Western Ontario McMaster Universities Osteoarthritis Index (WOMAC), Magnetic Resonance Observation of Cartilage Repair Tissue (MOCART) score, and adverse events were the outcomes analyzed. Analysis was performed in R-platform using OpenMeta [Analyst] software. RESULTS 21 studies involving 936 patients were included for analysis. None of the studies made a direct comparison of the direct and vehicle-based delivery of MSCs, hence we pooled the results of all the included studies of both groups and made a comparative analysis of their outcomes. Although at 6 months, both direct and vehicle-based delivery of MSCs showed significantly better VAS improvement (p = 0.002, p = 0.010), it was not consistent at 1 year for the vehicle delivery (p = 0.973). During 6 months and 12 months, direct delivery of MSCs (p < 0.001, p < 0.001) outperformed vehicle delivery (p = 0.969, p = 0.922) compared to their control based on WOMAC scores respectively. Both direct (p = 0.713) and vehicle-based delivery (p = 0.123) of MSCs did not produce significant adverse events compared to their controls. CONCLUSION Our analysis of literature showed that current clinically employed methods of vehicle-based delivery of MSCs such as platelet-rich plasma, hyaluronic acid did not demonstrate superior results compared to direct delivery, concerning the efficacy of treatment measured by improvement in pain, functional outcomes, and safety. Hence, we urge future clinical trials to be conducted to validate the effectiveness of advanced delivery vehicles such as composite bioscaffolds to establish their practical utility in cartilage regeneration with respect to its encouraging in-vitro evidence.
Collapse
Affiliation(s)
- Madhan Jeyaraman
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
| | - B. Shivaraj
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Dr. RML National Law University, Lucknow, Uttar Pradesh, India
| | - Shiva Kumar Bingi
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Dr. RML National Law University, Lucknow, Uttar Pradesh, India
| | - Rajni Ranjan
- Department of Orthopaedics, School of Medical Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Sathish Muthu
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, India
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
- Department of Orthopaedics, Government Medical College and Hospital, Dindigul, Tamil Nadu, India
| | - Manish Khanna
- Indian Stem Cell Study Group (ISCSG) Association, Lucknow, Uttar Pradesh, India
| |
Collapse
|
32
|
Lee DH, Kim SJ, Kim SA, Ju GI. Past, present, and future of cartilage restoration: from localized defect to arthritis. Knee Surg Relat Res 2022; 34:1. [PMID: 35090574 PMCID: PMC8800252 DOI: 10.1186/s43019-022-00132-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/11/2022] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Osteoarthritis, one of the most common joint diseases, is characterized by the loss of joint function due to articular cartilage destruction. Herein, we review current and previous research involving the clinical applications of arthritis therapy and suggest potential therapeutic options for osteoarthritis in the future. PAST, PRESENT, AND FUTURE TREATMENT The arthroscopic cartilage regeneration procedure or realignment osteotomy has been performed as a joint-conserving procedure in cases where conservative treatment for damaged articular cartilage and early osteoarthritis failed. If cartilage regeneration is ineffective or if the joint damage progresses, arthroplasty is the main treatment option. The need for biological arthritis treatment has expanded as the healthy lifespan of the global population has increased. Accordingly, minimally invasive surgical treatment has been developed for the treatment of damaged cartilage and early osteoarthritis. However, patients generally prefer to avoid all types of surgery, including minimally invasive surgery. Therefore, in the future, the treatment of osteoarthritis will likely involve injection or medication. CONCLUSION Currently, arthritis management primarily involves the surgical application of therapeutic agents to the joints. However, nonsurgical or prophylactic methods are expected to become mainstream arthritis therapies in the future.
Collapse
Affiliation(s)
- Dong Hwan Lee
- Department of Orthopedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 271, Cheonbo-ro, Gyeonggi-do, 11765, Uijeongbu-si, Republic of Korea
| | - Seok Jung Kim
- Department of Orthopedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 271, Cheonbo-ro, Gyeonggi-do, 11765, Uijeongbu-si, Republic of Korea.
| | - Seon Ae Kim
- Department of Orthopedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 271, Cheonbo-ro, Gyeonggi-do, 11765, Uijeongbu-si, Republic of Korea
| | - Gang-Ik Ju
- Department of Orthopedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 271, Cheonbo-ro, Gyeonggi-do, 11765, Uijeongbu-si, Republic of Korea
| |
Collapse
|
33
|
Roncada T, Bonithon R, Blunn G, Roldo M. Soft substrates direct stem cell differentiation into the chondrogenic lineage without the use of growth factors. J Tissue Eng 2022; 13:20417314221122121. [PMID: 36199979 PMCID: PMC9528007 DOI: 10.1177/20417314221122121] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/03/2022] [Indexed: 12/03/2022] Open
Abstract
Mesenchymal stem cells (MSCs) hold great promise for the treatment of cartilage related injuries. However, selectively promoting stem cell differentiation in vivo is still challenging. Chondrogenic differentiation of MSCs usually requires the use of growth factors that lead to the overexpression of hypertrophic markers. In this study, for the first time the effect of stiffness on MSC differentiation has been tested without the use of growth factors. Three-dimensional collagen and alginate scaffolds were developed and characterised. Stiffness significantly affected gene expression and ECM deposition. While, all hydrogels supported chondrogenic differentiation and allowed deposition of collagen type II and aggrecan, the 5.75 kPa hydrogel showed limited production of collagen type I compared to the other two formulations. These findings demonstrated for the first time that stiffness can guide MSCs differentiation without the use of growth factors within a tissue engineering scaffold suitable for the treatment of cartilage defects.
