1
|
Chou LS, Zhang J, Jildeh TR. Metabolic Functions of the Infrapatellar Fat Pad: Implications for Knee Health and Pathology. JBJS Rev 2024; 12:01874474-202410000-00001. [PMID: 39361777 DOI: 10.2106/jbjs.rvw.24.00110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
» Despite being historically viewed as a vestigial structure, the infrapatellar fat pad (IPFP) is now recognized as a metabolically active structure, influencing knee health through cytokine production and metabolic pathways.» With distinct anatomical regions, the IPFP contains diverse cell types including adipocytes, fibroblasts, and immune cells, influencing its functional roles, pathology, and contributions to knee disorders.» The IPFP acts as an endocrine organ by releasing adipokines such as adiponectin, leptin, and tumor necrosis factor α, regulating energy balance, immune responses, and tissue remodelling, with implications for knee joint health.» The IPFP's metabolic interactions with neighboring tissues influence joint health, clinical conditions such as knee pain, osteoarthritis, postoperative complications, and ganglion cysts, highlighting its therapeutic potential and clinical relevance.» Understanding the multifaceted metabolic role of the IPFP opens avenues for collaborative approaches that integrate orthopaedics, endocrinology, and immunology to develop innovative therapeutic strategies targeting the intricate connections between adipokines, joint health, and immune responses.
Collapse
Affiliation(s)
- Lee S Chou
- Department of Orthopaedic Surgery, Michigan State University, East Lansing, Michigan
| | | | | |
Collapse
|
2
|
El Masri J, Fadlallah H, Al Sabsabi R, Afyouni A, Al-Sayegh M, Abou-Kheir W. Adipose-Derived Stem Cell Therapy in Spinal Cord Injury. Cells 2024; 13:1505. [PMID: 39273075 PMCID: PMC11394073 DOI: 10.3390/cells13171505] [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: 07/25/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Spinal cord injury (SCI) is a serious condition accompanied by severe adverse events that affect several aspects of the patient's life, such as motor, sensory, and functional impairment. Despite its severe consequences, definitive treatment for these injuries is still missing. Therefore, researchers have focused on developing treatment strategies aimed at ensuring full recovery post-SCI. Accordingly, attention has been drawn toward cellular therapy using mesenchymal stem cells. Considering their wide availability, decreased immunogenicity, wide expansion capacity, and impressive effectiveness in many therapeutic approaches, adipose-derived stem cell (ADSC) injections in SCI cases have been investigated and showed promising results. In this review, SCI pathophysiology and ADSC transplantation benefits are discussed independently, together with SCI animal models and adipose stem cell preparation and application techniques. The mechanisms of healing in an SCI post-ADSC injection, the outcomes of this therapeutic approach, and current clinical trials are also deliberated, in addition to the challenges and future perspectives, aiming to encourage further research in this field.
Collapse
Affiliation(s)
- Jad El Masri
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
- Faculty of Medical Sciences, Lebanese University, Beirut 1533, Lebanon
| | - Hiba Fadlallah
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| | - Rahaf Al Sabsabi
- Faculty of Medical Sciences, Lebanese University, Beirut 1533, Lebanon
| | - Ahmad Afyouni
- Faculty of Medical Sciences, Lebanese University, Beirut 1533, Lebanon
| | - Mohamed Al-Sayegh
- Biology Division, New York University Abu Dhabi, Abu Dhabi 2460, United Arab Emirates
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon
| |
Collapse
|
3
|
Shimomura K, Ando W, Hart DA, Nakamura N. A novel scaffold-free mesenchymal stem cell-derived tissue engineered construct for articular cartilage restoration - From basic to clinic. Regen Ther 2024; 26:124-131. [PMID: 38883147 PMCID: PMC11176953 DOI: 10.1016/j.reth.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/04/2024] [Accepted: 05/19/2024] [Indexed: 06/18/2024] Open
Abstract
Treatments for articular cartilage injuries are still challenging, due in part to its avascular and aneural surroundings. Since the first report of autologous chondrocyte implantation, cell-based therapies have been extensively studied with a variety of cell sources, including chondrocytes and mesenchymal stem/stromal cells (MSCs). Recently, MSC-based therapy has received considerable research attention because of the relative ease in handling for tissue harvest, and subsequent cell expansion and differentiation. Using such cells, we have originally developed a 3-dimensional scaffold-free tissue-engineered construct (TEC) through simple-cell culture methods and demonstrated its feasibility for cartilage repair and regeneration in the first-in-human clinical trial. This review summarizes our novel scaffold-free approaches to use MSC for the restoration of damaged articular cartilage, documenting the progression from basic to clinical studies.
Collapse
Affiliation(s)
- Kazunori Shimomura
- Department of Rehabilitation, Kansai University of Welfare Sciences, Osaka, Japan
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Wataru Ando
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Orthopaedic Surgery, Kansai Rosai Hospital, Hyogo, Japan
| | - David A Hart
- McCaig Institute for Bone & Joint Health, University of Calgary, Calgary, Alberta, Canada
| | - Norimasa Nakamura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Institute for Medical Science in Sports, Osaka Health Science University, Osaka, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
| |
Collapse
|
4
|
Yin Z, Qin C, Pan S, Shi C, Wu G, Feng Y, Zhang J, Yu Z, Liang B, Gui J. Injectable hyperbranched PEG crosslinked hyaluronan hydrogel microparticles containing mir-99a-3p modified subcutaneous ADSCs-derived exosomes was beneficial for long-term treatment of osteoarthritis. Mater Today Bio 2023; 23:100813. [PMID: 37822452 PMCID: PMC10562164 DOI: 10.1016/j.mtbio.2023.100813] [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: 06/16/2023] [Revised: 08/19/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023] Open
Abstract
Exosomes (Exos) secreted by adipose-derived stem cells (ADSCs) have shown potential in alleviating osteoarthritis (OA). Previous studies indicated that infrapatellar fat pad (IPFP) derived stem cells (IPFSCs) may be more suitable for the treatment of OA than subcutaneous adipose tissue (ScAT) derived stem cells (ScASCs). However, it remains unclear which type of Exos offers superior therapeutic benefit for OA. This study first compared the differences between Exos derived from IPFP stem cells (ExosIPFP) and ScAT stem cells (ExosScAT) in OA treatment. Results suggested that ExosIPFP significantly inhibit the degradation of cartilage extracellular matrix (ECM) than ExosScAT, following this, the differences in microRNA (miRNA) expression between the two types of Exos using small RNA sequencing were performed. Subsequently, miR-99 b-3p was chosen and over-expressed in ExosScAT (ExosScAT-99b-3p), both in vivo and in vitro experiments demonstrated its efficacy in inhibiting the expression of ADAMTS4, promoting the repair of the ECM in OA. Finally, microfluidic technology was performed to fabricate a hyaluronan-based hydrogel microparticles (HMPs) for encapsulating Exos (HMPs@exos), the injectability, sustained release of Exos and long-term therapeutic effect on OA were validated. In summary, these results suggest miR-99 b-3p regulates the degradation of cartilage ECM by targeting ADAMTS4, the upregulation of miR-99 b-3p in ExosScAT would enable them to exhibit comparable or even superior effectiveness to ExosIPFP for OA treatment, making it a promising approach for OA treatment. Considering the abundant resources of ScAT and the limited availability of IPFP, ScAT harvested through liposuction could be genetically engineered to yield Exos for OA treatment. Furthermore, the encapsulation of Exos in HMPs provides an injectable sustained local drug release system, which could potentially enhance the efficacy of Exos and hold potential as future therapeutic strategies.