Collapse
Affiliation(s)
- Tosca Roncada
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Roxane Bonithon
- School of Mechanical and Design Engineering, University of Portsmouth, Portsmouth, UK
| | - Gordon Blunn
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
| | - Marta Roldo
- School of Pharmacy and Biomedical Sciences, University of Portsmouth, Portsmouth, UK
- Marta Roldo, School of Pharmacy and Biomedical Sciences, University of Portsmouth, White Swan Road, Portsmouth, PO1 2DT, UK.
| |
Collapse
|
34
|
James JL, Umapathy A, Srinivasan S, Barker CN, Brooks A, Hearn J, Chhana A, Williams E, Sheppard H, McGlashan SR. The Chondrogenic Potential of First-Trimester and Term Placental Mesenchymal Stem/Stromal Cells. Cartilage 2021; 13:544S-558S. [PMID: 34521248 PMCID: PMC8804733 DOI: 10.1177/19476035211044822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVES Mesenchymal stem/stromal cells (MSCs) are a well-established cell source for cartilage engineering, but challenges remain as differentiation often results in chondrocyte hypertrophy. Chondrogenic potential also varies with MSC source and donor age. We assessed the chondrogenic potential of first-trimester and term placental MSCs and compared their response to commonly used bone marrow MSCs (BM-MSCs). DESIGN MSCs were isolated from first-trimester and term placentae. BM-MSCs were commercially obtained. Chondrogenesis was induced by micromass culture in commercial chondrogenic media for 7, 14, or 21 days. Pellets were assessed for glycosaminoglycan (GAG) content, and types I, II, and X collagen. Gene expression was profiled using Qiagen RT2 human MSC arrays. RESULTS At day 0, first-trimester and term MSCs expression levels of many chondrogenic genes to BM-MSC after 21 days of culture. Only first trimester MSCs showed significant changes in chondrogenic gene expression during induction compared to day 0 undifferentiated MSCs (greater BMP4, KAT2B, and reduced GDF6 expression). Additionally, first-trimester MSCs showed significantly greater expression of ABCB1 (at days 14 and 21) and BMP4 (at days 7, 14, 21) compared with term MSCs. Both first-trimester and term pellets showed increased GAG content over time and term MSCs had significantly GAG greater compared with BM-MSCs at days 7 and 14. Type II collagen was present in all pellets but unlike BM-MSCs, type I collagen was not observed in first-trimester or term MSC pellets. CONCLUSIONS These data highlight differences in BM-MSC and placental MSC chondrogenesis and demonstrate that placental MSCs may be an alternative cell source.
Collapse
Affiliation(s)
- Joanna L. James
- Obstetrics and Gynaecology, The
University of Auckland, Auckland, New Zealand
| | - Anandita Umapathy
- Obstetrics and Gynaecology, The
University of Auckland, Auckland, New Zealand
| | - Sonia Srinivasan
- Obstetrics and Gynaecology, The
University of Auckland, Auckland, New Zealand
| | - Claire N. Barker
- Anatomy and Medical Imaging, The
University of Auckland, Auckland, New Zealand
| | - Anna Brooks
- School of Biological Sciences, The
University of Auckland, Auckland, New Zealand
| | - James Hearn
- Molecular Medicine and Pathology, The
University of Auckland, Auckland, New Zealand
| | - Ashika Chhana
- Anatomy and Medical Imaging, The
University of Auckland, Auckland, New Zealand
| | - Eloise Williams
- Obstetrics and Gynaecology, The
University of Auckland, Auckland, New Zealand
| | - Hilary Sheppard
- School of Biological Sciences, The
University of Auckland, Auckland, New Zealand
| | - Sue R. McGlashan
- Anatomy and Medical Imaging, The
University of Auckland, Auckland, New Zealand,Sue R. McGlashan, Department of Anatomy and
Medical Imaging, School of Medical Sciences, The University of Auckland, Faculty
of Medical and Health Sciences, 85 Park Road, Grafton, Auckland 1142, New
Zealand.