Collapse
Affiliation(s)
- Zhaowei Yin
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Chaoren Qin
- Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Shaowei Pan
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Chen Shi
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Guanfu Wu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing 211816, PR China
| | - Yan Feng
- Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jing Zhang
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing 211816, PR China
| | - Ziyi Yu
- State Key Laboratory of Materials-Oriented Chemical Engineering, College of Chemical Engineering, Nanjing Tech University, 30 Puzhu South Road, Nanjing 211816, PR China
| | - Bin Liang
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jianchao Gui
- Sports Medicine and Joint Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
5
|
Xiao J, Gong X, Fu Z, Song X, Ma Q, Miao J, Cai R, Yan Z, Wang S, Li Q, Chen Y, Yang L, Bian X, Chen Y. The influence of inflammation on the characteristics of adipose-derived mesenchymal stem cells (ADMSCs) and tissue repair capability in a hepatic injury mouse model. Stem Cell Res Ther 2023; 14:334. [PMID: 37981679 PMCID: PMC10659042 DOI: 10.1186/s13287-023-03532-z] [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: 04/18/2023] [Accepted: 10/10/2023] [Indexed: 11/21/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) are adult stem cells with self-renewal and multi-directional differentiation potential and possess the functions of immunomodulation, regulation of cell growth, and repair of damage. Over recent years, MSCs have been found to regulate the secretion of inflammatory factors and to exert regulatory effects on various lymphocytes in inflammatory states, and on the subsequent repair of tissue damage caused by inflammation. In the present study, we analyzed the effects of tissue inflammation on the characteristics of MSCs. METHODS Human fat derived from the infrapatellar fat pad (IPFP) of knees with differing degrees of inflammation was extracted from specimens derived from total knee arthroplasties. HE and immunohistochemical staining was performed to directly observe the evidence and degree of inflammation in human infrapatellar fat pad tissue in order to classify MSCs cells, by their origin, into highly inflamed and lowly inflamed groups, and to study the effect of tissue inflammation on cell acquisition rates via cellular counting data. Flow cytometry assays were performed to investigate the effect of tissue inflammation on MSC surface marker expression. Trilineage differentiation, including osteogenesis, adipogenesis, and chondrogenesis, was performed to assess the effect of tissue inflammation on the ability of MSCs to undergo directed differentiation. The effect of tissue inflammation on the ability of MSCs to proliferate was investigated via clone formation studies. RNA-sequencing was performed to evaluate the transcriptomes of MSCs derived from different areas of inflammation. The effect of tissue inflammation on tissue repair capacity and safety of MSCs was investigated via a murine model of acute liver injury. RESULTS The results of cell count data indicate that a high degree of tissue inflammation significantly decreases the acquisition rate of MSCs, and the proportion of CD34+ and CD146+ cells. The results of our trilineage differentiation assay show that a higher degree of inflammation decreases osteogenic differentiation and enhances adipogenic and chondrogenic differentiation of MSCs. However, these differences were not statistically significant. Clone formation assays indicate that the degree of tissue inflammation at the MSC source does not significantly affect the proliferative capacity of MSCs. The transcriptomes of MSCs remain relatively stable in fat pad tissues derived from both highly and lowly inflamed samples. The results of acute liver injury investigations in mice indicate that MSCs of high and low inflammatory tissue origin have no significant difference in their tissue repair capability. CONCLUSIONS High tissue inflammation at the source of MSCs reduces the acquisition rate of MSCs and the percentage of CD34+ and CD146+ cells acquisition. However, source tissue inflammation may not significantly affect trilineage differentiation potential and proliferative capacity of MSCs. Also, MSCs obtained from differing source degrees of inflammation retain stable and similar transcriptomic profile and are both safe and efficacious for tissue repair/regeneration without detectable differences.
Collapse
Affiliation(s)
- Jingfang Xiao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Xiaoyuan Gong
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Zhenlan Fu
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Xiongbo Song
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Qinghua Ma
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Jingya Miao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Ruili Cai
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Zexuan Yan
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Shuai Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Qian Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China
| | - Yaokai Chen
- Biobank and Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, People's Republic of China
| | - Liu Yang
- Center for Joint Surgery, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China.
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, Chongqing, People's Republic of China.
| | - Yemiao Chen
- Biobank and Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, People's Republic of China.
| |
Collapse
|
6
|
Xu X, Xu L, Xia J, Wen C, Liang Y, Zhang Y. Harnessing knee joint resident mesenchymal stem cells in cartilage tissue engineering. Acta Biomater 2023; 168:372-387. [PMID: 37481194 DOI: 10.1016/j.actbio.2023.07.024] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/26/2023] [Accepted: 07/17/2023] [Indexed: 07/24/2023]
Abstract
Osteoarthritis (OA) is a widespread clinical disease characterized by cartilage degeneration in middle-aged and elderly people. Currently, there is no effective treatment for OA apart from total joint replacement in advanced stages. Mesenchymal stem cells (MSCs) are a type of adult stem cell with diverse differentiation capabilities and immunomodulatory potentials. MSCs are known to effectively regulate the cartilage microenvironment, promote cartilage regeneration, and alleviate OA symptoms. As a result, they are promising sources of cells for OA therapy. Recent studies have revealed the presence of resident MSCs in synovial fluid, synovial membrane, and articular cartilage, which can be collected as knee joint-derived MSCs (KJD-MSC). Several preclinical and clinical studies have demonstrated that KJD-MSCs have great potential for OA treatment, whether applied alone, in combination with biomaterials, or as exocrine MSCs. In this article, we will review the characteristics of MSCs in the joints, including their cytological characteristics, such as proliferation, cartilage differentiation, and immunomodulatory abilities, as well as the biological function of MSC exosomes. We will also discuss the use of tissue engineering in OA treatment and introduce the concept of a new generation of stem cell-based tissue engineering therapy, including the use of engineering, gene therapy, and gene editing techniques to create KJD-MSCs or KJD-MSC derivative exosomes with improved functionality and targeted delivery. These advances aim to maximize the efficiency of cartilage tissue engineering and provide new strategies to overcome the bottleneck of OA therapy. STATEMENT OF SIGNIFICANCE: This research will provide new insights into the medicinal benefit of Joint resident Mesenchymal Stem Cells (MSCs), specifically on its cartilage tissue engineering ability. Through this review, the community will further realize promoting joint resident mesenchymal stem cells, especially cartilage progenitor/MSC-like progenitor cells (CPSC), as a preventive measure against osteoarthritis and cartilage injury. People and medical institutions may also consider cartilage derived MSC as an alternative approach against cartilage degeneration. Moreover, the discussion presented in this study will convey valuable information for future research that will explore the medicinal benefits of cartilage derived MSC.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Orthopedics, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Limei Xu
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Jiang Xia
- Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Caining Wen
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China
| | - Yujie Liang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China; Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| | - Yuanmin Zhang
- Department of Joint Surgery and Sports Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272029, China.
| |
Collapse
|
7
|
Neubauer M, Kramer K, Neugebauer J, Moser L, Moser A, Dammerer D, Nehrer S. Isolation and Cultivation of Adipose-Derived Mesenchymal Stem Cells Originating from the Infrapatellar Fat Pad Differentiated with Blood Products: Method and Protocol. Methods Protoc 2022; 6:mps6010003. [PMID: 36648952 PMCID: PMC9844469 DOI: 10.3390/mps6010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) are a promising source for clinical application in regenerative orthopedics. ASCs derived from the infra-patellar fat pad (IFP)-a distinct adipose structure in the knee-show superior regenerative potential compared to subcutaneous-fat-derived cells. Furthermore, it has been shown that blood products enhance ASCs' viability. A major challenge for clinical translation of both ASCs and blood products is the low comparability of obtained data due to non-standardized harvesting, isolation and preparation methods. The aim of this method-paper is to provide reproducible protocols to help standardize basic research in the field to build a sound basis for clinical translation with an emphasize on practicability. The presented protocols include (i) ASC isolation from the IFP, (ii) blood product preparation and (iii) ASC incubation with blood products.
Collapse
Affiliation(s)
- Markus Neubauer
- Center for Regenerative Medicine and Orthopedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500 Krems, Austria
- Department of Orthopedics and Traumatology, Karl Landsteiner University of Health Sciences, University Hospital Krems, Mitterweg 10, 3500 Krems, Austria
| | - Karina Kramer
- Center for Regenerative Medicine and Orthopedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500 Krems, Austria
| | - Johannes Neugebauer
- Center for Regenerative Medicine and Orthopedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500 Krems, Austria
- Department of Orthopedics and Traumatology, Karl Landsteiner University of Health Sciences, University Hospital Krems, Mitterweg 10, 3500 Krems, Austria
| | - Lukas Moser
- Center for Regenerative Medicine and Orthopedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500 Krems, Austria
- Department of Orthopedics and Traumatology, Karl Landsteiner University of Health Sciences, University Hospital Krems, Mitterweg 10, 3500 Krems, Austria
| | - Anna Moser
- Center for Regenerative Medicine and Orthopedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500 Krems, Austria
- Department of Orthopedics and Traumatology, Karl Landsteiner University of Health Sciences, University Hospital Krems, Mitterweg 10, 3500 Krems, Austria
| | - Dietmar Dammerer
- Center for Regenerative Medicine and Orthopedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500 Krems, Austria
- Department of Orthopedics and Traumatology, Karl Landsteiner University of Health Sciences, University Hospital Krems, Mitterweg 10, 3500 Krems, Austria
| | - Stefan Nehrer
- Center for Regenerative Medicine and Orthopedics, Danube University Krems, Dr. Karl-Dorrek-Str. 30, 3500 Krems, Austria
- Department of Orthopedics and Traumatology, Karl Landsteiner University of Health Sciences, University Hospital Krems, Mitterweg 10, 3500 Krems, Austria
- Correspondence: ; Tel.: +43-2732-893-2608
| |
Collapse
|
8
|
Cankaya D, Akti S, Yasar NE, Karakus D, Unal KO, Karhan TE, Sezgin EA. Total Infrapatellar Fat Pad Excision Leads to Worse Isokinetic Performance in Total Knee Arthroplasty: A Randomized Controlled Trial. J Knee Surg 2022; 35:1544-1548. [PMID: 33792001 DOI: 10.1055/s-0041-1727114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There are concerns that total infrapatellar fat pad (IPFP) excision in total knee arthroplasty (TKA) results in patellar tendon shortening due to ischemic contracture, but individual preference of the surgeon is still the main determinant between total or partial excision. The aim of this randomized controlled study was to compare isokinetic performance and clinical outcome of TKAs with total and partial excision of the IPFP. Seventy-two patients scheduled to undergo TKA for primary knee osteoarthritis by a single surgeon were randomly assigned to either total or partial excision group. Patients were evaluated preoperatively and at postoperative 1 year, with Knee Society Score (KSS) and isokinetic measurements. The physiatrist performing isokinetic tests and patients were blinded to the study. There were no significant differences between the groups in respect of age, body mass index, gender, and preoperative KSS and isokinetic performance. Postoperatively, both groups had improved KSS knee and KSS function scores, with no difference determined. Knee extension peak torque was significantly higher postoperatively in the partial excision group at postoperative 1 year (p = 0.036). However, there were no significant differences in knee flexion peak torque following TKA (p = 0.649). The results of this study demonstrated that total excision of the IPFP during TKA is associated with worse isokinetic performance, which is most likely due to changes in the knee biomechanics with the development of patella baja. Partial excision of the IPFP appears to be a valid alternative to overcome this potential detrimental effect without impeding exposure to the lateral compartment. This is a Level I, therapeutic study.
Collapse
Affiliation(s)
- Deniz Cankaya
- Department of Orthopaedic and Traumatology, Aksaray University Education and Research Hospital, Aksaray, Turkey
| | - Sefa Akti
- Department of Orthopaedic and Traumatology, Aksaray University Education and Research Hospital, Aksaray, Turkey
| | - Niyazi Erdem Yasar
- Department of Orthopaedic and Traumatology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Dilek Karakus
- Department of Physical Medicine and Rehabilition, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Kazim Onur Unal
- Department of Orthopaedic and Traumatology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Taha Esref Karhan
- Department of Orthopaedic and Traumatology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Erdem Aras Sezgin
- Department of Orthopaedic and Traumatology, Aksaray University Education and Research Hospital, Aksaray, Turkey
| |
Collapse
|
9
|
Najafi-Ghalehlou N, Feizkhah A, Mobayen M, Pourmohammadi-Bejarpasi Z, Shekarchi S, Roushandeh AM, Roudkenar MH. Plumping up a Cushion of Human Biowaste in Regenerative Medicine: Novel Insights into a State-of-the-Art Reserve Arsenal. Stem Cell Rev Rep 2022; 18:2709-2739. [PMID: 35505177 PMCID: PMC9064122 DOI: 10.1007/s12015-022-10383-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 12/03/2022]
Abstract
Major breakthroughs and disruptive methods in disease treatment today owe their thanks to our inch by inch developing conception of the infinitive aspects of medicine since the very beginning, among which, the role of the regenerative medicine can on no account be denied, a branch of medicine dedicated to either repairing or replacing the injured or diseased cells, organs, and tissues. A novel means to accomplish such a quest is what is being called "medical biowaste", a large assortment of biological samples produced during a surgery session or as a result of physiological conditions and biological activities. The current paper accentuating several of a number of promising sources of biowaste together with their plausible applications in routine clinical practices and the confronting challenges aims at inspiring research on the existing gap between clinical and basic science to further extend our knowledge and understanding concerning the potential applications of medical biowaste.
Collapse
Affiliation(s)
- Nima Najafi-Ghalehlou
- Department of Medical Laboratory Sciences, Faculty of Paramedicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Feizkhah
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammadreza Mobayen
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Zahra Pourmohammadi-Bejarpasi
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Shima Shekarchi
- Anatomical Sciences Department, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Amaneh Mohammadi Roushandeh
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| | - Mehryar Habibi Roudkenar
- Burn and Regenerative Medicine Research Center, School of Medicine, Velayat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
- Cardiovascular Diseases Research Center, Department of Cardiology, School of Medicine, Heshmat Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
10
|
Kurenkova AD, Romanova IA, Kibirskiy PD, Timashev P, Medvedeva EV. Strategies to Convert Cells into Hyaline Cartilage: Magic Spells for Adult Stem Cells. Int J Mol Sci 2022; 23:11169. [PMID: 36232468 PMCID: PMC9570095 DOI: 10.3390/ijms231911169] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/30/2022] Open
Abstract
Damaged hyaline cartilage gradually decreases joint function and growing pain significantly reduces the quality of a patient's life. The clinically approved procedure of autologous chondrocyte implantation (ACI) for treating knee cartilage lesions has several limits, including the absence of healthy articular cartilage tissues for cell isolation and difficulties related to the chondrocyte expansion in vitro. Today, various ACI modifications are being developed using autologous chondrocytes from alternative sources, such as the auricles, nose and ribs. Adult stem cells from different tissues are also of great interest due to their less traumatic material extraction and their innate abilities of active proliferation and chondrogenic differentiation. According to the different adult stem cell types and their origin, various strategies have been proposed for stem cell expansion and initiation of their chondrogenic differentiation. The current review presents the diversity in developing applied techniques based on autologous adult stem cell differentiation to hyaline cartilage tissue and targeted to articular cartilage damage therapy.