| |
Collapse
|
35
|
Menshikov M, Zubkova E, Stafeev I, Parfyonova Y. Autophagy, Mesenchymal Stem Cell Differentiation, and Secretion. Biomedicines 2021; 9:biomedicines9091178. [PMID: 34572364 PMCID: PMC8467641 DOI: 10.3390/biomedicines9091178] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSC) are multipotent cells capable to differentiate into adipogenic, osteogenic, and chondrogenic directions, possessing immunomodulatory activity and a capability to stimulate angiogenesis. A scope of these features and capabilities makes MSC a significant factor of tissue homeostasis and repair. Among factors determining the fate of MSC, a prominent place belongs to autophagy, which is activated under different conditions including cell starvation, inflammation, oxidative stress, and some others. In addition to supporting cell homeostasis by elimination of protein aggregates, and non-functional and damaged proteins, autophagy is a necessary factor of change in cell phenotype on the process of cell differentiation. In present review, some mechanisms providing participation of autophagy in cell differentiation are discussed
Collapse
|
36
|
Lee SJ, Nam Y, Rim YA, Lee K, Ju JH, Kim DS. Perichondrium-inspired permeable nanofibrous tube well promoting differentiation of hiPSC-derived pellet toward hyaline-like cartilage pellet. Biofabrication 2021; 13. [PMID: 34404032 DOI: 10.1088/1758-5090/ac1e76] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 08/17/2021] [Indexed: 01/22/2023]
Abstract
The pellet formation has been regarded as a golden standard forin vitrochondrogenic differentiation. However, a spatially inhomogeneous chondrogenic microenvironment around a pellet resulted from the use of a traditional impermeable narrow tube, such as the conical tube, undermines the differentiation performance and therapeutic potential of differentiated cartilage pellet in defective articular cartilage treatment. To address this drawback, a perichondrium-inspired permeable nanofibrous tube (PINaT) well with a nanofibrous wall permeable to gas and soluble molecules is proposed. The PINaT well was fabricated with a micro deep drawing process where a flat thin nanofibrous membrane was transformed to a 3.5 mm deep tube well with a ∼50µm thick nanofibrous wall. Similar toin vivoperichondrium, the PINaT well was found to allow oxygen and growth factor diffusion required for chondrogenic differentiation across the entire nanofibrous wall. Analyses of gene expressions (COL2A1, COL10A1, ACAN, and SOX9), proteins (type II and X collagen), and glycosaminoglycans contents were conducted to assess the differentiation performance and clinical efficacy of differentiated cartilage pellet. The regulated spatially homogeneous chondrogenic microenvironment around the human induced pluripotent stem cell-derived pellet (3 × 105cells per pellet) in the PINaT well remarkably improved the quality of the differentiated pellet toward a more hyaline-like cartilage pellet. Furthermore, an accelerated chondrogenic differentiation process of the pellet produced by the PINaT well was achieved for 14 days, demonstrating a hyaline cartilage-specific marker similar to the control pellet differentiated for 20 days. Finally, the enhanced clinical efficacy of the hyaline-like cartilage pellet was confirmed using an osteochondral defect rat model, with the repaired tissue resembling hyaline cartilage rather than fibrous cartilage after 8 weeks of regeneration.
Collapse
Affiliation(s)
- Seong Jin Lee
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea
| | - Yoojun Nam
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Yeri Alice Rim
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Kijun Lee
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Ji Hyeon Ju
- Catholic iPSC Research Center, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, Institute of Medical Science, College of Medicine, The Catholic University of Korea, 20 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Dong Sung Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea.,Department of Chemical Engineering, Pohang University of Science and Technology (POSTECH), 77, Cheongam-ro, Nam-gu, Pohang, Gyeongbuk 37673, Republic of Korea.,Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50, Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
37
|
Aldrich ED, Cui X, Murphy CA, Lim KS, Hooper GJ, McIlwraith CW, Woodfield TBF. Allogeneic mesenchymal stromal cells for cartilage regeneration: A review of in vitro evaluation, clinical experience, and translational opportunities. Stem Cells Transl Med 2021; 10:1500-1515. [PMID: 34387402 PMCID: PMC8550704 DOI: 10.1002/sctm.20-0552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 05/19/2021] [Accepted: 06/06/2021] [Indexed: 12/14/2022] Open
Abstract
The paracrine signaling, immunogenic properties and possible applications of mesenchymal stromal cells (MSCs) for cartilage tissue engineering and regenerative medicine therapies have been investigated through numerous in vitro, animal model and clinical studies. The emerging knowledge largely supports the concept of MSCs as signaling and modulatory cells, exerting their influence through trophic and immune mediation rather than as a cell replacement therapy. The virtues of allogeneic cells as a ready‐to‐use product with well‐defined characteristics of cell surface marker expression, proliferative ability, and differentiation capacity are well established. With clinical applications in mind, a greater focus on allogeneic cell sources is evident, and this review summarizes the latest published and upcoming clinical trials focused on cartilage regeneration adopting allogeneic and autologous cell sources. Moreover, we review the current understanding of immune modulatory mechanisms and the role of trophic factors in articular chondrocyte‐MSC interactions that offer feasible targets for evaluating MSC activity in vivo within the intra‐articular environment. Furthermore, bringing labeling and tracking techniques to the clinical setting, while inherently challenging, will be extremely informative as clinicians and researchers seek to bolster the case for the safety and efficacy of allogeneic MSCs. We therefore review multiple promising approaches for cell tracking and labeling, including both chimerism studies and imaging‐based techniques, that have been widely explored in vitro and in animal models. Understanding the distribution and persistence of transplanted MSCs is necessary to fully realize their potential in cartilage regeneration techniques and tissue engineering applications.