Collapse
Affiliation(s)
- Anastasiia D. Kurenkova
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia or
| | - Irina A. Romanova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Pavel D. Kibirskiy
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia or
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia or
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Ekaterina V. Medvedeva
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia or
| |
Collapse
|
11
|
Altaie A, Baboolal TG, Wall O, Pandit H, Jones E, McGonagle D. Device-Based Enrichment of Knee Joint Synovial Cells to Drive MSC Chondrogenesis Without Prior Culture Expansion In Vitro: A Step Closer to 1-Stage Orthopaedic Procedures. Am J Sports Med 2022; 50:152-161. [PMID: 34779670 PMCID: PMC8739599 DOI: 10.1177/03635465211055164] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 07/27/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND Synovial fluid (SF) mesenchymal stem cells (MSCs) are derived from the synovial membrane and have cartilage repair potential. Their current use in clinical practice is largely exploratory. As their numbers tend to be small, therapeutic procedures using MSCs typically require culture expansion. Previous reports indicate that the stem cell-mobilizing device (STEM device) intraoperatively increases SF-MSCs. PURPOSE This study evaluated the chondrogenic potential of non-culture expanded synovium-mobilized MSCs and SF-microfragments obtained after enrichment using the STEM device and ascertained if device-mediated synovial membrane manipulation facilitated ongoing MSC release. STUDY DESIGN Controlled laboratory study. METHODS Two samples of aspiration fluid were collected intraoperatively before and after STEM device utilization from patients (n = 16) undergoing diagnostic or therapeutic knee arthroscopy. Human knee synovium (n = 5) was collected during total knee replacement, and a suspended culture was performed to assess the effect of the STEM device on ongoing MSC release. Colony forming unit-fibroblastic assays were used to determine the number of MSCs. Additionally, cytometric characterization of stromal and immune cells and chondrogenesis differentiation assay were performed without culture expansion. Filtered platelet concentrates were prepared using the HemaTrate system. RESULTS After STEM device use, a significant increase was evident in SF-MSCs (P = .03) and synovial fluid-resident synovial tissue microfragments (P = .03). In vitro-suspended synovium released significantly more MSCs following STEM device use than nonstimulated synovium (P = .01). The STEM device-released total cellular fraction produced greater in vitro chondrogenesis with significantly more glycosaminoglycans (GAGs; P < .0001) when compared with non-STEM device synovial fluid material. Nonexpanded SF-MSCs and SF-microfragments combined with autologous filtered platelet concentrate produced significantly more GAGs than the complete chondrogenic media (P < .0001). The STEM device-mobilized cells contained more M2 macrophage cells and fewer M1 cells. CONCLUSION Non-culture expanded SF-MSCs and SF-microfragments had the potential to undergo chondrogenesis without culture expansion, which can be augmented using the STEM device with increased MSC release from manipulated synovium for several days. Although preliminary, these findings offer proof of concept toward manipulation of the knee joint environment to facilitate endogenous repair responses. CLINICAL RELEVANCE Although numbers were small, this study highlights 3 factors relevant to 1-stage joint repair using the STEM device: increased SF-MSCs and SF-microfragments and prolonged synovial release of MSCs. Joint repair strategies involving endogenous MSCs for cartilage repair without the need for culture expansion in a 1-stage procedure may be possible.
Collapse
Affiliation(s)
- Ala Altaie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Thomas G. Baboolal
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Owen Wall
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Leeds Biomedical Research Centre, National Institute for Health Research, Leeds, UK
| | - Hemant Pandit
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Leeds Biomedical Research Centre, National Institute for Health Research, Leeds, UK
| | - Elena Jones
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- Leeds Biomedical Research Centre, National Institute for Health Research, Leeds, UK
| |
Collapse
|
12
|
Sanooghi D, Amini N, Azedi F, Bagher Z, Parvishan A, Lotfi A, Rashidi N, Lotfi E, Sayahpour FA, Faghihi F. Differentiation of Mesenchymal Stem Cells Derived From Human Adipose Tissue Into Cholinergic-like Cells: An in Vitro Study. Basic Clin Neurosci 2021; 12:315-323. [PMID: 34917291 PMCID: PMC8666926 DOI: 10.32598/bcn.2021.1008.2] [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: 08/23/2019] [Revised: 10/10/2019] [Accepted: 02/15/2020] [Indexed: 11/20/2022] Open
Abstract
Introduction: Cholinergic-associated diseases currently constitute a significant cause of neurological and neurodegenerative disabilities. As the drugs are not efficient in improving the suffered tissues, stem cell treatment is considered an effective strategy for substituting the lost cells. Methods: In the current study, we set out to investigate the differentiation properties of human Adipose-Derived Mesenchymal Stem Cells (AD-MSCs) into cholinergic-like cells by two morphogens of Retinoic Acid (RA) and Sonic Hedgehog (Shh) using a three-step in vitro procedure. The results were evaluated using real-time PCR, flow cytometry, and immunocytochemistry for two weeks. Results: Our data showed that the cells could express cholinergic specific markers, including Islet-1, Acetylcholinesterase (AChE), SMI-32, and Nestin, at mRNA and protein levels. We could also quantitatively evaluate the expression of Islet-1, AChE, and Nestin at 14 days post-induction using flow cytometry. Conclusion: Human AD-MSCs are potent cells to differentiate into cholinergic-like cells in the presence of RA and Shh through a three-step protocol. Thus, they could be a suitable cell candidate for the regeneration of cholinergic-associated diseases. However, more functional and electrophysiological analyses are needed in this regard.
Collapse
Affiliation(s)
- Davood Sanooghi
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Naser Amini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Azedi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zohreh Bagher
- ENT and Head & Neck Research Center, The Five Basic Sensory Institute, Hazrat Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering & Regenerative Medicin, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Asghar Parvishan
- Department of Neuroscience, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Lotfi
- Damavand Agricultural College, Technical and Vocational University, Tehran, Iran
| | - Nooshin Rashidi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Erfan Lotfi
- School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Forough Azam Sayahpour
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Academic Center for Education, Culture and Research, Tehran, Iran
| | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Saxena S, Patel DD, Shah A, Doctor M. Fat Chance for Hidden Lesions: Pictorial Review of Hoffa's Fat Pad Lesions. Indian J Radiol Imaging 2021; 31:961-974. [PMID: 35136510 PMCID: PMC8817800 DOI: 10.1055/s-0041-1739383] [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: 11/01/2022] Open
Abstract
AbstractHoffa's fat pad (HFP) is the most commonly afflicted among all the knee fat pads. Anterior knee pain is common in various pathologies of HFP, as it is richly innervated. A potpourri of the intrinsic and extrinsic pathologies and the tumors and tumor-like conditions affect HFP, and MRI remains the fundamental modality to assess them and provide a specific diagnosis.
Collapse
Affiliation(s)
- Suvinay Saxena
- Department of Diagnostic and Interventional Radiology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | - Drushi D. Patel
- Department of Radiology, Gujarat Imaging Centre, Samved Hospital Post Graduate Institute of Radiology and Imaging, Ahmedabad, Gujarat, India
| | - Ankur Shah
- Department of Radiology, Sadbhav Imaging Centre and Gujarat Imaging Centre Postgraduate Institute of Radiology and Imaging, Ahmedabad, Gujarat, India
| | - Mrugesh Doctor
- Department of Radiology, Gujarat Imaging Centre, Samved Hospital Post Graduate Institute of Radiology and Imaging, Ahmedabad, Gujarat, India
| |
Collapse
|
14
|
Itoh Y, Itoh S, Naruse H, Kagioka T, Hue MT, Abe M, Hayashi M. Intracellular density is a novel indicator of differentiation stages of murine osteoblast lineage cells. J Cell Biochem 2021; 122:1805-1816. [PMID: 34427353 DOI: 10.1002/jcb.30135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/15/2021] [Accepted: 08/05/2021] [Indexed: 11/12/2022]
Abstract
Osteoblasts are primary bone-making cells originating from mesenchymal stem cells (MSCs) in the bone marrow. The differentiation of MSCs to mature osteoblasts involves an intermediate stage called preosteoblasts, but the details of this process remain unclear. This study focused on the intracellular density of immature osteoblast lineage cells and hypothesized that the density might vary during differentiation and might be associated with the differentiation stages of osteoblast lineage cells. This study aimed to clarify the relationship between intracellular density and differentiation stages using density gradient centrifugation. Primary murine bone marrow stromal cell cultures were prepared in an osteogenic induction medium, and cells were separated into three fractions (low, intermediate, and high-density). The high-density fraction showed elevated expression of osteoblast differentiation markers (Sp7, Col1a1, Spp1, and Bglap) and low expression of MSC surface markers (Sca-1, CD73, CD105, and CD106). In contrast, the low-density fraction showed a high expression of MSC surface markers. These results indicated that intracellular density increased during differentiation from preosteoblasts to committed osteoblasts. Intracellular density may be a novel indicator for osteoblast differentiation stages. Density gradient centrifugation is a novel technique to study the process by which preosteoblasts transform into bone-forming cells.