Collapse
Affiliation(s)
- Ellison D Aldrich
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand.,School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Xiaolin Cui
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| | - Caroline A Murphy
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| | - Khoon S Lim
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| | - Gary J Hooper
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| | - C Wayne McIlwraith
- Orthopedic Research Center, C. Wayne McIlwraith Translational Medicine Institute, Colorado State University, Fort Collins, Colorado, USA
| | - Tim B F Woodfield
- Christchurch Regenerative Medicine and Tissue Engineering (CReaTE) Group, Department of Orthopedic Surgery & Musculoskeletal Medicine, University of Otago, Christchurch, New Zealand
| |
Collapse
|
38
|
Ren L, Cong N, Han H, Zhang Z, Deng C, Zhang N, Li D. The effect of sodium metasilicate on the three-dimensional chondrogenesis of mesenchymal stem cells. Dent Mater J 2021; 40:853-862. [PMID: 34193723 DOI: 10.4012/dmj.2020-214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The benefits of different silicic concentrations on chondrogenesis of mesenchymal stem cell (MSC) are unclear. Here an in vitro scaffoldless model was used to determine the impact of different silicic concentrations on the three-dimensional chondrogenesis of MSCs. Sodium metasilicate solutions were used as the source of silica, and were added in the chondrogenic medium and replenished every 3 days. The thickness and area of cartilage; the expression of collagen II, aggrecan, and the collagen type II/I ratio; the glycosaminoglycan and cell contents; and the tangent modulus of the constructs were all significantly higher in 100 and 200 ng/mL groups compared with those in 0 and 10 ng/mL groups. All the above parameters, as well as several mechanical parameters of cartilage constructs were highest in 200 ng/mL group. Thus, 200 ng/mL sodium metasilicate could promote the chondrogenic differentiation of MSCs and the mechanical and biochemical properties of the cartilage constructs.
Collapse
Affiliation(s)
- Le Ren
- Department of Oral, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Nuonuo Cong
- Department of Stomatology, Beijing Friendship Hospital, Capital Medical University
| | - Hao Han
- Medical Emergency Center, Xi'an Xiangyang International Airport
| | - Zhe Zhang
- Department of Oral, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Chunni Deng
- Department of Oral, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Nan Zhang
- Department of Oral, The First Affiliated Hospital of Xi'an Jiaotong University
| | - Daxu Li
- Department of Oral, The First Affiliated Hospital of Xi'an Jiaotong University
| |
Collapse
|
39
|
Zhao X, Hu DA, Wu D, He F, Wang H, Huang L, Shi D, Liu Q, Ni N, Pakvasa M, Zhang Y, Fu K, Qin KH, Li AJ, Hagag O, Wang EJ, Sabharwal M, Wagstaff W, Reid RR, Lee MJ, Wolf JM, El Dafrawy M, Hynes K, Strelzow J, Ho SH, He TC, Athiviraham A. Applications of Biocompatible Scaffold Materials in Stem Cell-Based Cartilage Tissue Engineering. Front Bioeng Biotechnol 2021; 9:603444. [PMID: 33842441 PMCID: PMC8026885 DOI: 10.3389/fbioe.2021.603444] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 02/08/2021] [Indexed: 12/16/2022] Open
Abstract
Cartilage, especially articular cartilage, is a unique connective tissue consisting of chondrocytes and cartilage matrix that covers the surface of joints. It plays a critical role in maintaining joint durability and mobility by providing nearly frictionless articulation for mechanical load transmission between joints. Damage to the articular cartilage frequently results from sport-related injuries, systemic diseases, degeneration, trauma, or tumors. Failure to treat impaired cartilage may lead to osteoarthritis, affecting more than 25% of the adult population globally. Articular cartilage has a very low intrinsic self-repair capacity due to the limited proliferative ability of adult chondrocytes, lack of vascularization and innervation, slow matrix turnover, and low supply of progenitor cells. Furthermore, articular chondrocytes are encapsulated in low-nutrient, low-oxygen environment. While cartilage restoration techniques such as osteochondral transplantation, autologous chondrocyte implantation (ACI), and microfracture have been used to repair certain cartilage defects, the clinical outcomes are often mixed and undesirable. Cartilage tissue engineering (CTE) may hold promise to facilitate cartilage repair. Ideally, the prerequisites for successful CTE should include the use of effective chondrogenic factors, an ample supply of chondrogenic progenitors, and the employment of cell-friendly, biocompatible scaffold materials. Significant progress has been made on the above three fronts in past decade, which has been further facilitated by the advent of 3D bio-printing. In this review, we briefly discuss potential sources of chondrogenic progenitors. We then primarily focus on currently available chondrocyte-friendly scaffold materials, along with 3D bioprinting techniques, for their potential roles in effective CTE. It is hoped that this review will serve as a primer to bring cartilage biologists, synthetic chemists, biomechanical engineers, and 3D-bioprinting technologists together to expedite CTE process for eventual clinical applications.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Daniel A. Hu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Di Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Fang He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hao Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Linjuan Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Nephrology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Deyao Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Orthopaedic Surgery, Union Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Spine Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Na Ni
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Ministry of Education Key Laboratory of Diagnostic Medicine, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Mikhail Pakvasa
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Yongtao Zhang
- Department of Orthopaedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Kai Fu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Departments of Neurosurgery, The Affiliated Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kevin H. Qin
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Alexander J. Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Ofir Hagag
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Eric J. Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Maya Sabharwal
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
- Department of Surgery, Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL, United States
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Jennifer Moriatis Wolf
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Mostafa El Dafrawy
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Kelly Hynes
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Sherwin H. Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| | - Aravind Athiviraham
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, United States
| |
Collapse
|
40
|
Lam ATL, Lee AP, Jayaraman P, Tan KY, Raghothaman D, Lim HL, Cheng H, Zhou L, Tan AHM, Reuveny S, Oh S. Multiomics analyses of cytokines, genes, miRNA, and regulatory networks in human mesenchymal stem cells expanded in stirred microcarrier-spinner cultures. Stem Cell Res 2021; 53:102272. [PMID: 33676128 DOI: 10.1016/j.scr.2021.102272] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/19/2021] [Accepted: 02/21/2021] [Indexed: 01/09/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are of great clinical interest as a form of allogenic therapy due to their excellent regenerative and immunomodulatory effects for various therapeutic indications. Stirred suspension bioreactors using microcarriers (MC) have been used for large-scale production of MSCs compared to planar cultivation systems. Previously, we have demonstrated that expansion of MSCs in MC-spinner cultures improved chondrogenic, osteogenic, and cell migration potentials as compared to monolayer-static cultures. In this study, we sought to address this by analyzing global gene expression patterns, miRNA profiles and secretome under both monolayer-static and MC-spinner cultures in serum-free medium at different growth phases. The datasets revealed differential expression patterns that correlated with potentially improved MSC properties in cells from MC-spinner cultures compared to those of monolayer-static cultures. Transcriptome analysis identified a unique expression signature for cells from MC-spinner cultures, which correlated well with miRNA expression, and cytokine secretion involved in key MSC functions. Importantly, MC-spinner cultures and conditioned medium showed increased expression of factors that possibly enhance pathways of extracellular matrix dynamics, cellular metabolism, differentiation potential, immunoregulatory function, and wound healing. This systematic analysis provides insights for the efficient optimization of stem cell bioprocessing and infers that MC-based bioprocess manufacturing could improve post-expansion cellular properties for stem cell therapies.