Collapse
Affiliation(s)
- Yuki Itoh
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Shousaku Itoh
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Haruna Naruse
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Takumi Kagioka
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Mai Thi Hue
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Makoto Abe
- Department of Oral Anatomy and Developmental Biology, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Mikako Hayashi
- Department of Restorative Dentistry and Endodontology, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
15
|
Rahman S, Szojka ARA, Liang Y, Kunze M, Goncalves V, Mulet-Sierra A, Jomha NM, Adesida AB. Inability of Low Oxygen Tension to Induce Chondrogenesis in Human Infrapatellar Fat Pad Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:703038. [PMID: 34381784 PMCID: PMC8350173 DOI: 10.3389/fcell.2021.703038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Articular cartilage of the knee joint is avascular, exists under a low oxygen tension microenvironment, and does not self-heal when injured. Human infrapatellar fat pad-sourced mesenchymal stem cells (IFP-MSC) are an arthroscopically accessible source of mesenchymal stem cells (MSC) for the repair of articular cartilage defects. Human IFP-MSC exists physiologically under a low oxygen tension (i.e., 1-5%) microenvironment. Human bone marrow mesenchymal stem cells (BM-MSC) exist physiologically within a similar range of oxygen tension. A low oxygen tension of 2% spontaneously induced chondrogenesis in micromass pellets of human BM-MSC. However, this is yet to be demonstrated in human IFP-MSC or other adipose tissue-sourced MSC. In this study, we explored the potential of low oxygen tension at 2% to drive the in vitro chondrogenesis of IFP-MSC. We hypothesized that 2% O2 will induce stable chondrogenesis in human IFP-MSC without the risk of undergoing endochondral ossification at ectopic sites of implantation. METHODS Micromass pellets of human IFP-MSC were cultured under 2% O2 or 21% O2 (normal atmosphere O2) in the presence or absence of chondrogenic medium with transforming growth factor-β3 (TGFβ3) for 3 weeks. Following in vitro chondrogenesis, the resulting pellets were implanted in immunodeficient athymic nude mice for 3 weeks. RESULTS A low oxygen tension of 2% was unable to induce chondrogenesis in human IFP-MSC. In contrast, chondrogenic medium with TGFβ3 induced in vitro chondrogenesis. All pellets were devoid of any evidence of undergoing endochondral ossification after subcutaneous implantation in athymic mice.
Collapse
Affiliation(s)
- Samia Rahman
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Alexander R. A. Szojka
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Yan Liang
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Melanie Kunze
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Victoria Goncalves
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Aillette Mulet-Sierra
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Nadr M. Jomha
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Adetola B. Adesida
- Laboratory of Stem Cell Biology and Orthopedic Tissue Engineering, Division of Orthopedic Surgery and Surgical Research, Department of Surgery, University of Alberta, Edmonton, AB, Canada
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of Alberta Hospital, Edmonton, AB, Canada
| |
Collapse
|
16
|
Zhou Y, Li H, Xiang D, Shao J, Fu Q, Han Y, Zhu J, Chen Y, Qian Q. The clinical efficacy of arthroscopic therapy with knee infrapatellar fat pad cell concentrates in treating knee cartilage lesion: a prospective, randomized, and controlled study. J Orthop Surg Res 2021; 16:87. [PMID: 33509248 PMCID: PMC7841893 DOI: 10.1186/s13018-021-02224-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
Introduction To evaluate the clinical efficacy of arthroscopic therapy with infrapatellar fat pad cell concentrates in treating knee cartilage lesions, we conducted a prospective randomized single-blind clinical study of controlled method. Methods Sixty cases from Shanghai Changzheng Hospital from April 2018 to December 2019 were chosen and randomly divided into 2 groups equally. Patients in the experiment group were treated through knee arthroscopy with knee infrapatellar fat pad cell concentrates containing mesenchymal stromal cells, while patients in the control group were treated through regular knee arthroscopic therapy. VAS and WOMAC scores were assessed at pre-operation, and 6 weeks, 12 weeks, 6 months, and 12 months after intervention. MORCART scores were assessed at pre-operation and 12 months after intervention. Results Twenty-nine cases in the experiment group and 28 cases in the control group were followed up. No significant difference in VAS, WOMAC, and MOCART scores were found between the two groups before surgery (P > 0.05). The WOMAC total and WOMAC function scores of the experiment group were significantly lower than those of the control group 6 months and 12 months after surgery (P < 0.05). The VAS rest and VAS motion scores of the experiment group were found significantly lower than those of the control group 12 months after surgery (P < 0.05). The MOCART scores of the experiment group were found significantly higher compared with the control group 12 months after surgery (P < 0.05). No significant difference in WOMAC stiffness scores were found between the two groups. Conclusions The short-term results of our study are encouraging and demonstrate that knee arthroscopy with infrapatellar fat pad cell concentrates containing mesenchymal stromal cells is safe and provides assistance in reducing pain and improving function in patients with knee cartilage lesions. Trial registration ChiCTR1800015379. Registered on 27 March 2018, http://www.chictr.org.cn/showproj.aspx?proj=25901.
Collapse
Affiliation(s)
- Yiqin Zhou
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Haobo Li
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Dong Xiang
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Jiahua Shao
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Qiwei Fu
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Yaguang Han
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China
| | - Jun Zhu
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China.
| | - Yi Chen
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China.
| | - Qirong Qian
- Department of Joint Surgery and Sports Medicine, Shanghai Changzheng Hospital, Naval Medical University, No.415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
17
|
Bui TVA, Hwang JW, Lee JH, Park HJ, Ban K. Challenges and Limitations of Strategies to Promote Therapeutic Potential of Human Mesenchymal Stem Cells for Cell-Based Cardiac Repair. Korean Circ J 2021; 51:97-113. [PMID: 33525065 PMCID: PMC7853896 DOI: 10.4070/kcj.2020.0518] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) represent a population of adult stem cells residing in many tissues, mainly bone marrow, adipose tissue, and umbilical cord. Due to the safety and availability of standard procedures and protocols for isolation, culturing, and characterization of these cells, MSCs have emerged as one of the most promising sources for cell-based cardiac regenerative therapy. Once transplanted into a damaged heart, MSCs release paracrine factors that nurture the injured area, prevent further adverse cardiac remodeling, and mediate tissue repair along with vasculature. Numerous preclinical studies applying MSCs have provided significant benefits following myocardial infarction. Despite promising results from preclinical studies using animal models, MSCs are not up to the mark for human clinical trials. As a result, various approaches have been considered to promote the therapeutic potency of MSCs, such as genetic engineering, physical treatments, growth factor, and pharmacological agents. Each strategy has targeted one or multi-potentials of MSCs. In this review, we will describe diverse approaches that have been developed to promote the therapeutic potential of MSCs for cardiac regenerative therapy. Particularly, we will discuss major characteristics of individual strategy to enhance therapeutic efficacy of MSCs including scientific principles, advantages, limitations, and improving factors. This article also will briefly introduce recent novel approaches that MSCs enhanced therapeutic potentials of other cells for cardiac repair.