Collapse
Affiliation(s)
- Alan Tin-Lun Lam
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Alison P Lee
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Premkumar Jayaraman
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Kah Yong Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Deepak Raghothaman
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Hsueh Lee Lim
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - He Cheng
- MiRXES, 2 Tukang Innovation Grove, JTC MedTech Hub, Singapore
| | - Lihan Zhou
- MiRXES, 2 Tukang Innovation Grove, JTC MedTech Hub, Singapore
| | - Andy Hee-Meng Tan
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shaul Reuveny
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Steve Oh
- Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore.
| |
Collapse
|
41
|
Wong KU, Zhang A, Akhavan B, Bilek MM, Yeo GC. Biomimetic Culture Strategies for the Clinical Expansion of Mesenchymal Stromal Cells. ACS Biomater Sci Eng 2021. [PMID: 33599471 DOI: 10.1021/acsbiomaterials.0c01538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mesenchymal stromal/stem cells (MSCs) typically require significant ex vivo expansion to achieve the high cell numbers required for research and clinical applications. However, conventional MSC culture on planar (2D) plastic surfaces has been shown to induce MSC senescence and decrease cell functionality over long-term proliferation, and usually, it has a high labor requirement, a high usage of reagents, and therefore, a high cost. In this Review, we describe current MSC-based therapeutic strategies and outline the important factors that need to be considered when developing next-generation cell expansion platforms. To retain the functional value of expanded MSCs, ex vivo culture systems should ideally recapitulate the components of the native stem cell microenvironment, which include soluble cues, resident cells, and the extracellular matrix substrate. We review the interplay between these stem cell niche components and their biological roles in governing MSC phenotype and functionality. We discuss current biomimetic strategies of incorporating biochemical and biophysical cues in MSC culture platforms to grow clinically relevant cell numbers while preserving cell potency and stemness. This Review summarizes the current state of MSC expansion technologies and the challenges that still need to be overcome for MSC clinical applications to be feasible and sustainable.
Collapse
Affiliation(s)
- Kuan Un Wong
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Anyu Zhang
- School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Behnam Akhavan
- School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Marcela M Bilek
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Biomedical Engineering, The University of Sydney, Sydney, New South Wales 2006, Australia.,The University of Sydney Nano Institute, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Giselle C Yeo
- Charles Perkins Center, The University of Sydney, Sydney, New South Wales 2006, Australia.,School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
42
|
Futrega K, Music E, Robey PG, Gronthos S, Crawford R, Saifzadeh S, Klein TJ, Doran MR. Characterisation of ovine bone marrow-derived stromal cells (oBMSC) and evaluation of chondrogenically induced micro-pellets for cartilage tissue repair in vivo. Stem Cell Res Ther 2021; 12:26. [PMID: 33413652 PMCID: PMC7791713 DOI: 10.1186/s13287-020-02045-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract Bone marrow stromal cells (BMSC) show promise in cartilage repair, and sheep are the most common large animal pre-clinical model. Objective The objective of this study was to characterise ovine BMSC (oBMSC) in vitro, and to evaluate the capacity of chondrogenic micro-pellets manufactured from oBMSC or ovine articular chondrocytes (oACh) to repair osteochondral defects in sheep. Design oBMSC were characterised for surface marker expression using flow cytometry and evaluated for tri-lineage differentiation capacity. oBMSC micro-pellets were manufactured in a microwell platform, and chondrogenesis was compared at 2%, 5%, and 20% O2. The capacity of cartilage micro-pellets manufactured from oBMSC or oACh to repair osteochondral defects in adult sheep was evaluated in an 8-week pilot study. Results Expanded oBMSC were positive for CD44 and CD146 and negative for CD45. The common adipogenic induction ingredient, 3-Isobutyl-1-methylxanthine (IBMX), was toxic to oBMSC, but adipogenesis could be restored by excluding IBMX from the medium. BMSC chondrogenesis was optimal in a 2% O2 atmosphere. Micro-pellets formed from oBMSC or oACh appeared morphologically similar, but hypertrophic genes were elevated in oBMSC micro-pellets. While oACh micro-pellets formed cartilage-like repair tissue in sheep, oBMSC micro-pellets did not. Conclusion The sensitivity of oBMSC, compared to human BMSC, to IBMX in standard adipogenic assays highlights species-associated differences. Micro-pellets manufactured from oACh were more effective than micro-pellets manufactured from oBMSC in the repair of osteochondral defects in sheep. While oBMSC can be driven to form cartilage-like tissue in vitro, the effective use of these cells in cartilage repair will depend on the successful mitigation of hypertrophy and tissue integration. Supplementary information The online version contains supplementary material available at 10.1186/s13287-020-02045-3.
Collapse
Affiliation(s)
- K Futrega
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia.,National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA.,Translational Research Institute (TRI), Brisbane, Queensland, Australia
| | - E Music
- Translational Research Institute (TRI), Brisbane, Queensland, Australia.,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - P G Robey
- National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - S Gronthos
- Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - R Crawford
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - S Saifzadeh
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - T J Klein
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia
| | - M R Doran
- Centre for Biomedical Technologies (CBT), Queensland University of Technology (QUT), Brisbane, Queensland, Australia. .,National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, Maryland, USA. .,Translational Research Institute (TRI), Brisbane, Queensland, Australia. .,School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, Queensland, Australia. .,Mater Research Institute - University of Queensland (UQ), Translational Research Institute (TRI), Brisbane, Queensland, Australia.
| |
Collapse
|
43
|
Microparticles from glycidylmethacrylated gelatin as cell carriers prepared in an aqueous two-phase system. Eur Polym J 2021. [DOI: 10.1016/j.eurpolymj.2020.110148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
44
|
Wasyłeczko M, Sikorska W, Chwojnowski A. Review of Synthetic and Hybrid Scaffolds in Cartilage Tissue Engineering. MEMBRANES 2020; 10:E348. [PMID: 33212901 PMCID: PMC7698415 DOI: 10.3390/membranes10110348] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023]
Abstract
Cartilage tissue is under extensive investigation in tissue engineering and regenerative medicine studies because of its limited regenerative potential. Currently, many scaffolds are undergoing scientific and clinical research. A key for appropriate scaffolding is the assurance of a temporary cellular environment that allows the cells to function as in native tissue. These scaffolds should meet the relevant requirements, including appropriate architecture and physicochemical and biological properties. This is necessary for proper cell growth, which is associated with the adequate regeneration of cartilage. This paper presents a review of the development of scaffolds from synthetic polymers and hybrid materials employed for the engineering of cartilage tissue and regenerative medicine. Initially, general information on articular cartilage and an overview of the clinical strategies for the treatment of cartilage defects are presented. Then, the requirements for scaffolds in regenerative medicine, materials intended for membranes, and methods for obtaining them are briefly described. We also describe the hybrid materials that combine the advantages of both synthetic and natural polymers, which provide better properties for the scaffold. The last part of the article is focused on scaffolds in cartilage tissue engineering that have been confirmed by undergoing preclinical and clinical tests.
Collapse
Affiliation(s)
- Monika Wasyłeczko
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Trojdena 4 str., 02-109 Warsaw, Poland; (W.S.); (A.C.)
| | | | | |
Collapse
|
45
|
Oliver JD, Jia S, Halpern LR, Graham EM, Turner EC, Colombo JS, Grainger DW, D'Souza RN. Innovative Molecular and Cellular Therapeutics in Cleft Palate Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2020; 27:215-237. [PMID: 32873216 DOI: 10.1089/ten.teb.2020.0181] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Clefts of the lip and/or palate are the most prevalent orofacial birth defects occurring in about 1:700 live human births worldwide. Early postnatal surgical interventions are extensive and staged to bring about optimal growth and fusion of palatal shelves. Severe cleft defects pose a challenge to correct with surgery alone, resulting in complications and sequelae requiring life-long, multidisciplinary care. Advances made in materials science innovation, including scaffold-based delivery systems for precision tissue engineering, now offer new avenues for stimulating bone formation at the site of surgical correction for palatal clefts. In this study, we review the present scientific literature on key developmental events that can go awry in palate development and the common surgical practices and challenges faced in correcting cleft defects. How key osteoinductive pathways implicated in palatogenesis inform the design and optimization of constructs for cleft palate correction is discussed within the context of translation to humans. Finally, we highlight new osteogenic agents and innovative delivery systems with the potential to be adopted in engineering-based therapeutic approaches for the correction of palatal defects. Impact statement Tissue-engineered scaffolds supplemented with osteogenic growth factors have attractive, largely unexplored possibilities to modulate molecular signaling networks relevant to driving palatogenesis in the context of congenital anomalies (e.g., cleft palate). Constructs that address this need may obviate current use of autologous bone grafts, thereby avoiding donor-site morbidity and other regenerative challenges in patients afflicted with palatal clefts. Combinations of biomaterials and drug delivery of diverse regenerative cues and biologics are currently transforming strategies exploited by engineers, scientists, and clinicians for palatal cleft repair.