Collapse
Affiliation(s)
- Thi Van Anh Bui
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Ji Won Hwang
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea
| | - Jung Hoon Lee
- Department of Chemistry, City University of Hong Kong, Hong Kong SAR, China
| | - Hun Jun Park
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Korea.,Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Korea.,Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
18
|
van Schaik TJA, Gaul F, Dorthé EW, Lee EE, Grogan SP, D’Lima DD. Development of an Ex Vivo Murine Osteochondral Repair Model. Cartilage 2021; 12:112-120. [PMID: 30373381 PMCID: PMC7755972 DOI: 10.1177/1947603518809402] [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] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE Mouse models are commonly used in research applications due to the relatively low cost, highly characterized strains, as well as the availability of many genetically modified phenotypes. In this study, we characterized an ex vivo murine osteochondral repair model using human infrapatellar fat pad (IPFP) progenitor cells. DESIGN Femurs from euthanized mice were removed and clamped in a custom multidirectional vise to create cylindrical osteochondral defects 0.5 mm in diameter and 0.5 mm deep in both condyles. The IPFP contains progenitors that are a promising cell source for the repair of osteochondral defects. For proof of concept, human IPFP-derived progenitor cells, from osteoarthritic (OA) patients, cultured as pellets, were implanted into the defects and cultured in serum-free medium with TGFβ3 for 3 weeks and then processed for histology and immunostaining. RESULTS The custom multidirectional vise enabled reproducible creation of osteochondral defects in murine femoral condyles. Implantation of IPFP-derived progenitor cells led to development of cartilaginous tissue with Safranin O staining and deposition of collagen type II in the extracellular matrix. CONCLUSIONS We showed feasibility in creating ex vivo osteochondral defects and demonstrated the regenerative potential of OA human IPFP-derived progenitors in mouse femurs. The murine model can be used to study the effects of aging and OA on tissue regeneration and to explore molecular mechanisms of cartilage repair using genetically modified mice.
Collapse
Affiliation(s)
- Thomas J. A. van Schaik
- Orthopaedic Research Laboratory, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Florian Gaul
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Erik W. Dorthé
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Emily E. Lee
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Shawn P. Grogan
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Darryl D. D’Lima
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA, USA,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA,Darryl D. D’Lima, Scripps Health, Shiley Center for Orthopaedic Research and Education at Scripps Clinic, 10666 North Torrey Pines Road, MS126, La Jolla, CA 92027, USA.
| |
Collapse
|
19
|
Siengdee P, Oster M, Reyer H, Viergutz T, Wimmers K, Ponsuksili S. Morphological and Molecular Features of Porcine Mesenchymal Stem Cells Derived From Different Types of Synovial Membrane, and Genetic Background of Cell Donors. Front Cell Dev Biol 2020; 8:601212. [PMID: 33363158 PMCID: PMC7755640 DOI: 10.3389/fcell.2020.601212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 11/19/2020] [Indexed: 01/22/2023] Open
Abstract
Synovial mesenchymal stem cells (SMSCs) have become a great cell source for musculoskeletal stem cell research, especially related to cartilage and bone tissue regeneration, due to their superior cell proliferation properties and multidifferentiation potential into various cell lineages. This study revealed isolation methods, culture conditions, and morphological and molecular characterization of SMSCs derived fibrous synovium (FS) and adipose synovium (FP) of two pig breeds differing in growth performance [German Landrace (DL), and fat deposition (Angeln Saddleback (AS)]. Herein, FS possessed nucleated cell numbers nearly twice as high as those of FP at Passage 0. SMSCs derived from different types of synovial membrane and genetic background show similar cell morphologies and immunophenotypes, which were assessed by cell surface epitopes and multilineage differentiation potential, but differ significantly in their molecular characteristics. In addition, transcripts of SMSCs from AS were more enriched in IGF-1 signaling and VEGF ligand receptor, while SMSCs from DL were more enriched in growth hormone signaling and bone metabolism. The results indicate that genetics and tissues play significant roles for SMSC characteristics so that SMSCs can be traced back to the original cell donor and be used for fine turning in applications of medical research and therapies.
Collapse
Affiliation(s)
- Puntita Siengdee
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Michael Oster
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Henry Reyer
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Torsten Viergutz
- Institute for Reproductive Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Klaus Wimmers
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Institute for Genome Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
20
|
Basic Science of Resident Stem Cells. OPER TECHN SPORT MED 2020. [DOI: 10.1016/j.otsm.2020.150776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
21
|
Water-transport and intracellular ice formation of human adipose-derived stem cells during freezing. J Therm Biol 2020; 93:102689. [PMID: 33077114 DOI: 10.1016/j.jtherbio.2020.102689] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 07/22/2020] [Accepted: 08/05/2020] [Indexed: 11/22/2022]
Abstract
The key to optimizing the cryopreservation strategy of human adipose-derived stem cells (hADSCs) is to identify the biophysical characteristics during freezing. Systematic freezing experiments were conducted under a cryo-microscope system to investigate the cryoinjury mechanism for hADSCs at different cooling rates. By simultaneously fitting morphological change data to the water-transport equation at 5, 10 and 20 °C/min, the plasma membrane hydraulic conductivity, Lpg, and activation energy, ELp, were determined. Moreover, the optimal cooling rate was also predicted by using mathematical model methods. Additionally, the surface-catalyzed nucleation (SCN) parameters were calculated by fitting in numerical models, Ω0SCN and k0SCN were determined at cooling rates of 30, 45 and 60 °C/min. These results may provide potential application value for cryopreservation of hADSCs.
Collapse
|
22
|
Desai S, Jayasuriya CT. Implementation of Endogenous and Exogenous Mesenchymal Progenitor Cells for Skeletal Tissue Regeneration and Repair. Bioengineering (Basel) 2020; 7:E86. [PMID: 32759659 PMCID: PMC7552784 DOI: 10.3390/bioengineering7030086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/25/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
Harnessing adult mesenchymal stem/progenitor cells to stimulate skeletal tissue repair is a strategy that is being actively investigated. While scientists continue to develop creative and thoughtful ways to utilize these cells for tissue repair, the vast majority of these methodologies can ultimately be categorized into two main approaches: (1) Facilitating the recruitment of endogenous host cells to the injury site; and (2) physically administering into the injury site cells themselves, exogenously, either by autologous or allogeneic implantation. The aim of this paper is to comprehensively review recent key literature on the use of these two approaches in stimulating healing and repair of different skeletal tissues. As expected, each of the two strategies have their own advantages and limitations (which we describe), especially when considering the diverse microenvironments of different skeletal tissues like bone, tendon/ligament, and cartilage/fibrocartilage. This paper also discusses stem/progenitor cells commonly used for repairing different skeletal tissues, and it lists ongoing clinical trials that have risen from the implementation of these cells and strategies. Lastly, we discuss our own thoughts on where the field is headed in the near future.