Collapse
Affiliation(s)
- Jeremie D Oliver
- School of Dentistry, University of Utah Health Sciences, Salt Lake City, Utah, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Shihai Jia
- School of Dentistry, University of Utah Health Sciences, Salt Lake City, Utah, USA
| | - Leslie R Halpern
- School of Dentistry, University of Utah Health Sciences, Salt Lake City, Utah, USA
| | - Emily M Graham
- School of Medicine, University of Utah Health Sciences, Salt Lake City, Utah, USA
| | - Emma C Turner
- University of Western Australia Dental School, Perth, Western Australia
| | - John S Colombo
- University of Las Vegas at Nevada School of Dental Medicine, Las Vegas, Nevada, USA
| | - David W Grainger
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA.,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah Health Sciences, Salt Lake City, Utah, USA
| | - Rena N D'Souza
- School of Dentistry, University of Utah Health Sciences, Salt Lake City, Utah, USA.,Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA.,School of Medicine, University of Utah Health Sciences, Salt Lake City, Utah, USA
| |
Collapse
|
46
|
Cartilage repair using stem cells & biomaterials: advancement from bench to bedside. Mol Biol Rep 2020; 47:8007-8021. [PMID: 32888123 DOI: 10.1007/s11033-020-05748-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 08/28/2020] [Indexed: 10/23/2022]
Abstract
Osteoarthritis (OA) involves gradual destruction of articular cartilagemanifested by pain, stiffness of joints, and impaired movement especially in knees and hips. Non-vascularity of this tissue hinders its self-regenerative capacity and thus, the application of reparative or restorative modalities becomes imperative in OA treatment. In recent years, stem cell-based therapies have been explored as potential modalities for addressing OA complications. While mesenchymal stem cells (MSCs) hold immense promise, the recapitulation of native articular cartilage usingMSCs remains elusive. In this review, we have highlighted the chondrogenic potential of MSCs, factors guiding in vitro chondrogenic differentiation, biomaterials available for cartilage repair, their current market status, and the outcomes of major clinical trials. Our search on ClinicalTrials.gov using terms "stem cell" and "osteoarthritis" yielded 83 results. An analysis of the 29 trials that have been completed revealed differences in source of MSCs (bone marrow, adipose tissue, umbilical cord etc.), cell type (autologous or allogenic), and dose administered. Moreover, only 02 out of 29 studies have reported the use of matrix for cartilage repair. From future perspective, aconsensus on choice of cells, differentiation inducers, biomaterials, and clinical settings might pave a way for concocting robust strategies to improve the clinical applicability of biomimetic neocartilage constructs.
Collapse
|
47
|
Shelat R, Bhatt LK, Paunipagar B, Kurian T, Khanna A, Chandra S. Regeneration of hyaline cartilage in osteochondral lesion model using L-lysine magnetic nanoparticles labeled mesenchymal stem cells and their in vivo imaging. J Tissue Eng Regen Med 2020; 14:1604-1617. [PMID: 32840054 DOI: 10.1002/term.3120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 12/16/2022]
Abstract
Treatment of osteochondral defects continues to pose a major challenge for patients and orthopedic surgeons due to the limited healing potential of articular cartilage. Mesenchymal stem cells (MSCs) possess therapeutic potential for the treatment of osteochondral pain and pathology. However, it is necessary to use proper labeling and imaging agent of stem cells that can decipher its role posttransplantation. A major limitation of routinely used contrast agents is signal dilution over a period of time which limits its use for further studies. At the same time, regeneration of fibrocartilage over native hyaline cartilage also limits the use of conventional therapies. The present study evaluates the efficacy of bone marrow-derived mesenchymal stem cells (BMSCs) for the treatment of osteochondral defect in rats with the regeneration of hyaline cartilage in situ and in vivo monitoring of the stem cells using L-lysine functionalized magnetic iron oxide nanoparticles (lys-IONPs). L-lysine stabilizes the iron oxide nanoparticles, enhances the biocompatibility, and provides functionalities for efficient stem cell labeling. in vitro toxic effects of lys-IONPs on mitochondrial impairment, morphological alterations, and actin cytoskeleton reveal minimum damage to BM-MSCs. Histological data (H and E, Masson's trichrome and immunohistochemistry) describe the early initiation of healing and regeneration of hyaline-like cartilage over fibrocartilage in stem cell treated groups. MR scans demonstrate generation of hypointense signals in lys-IONPs-BMSCs with improved signal intensity and minimum loss over 28 days revealing its use as a long-term stem cell labeling and imaging agent.
Collapse
Affiliation(s)
- Ruchita Shelat
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University, Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | | | | | - Aparna Khanna
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University, Mumbai, India.,Computational Biology and Translational Research, Amity University, Mumbai, India
| | - Sudeshna Chandra
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University, Mumbai, India
| |
Collapse
|
48
|
Dual delivery of stem cells and insulin-like growth factor-1 in coacervate-embedded composite hydrogels for enhanced cartilage regeneration in osteochondral defects. J Control Release 2020; 327:284-295. [PMID: 32763434 DOI: 10.1016/j.jconrel.2020.08.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/07/2020] [Accepted: 08/02/2020] [Indexed: 12/31/2022]
Abstract
Exogenous dual delivery of progenitor cell population and therapeutic growth factors (GFs) is one of alternative tissue engineering strategies for osteochondral tissue regeneration. In the present study, an implantable dual delivery platform was developed using coacervates (Coa) (i.e., a tertiary complex of poly(ethylene argininylaspartate diglyceride) (PEAD) polycation, heparin, and cargo insulin-like growth factor-1 (IGF-1), in thiolated gelatin (gelatin-SH)/ poly(ethylene glycol) diacrylate (PEGDA) interpenetrating network (IPN) hydrogels. Since Coa is able to protect cargo GF and maintain its long-term bioactivity, it is speculated that Coa-mediated delivery of chondrogenic factor IGF-1 with the aid of adipose-derived stem cells (ADSCs) would synergistically facilitate osteochondral tissue repair during physiological regeneration process. Our results indicate that gelatin-SH/PEGDA IPN hydrogels demonstrated biocompatibility and mechanical properties for a possible long-term transplantation, and PEAD-base Coa exhibited a sustained release of bioactive IGF-1 over 3 weeks. Subsequently, released IGF-1 from Coa could effectively induce chondrogenic differentiation of embedded ADSCs in the hydrogel, by showing enhanced glycosaminoglycan deposition and expression of chondrogenesis-associated genes. More importantly, at 12 weeks post-implantation in a rabbit full thickness osteochondral defect model, the quality of regenerative tissues in both chondral and subchondral layers was significantly improved in dual delivery of ADSC and IGF-1 in Coa encapsulated in gelatin-SH/PEGDA IPN hydrogels, as compared with a single delivery of ADSC only and a dual delivery without Coa. Therefore, we conclude that our Coa-embedded composite hydrogel platform could effectively augment osteochondral tissue regeneration holds promise for a feasible osteoarthritis therapeutic application.