Collapse
Affiliation(s)
| | - Chathuraka T. Jayasuriya
- Department of Orthopaedics, Warren Alpert Medical School of Brown University and the Rhode Island Hospital, Providence, RI 02903, USA;
| |
Collapse
|
23
|
Greif DN, Kouroupis D, Murdock CJ, Griswold AJ, Kaplan LD, Best TM, Correa D. Infrapatellar Fat Pad/Synovium Complex in Early-Stage Knee Osteoarthritis: Potential New Target and Source of Therapeutic Mesenchymal Stem/Stromal Cells. Front Bioeng Biotechnol 2020; 8:860. [PMID: 32850724 PMCID: PMC7399076 DOI: 10.3389/fbioe.2020.00860] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022] Open
Abstract
The infrapatellar fat pad (IFP) has until recently been viewed as a densely vascular and innervated intracapsular/extrasynovial tissue with biomechanical roles in the anterior compartment of the knee. Over the last decade, secondary to the proposition that the IFP and synovium function as a single unit, its recognized tight molecular crosstalk with emerging roles in the pathophysiology of joint disease, and the characterization of immune-related resident cells with varying phenotypes (e.g., pro and anti-inflammatory macrophages), this structural complex has gained increasing attention as a potential therapeutic target in patients with various knee pathologies including osteoarthritis (KOA). Furthermore, the description of the presence of mesenchymal stem/stromal cells (MSC) as perivascular cells within the IFP (IFP-MSC), exhibiting immunomodulatory, anti-fibrotic and neutralizing activities over key local mediators, has promoted the IFP as an alternative source of MSC for cell-based therapy protocols. These complementary concepts have supported the growing notion of immune and inflammatory events participating in the pathogenesis of KOA, with the IFP/synovium complex engaging not only in amplifying local pathological responses, but also as a reservoir of potential therapeutic cell-based products. Consequently, the aim of this review is to outline the latest discoveries related with the IFP/synovium complex as both an active participant during KOA initiation and progression thus emerging as a potential target, and a source of therapeutic IFP-MSCs. Finally, we discuss how these notions may help the design of novel treatments for KOA through modulation of local cellular and molecular cascades that ultimately lead to joint destruction.
Collapse
Affiliation(s)
- Dylan N Greif
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Dimitrios Kouroupis
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Christopher J Murdock
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Anthony J Griswold
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Lee D Kaplan
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Thomas M Best
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Diego Correa
- Department of Orthopedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL, United States.,Diabetes Research Institute and Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
24
|
Are Stem Cells Derived from Synovium and Fat Pad Able to Treat Induced Knee Osteoarthritis in Rats? Int J Rheumatol 2020; 2020:9610261. [PMID: 32765610 PMCID: PMC7374223 DOI: 10.1155/2020/9610261] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 01/22/2023] Open
Abstract
Background Osteoarthritis (OA) is a chronic disease and a significant cause of joint pain, tenderness, and limitation of motion. At present, no specific treatment is available, and mesenchymal stem cells (MSCs) have shown promising potentials in this regard. Herein, we aimed to evaluate the repairing potentials of stem cells derived from the synovium and fat pad in the treatment of OA. Methods Twenty-eight male rats (220 ± 20 g, aged 10-12 weeks), were randomly divided into four groups (n = 7): C1: nontreated group, C2: Hyalgan-treated group, E1: adipose tissue-derived stem cell-treated group, and E2: synovial membrane-based stem cell-treated group. Collagenase type II was injected into the left knee; after eight weeks, OA was developed. Then, stem cells were injected, and rats were followed for three months. Afterward, specimens and radiological images were investigated. p value ≤ 0.05 was set as statistically significant. Results Compared to the C1 group, the E1 and E2 groups showed significantly better results in all six pathological criteria as well as joint space width and osteophytes of medial tibial, medial femoral, and medial fabellar condyles (p ≤ 0.001). Similarly, compared to the C2 group, the E1 and E2 groups had better scores regarding surface, matrix, cell distribution, and cell population viability (p < 0.05). E2 showed considerably higher scores compared to C2 regarding subchondral bone and cartilage mineralization (p < 0.05). The joint space width was similar between the C2 and E groups. Conclusion Treatment of OA with MSCs, particularly synovial membrane-derived stem cells, not only prevented but also healed OA of the knee to some extent in comparison to the Hyalgan and nontreatment groups.
Collapse
|
25
|
Francis SL, Yao A, Choong PFM. Culture Time Needed to Scale up Infrapatellar Fat Pad Derived Stem Cells for Cartilage Regeneration: A Systematic Review. Bioengineering (Basel) 2020; 7:bioengineering7030069. [PMID: 32635513 PMCID: PMC7552776 DOI: 10.3390/bioengineering7030069] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022] Open
Abstract
Adipose tissue is a rich source of stem cells, which are reported to represent 2% of the stromal vascular fraction (SVF). The infrapatellar fat pad (IFP) is a unique source of tissue, from which human adipose-derived stem cells (hADSCs) have been shown to harbour high chondrogenic potential. This review aims to calculate, based on the literature, the culture time needed before an average knee articular cartilage defect can be treated using stem cells obtained from arthroscopically or openly harvested IFP. Firstly, a systematic literature review was performed to search for studies that included the number of stem cells isolated from the IFP. Subsequent analysis was conducted to identify the amount of IFP tissue harvestable, stem cell count and the overall yield based on the harvesting method. We then determined the minimum time required before treating an average-sized knee articular cartilage defect with IFP-derived hADSCs by using our newly devised equation. The amount of fat tissue, the SVF cell count and the stem cell yield are all lower in arthroscopically harvested IFP tissue compared to that collected using arthrotomy. As an extrapolation, we show that an average knee defect can be treated in 20 or 17 days using arthroscopically or openly harvested IFP-derived hADSCs, respectively. In summary, the systematic review conducted in this study reveals that there is a higher amount of fat tissue, SVF cell count and overall yield (cells/volume or cells/gram) associated with open (arthrotomy) compared to arthroscopic IFP harvest. In addition to these review findings, we demonstrate that our novel framework can give an indication about the culture time needed to scale up IFP-derived stem cells for the treatment of articular cartilage defects based on harvesting method.
Collapse
Affiliation(s)
- Sam L. Francis
- Department of Surgery, The University of Melbourne, Melbourne, VIC 3065, Australia;
- Department of Orthopaedics, St Vincent’s Hospital, Melbourne, VIC 3056, Australia;
- Biofab 3D, Aikenhead Centre for Medical Discovery, Melbourne, VIC 3065, Australia
- Correspondence: ; Tel.: +61-466-640-801
| | - Angela Yao
- Department of Orthopaedics, St Vincent’s Hospital, Melbourne, VIC 3056, Australia;
| | - Peter F. M. Choong
- Department of Surgery, The University of Melbourne, Melbourne, VIC 3065, Australia;
- Department of Orthopaedics, St Vincent’s Hospital, Melbourne, VIC 3056, Australia;
- Biofab 3D, Aikenhead Centre for Medical Discovery, Melbourne, VIC 3065, Australia
| |
Collapse
|
26
|
Vaishya R, Kansagra A, Agarwal AK, Vijay V. A Giant Ganglion Cyst Arising from Lateral Hoffa's Fat Pad of the Knee. J Orthop Case Rep 2020; 9:36-39. [PMID: 32548025 PMCID: PMC7276596 DOI: 10.13107/jocr.2019.v09.i06.578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Introduction The ganglion cysts are benign fluid-filled sacs, which often arise from a tendon sheath or a joint capsule. Their origin from the fat pad of the knee is rare. Several studies have described intra-articular ganglion cysts in detail; however, extra-articular soft-tissue ganglion cysts have been reported sparingly. We report a rare case of giant ganglion cyst arising from lateral Hoffa’s fat pad (HFP). Case Report A 59-year-old male patient presented with a 3-year history of swelling of the left knee, with occasional pain. There was no history of trauma or any constitutional symptoms. There was an apparent swelling (10 cm ×5 cm in size) around the anterolateral aspect of the knee joint. A magnetic resonance imaging (MRI) scan revealed a multilobular, complex cystic lesion of the lateral HFP. Surgical excision of the cyst was done, and histopathological examination confirmed the diagnosis of the ganglion cyst. Conclusion Cysts and cystic-appearing lesions around the knee are not uncommon, but a ganglion cyst arising from HFP is rare. The presence of multipotent cells in the HFP may be responsible for producing a variety of cyst and cyst-like tumors around the anterior aspect of the knee joint. An MRI is the best imaging modality for the diagnosis of these cysts and cysts-like lesions around the knee. We recommend that the smaller intra-articular lesions can be resected arthroscopically, but larger lesions, with extraarticular extension, are best treated by open resection to avoid incomplete excision and recurrence.