Collapse
|
49
|
Debnath UK. Mesenchymal Stem Cell Therapy in Chondral Defects of Knee: Current Concept Review. Indian J Orthop 2020; 54:1-9. [PMID: 32952903 PMCID: PMC7474009 DOI: 10.1007/s43465-020-00198-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE Full-thickness cartilage defects if left alone would increase the risk of osteoarthritis (OA) with severe associated pain and functional disability. Articular cartilage defect may result from direct trauma or chronic degeneration. The capability of the mesenchymal stem cells (MSCs) to repair and regenerate cartilage has been widely investigated. This review describes current trends in MSC biology, the sourcing, expansion, application and role of MSCs in chondral defects of human knees. METHODS The studies referencing MSCs and knee osteoarthritis were searched (from1998 to 2020) using PubMed, EMBASE, Cochrane Library, Web of Science and the ClinicalTrials.gov with keywords (MSCs, chondral defects or cartilage degeneration of knee, cartilage regeneration, chondrogenesis, tissue engineering, efficacy and safety). The inclusion criteria were based on use of MSCs for treatment of chondral defects and osteoarthritis of the knee, English language and human studies. RESULTS The history of MSC research from the initial discovery of their multipotency to the more recent recognition of their role in cartilage defects of knee is elucidated. Several studies have demonstrated promising results in the clinical application for repair of chondral defects as an adjuvant or independent procedure. Intra-articular MSCs provide improvements in pain and function in knee osteoarthritis at short-term follow-up in many studies. The tendency of MSCs to differentiate into fibrocartilage affecting the outcome is a common issue faced by researchers. CONCLUSION Some efficacy has been shown of MSCs for cartilage repair in osteoarthritis; however, the evidence of efficacy of intra-articular MSCs on both clinical outcomes and cartilage repair remains limited. Despite the high quality of evidence to support, MSC therapy has emerged but further refinement of methodology will be necessary to support its routine clinical use.
Collapse
|
50
|
Kim SA, Sur YJ, Cho ML, Go EJ, Kim YH, Shetty AA, Kim SJ. Atelocollagen promotes chondrogenic differentiation of human adipose-derived mesenchymal stem cells. Sci Rep 2020; 10:10678. [PMID: 32606308 PMCID: PMC7327030 DOI: 10.1038/s41598-020-67836-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 06/16/2020] [Indexed: 12/19/2022] Open
Abstract
Effective engineering approaches for cartilage regeneration involve a combination of cells and biomaterial scaffolds. Multipotent mesenchymal stem cells (MSCs) are important sources for cartilage regeneration. Atelocollagen provides a suitable substrate for MSC attachment and enhancing chondrogenic differentiation. Here, we assessed the chondrogenic potential of adipose tissue derived human MSCs (hMSCs) mixed with atelocollagen gel. We observed cell attachment, viability, and microstructures by electron microscopy over 21 days. The levels of Sox9, type II collagen, aggrecan, type I collagen, Runx2, type X collagen, ALP, Osterix, and MMP13 were measured by RT-qPCR. Cartilage matrix-related proteins were assessed by enzyme-linked immunosorbent assay (ELISA), histology, and immunohistochemistry. hMSCs of all groups exhibited well-maintained cell survival, distribution and morphology. Abundant type II collagen fibers developed on day 21; while Sox9, type II collagen, and aggrecan expression increased over time in the atelocollagen group. However, type I collagen, RUNX2, type X collagen (CoL10A1), Osterix, and ALP were not expressed. These results corroborated the protein expression detected by ELISA. Further, histological analysis revealed lacunae-like structures, while staining demonstrated glycosaminoglycan accumulation. Cumulatively, these results indicate that atelocollagen scaffolds improve hMSC chondrogenic differentiation and are a potential approach for cartilage regeneration.
Collapse
Affiliation(s)
- Seon Ae Kim
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoo Joon Sur
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Eun Jeong Go
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yun Hwan Kim
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Asode Ananthram Shetty
- The Institute of Medical Sciences, Faculty of Health and Wellbeing, Canterbury Christ Church University, Kent, UK
| | - Seok Jung Kim
- Department of Orthopedic Surgery, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|