Collapse
Affiliation(s)
- Raju Vaishya
- Department of Orthopaedics, Indraprastha Apollo Hospitals, SaritaVihar, New Delhi - 110076, India
| | - Ankur Kansagra
- Department of Orthopaedics, Indraprastha Apollo Hospitals, SaritaVihar, New Delhi - 110076, India
| | - Amit Kumar Agarwal
- Department of Orthopaedics, Indraprastha Apollo Hospitals, SaritaVihar, New Delhi - 110076, India
| | - Vipul Vijay
- Department of Orthopaedics, Indraprastha Apollo Hospitals, SaritaVihar, New Delhi - 110076, India
| |
Collapse
|
27
|
Wu CL, Harasymowicz NS, Klimak MA, Collins KH, Guilak F. The role of macrophages in osteoarthritis and cartilage repair. Osteoarthritis Cartilage 2020; 28:544-554. [PMID: 31926267 PMCID: PMC7214213 DOI: 10.1016/j.joca.2019.12.007] [Citation(s) in RCA: 168] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/15/2019] [Accepted: 12/30/2019] [Indexed: 02/07/2023]
Abstract
Osteoarthritis (OA) is a family of degenerative diseases affecting multiple joint tissues. Despite the diverse etiology and pathogenesis of OA, increasing evidence suggests that macrophages can play a significant role in modulating joint inflammation, and thus OA severity, via various secreted mediators. Recent advances in next-generation sequencing technologies coupled with proteomic and epigenetic tools have greatly facilitated research to elucidate the embryonic origin of macrophages in various tissues including joint synovium. Furthermore, scientists have now begun to appreciate that macrophage polarization can span beyond the conventionally recognized binary states (i.e., pro-inflammatory M1-like vs anti-inflammatory M2-like) and may encompass a broad spectrum of phenotypes. Although the presence of these cells has been shown in multiple joint tissues, additional mechanistic studies are required to provide a comprehensive understanding of the precise role of these diverse macrophage populations in OA onset and progression. New approaches that can modulate macrophages into desired functional phenotypes may provide novel therapeutic strategies for preventing OA or enhancing cartilage repair and regeneration.
Collapse
Affiliation(s)
- C-L Wu
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - N S Harasymowicz
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - M A Klimak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA
| | - K H Collins
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA
| | - F Guilak
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA; Shriners Hospitals for Children - St. Louis, St. Louis, MO 63110, USA; Center of Regenerative Medicine, Washington University, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University, St. Louis, MO 63110, USA.
| |
Collapse
|
28
|
Transient Existence of Circulating Mesenchymal Stem Cells in the Deep Veins in Humans Following Long Bone Intramedullary Reaming. J Clin Med 2020; 9:jcm9040968. [PMID: 32244388 PMCID: PMC7230570 DOI: 10.3390/jcm9040968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 12/24/2022] Open
Abstract
The biology of mesenchymal stem cells (MSCs) in humans is incompletely understood and a possible role of systemically circulating cells in health and autoimmune disease remains controversial. Physiological movement of bone marrow MSCs to sites of injury would support the rationale for intravenous administration for relocation to damaged organs. We hypothesized that biophysical skeletal trauma rather than molecular cues may explain reported MSC circulation phenomena. Deep-femoral vein (FV) and matched peripheral vein blood samples (PVBs) were collected from patients undergoing lower-limb orthopaedic procedures during surgery (tibia using conventional sequential reaming, n = 9, femur using reamer/irrigator/aspirator (RIA), n = 15). PVBs were also taken from early (n = 15) and established (n = 12) rheumatoid arthritis (RA) patients and healthy donors (n = 12). Colony-forming unit-fibroblasts (CFU-Fs) were found in 17/36 FVBs but only 7/74 PVBs (mostly from femoral RIA); highly proliferative clonogenic cells were not generated. Only one colony was found in control/RA samples (n = 28). The rare CFU-Fs’ MSC nature was confirmed by phenotypic: CD105+/CD73+/CD90+ and CD19−/CD31−/CD33−/CD34−/CD45−/CD61−, and molecular profiles with 39/80 genes (including osteo-, chondro-, adipo-genic and immaturity markers) similar across multiple MSC tissue controls, but not dermal fibroblasts. Analysis of FVB-MSCs suggested that their likely origin was bone marrow as only two differences were observed between FVB-MSCs and IC-BM-MSCs (ACVR2A, p = 0.032 and MSX1, p = 0.003). Stromal cells with the phenotype and molecular profile of MSCs were scarcely found in the circulation, supporting the hypothesis that their very rare presence is likely linked to biophysical micro-damage caused by skeletal trauma (here orthopaedic manipulation) rather than specific molecular cues to a circulatory pool of MSCs capable of repair of remote organs or tissues. These findings support the use of organ resident cells or MSCs placed in situ to repair tissues rather than systemic administration.
Collapse
|
29
|
Zhong YC, Wang SC, Han YH, Wen Y. Recent Advance in Source, Property, Differentiation, and Applications of Infrapatellar Fat Pad Adipose-Derived Stem Cells. Stem Cells Int 2020; 2020:2560174. [PMID: 32215015 PMCID: PMC7081037 DOI: 10.1155/2020/2560174] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/12/2020] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Infrapatellar fat pad (IPFP) can be easily obtained during knee surgery, which avoids the damage to patients for obtaining IPFP. Infrapatellar fat pad adipose-derived stem cells (IPFP-ASCs) are also called infrapatellar fat pad mesenchymal stem cells (IPFP-MSCs) because the morphology of IPFP-ASCs is similar to that of bone marrow mesenchymal stem cells (BM-MSCs). IPFP-ASCs are attracting more and more attention due to their characteristics suitable to regenerative medicine such as strong proliferation and differentiation, anti-inflammation, antiaging, secreting cytokines, multipotential capacity, and 3D culture. IPFP-ASCs can repair articular cartilage and relieve the pain caused by osteoarthritis, so most of IPFP-related review articles focus on osteoarthritis. This article reviews the anatomy and function of IPFP, as well as the discovery, amplification, multipotential capacity, and application of IPFP-ASCs in order to explain why IPFP-ASC is a superior stem cell source in regenerative medicine.
Collapse
Affiliation(s)
- Yu-chen Zhong
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
- Class 4, Phase 102, China Medical University, Shenyang 110122, China
| | - Shi-chun Wang
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
- Class 4, Phase 102, China Medical University, Shenyang 110122, China
| | - Yin-he Han
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| | - Yu Wen
- Department of Histology and Embryology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China
| |
Collapse
|
30
|
The Effect of Blood-Derived Products on the Chondrogenic and Osteogenic Differentiation Potential of Adipose-Derived Mesenchymal Stem Cells Originated from Three Different Locations. Stem Cells Int 2019; 2019:1358267. [PMID: 32082382 PMCID: PMC7012275 DOI: 10.1155/2019/1358267] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/01/2019] [Accepted: 11/29/2019] [Indexed: 02/06/2023] Open
Abstract
Background Adipose-derived mesenchymal stem cells (AD-MSCs) from fat tissue considered “surgical waste” during joint surgery may provide a potent source for regenerative medicine. Intra-articular, homologous fat tissue (Hoffa's fat pad, pouch fat) might possess a superior chondrogenic and osteogenic differentiation potential in comparison to extra-articular, nonhomologous fat. Blood products might further enhance this potential. Methods AD-MSCs were isolated from fat tissue of 3 donors from 3 locations each, during total knee replacement. Isolated cells were analyzed via flow cytometry. Cells were supplemented with blood products: two types of platelet-rich plasma (EPRP—PRP prepared in the presence of EDTA; CPRP—PRP prepared in the presence of citrate), hyperacute serum (hypACT), and standard fetal calf serum (FCS) as a positive control. The viability of the cells was determined by XTT assay, and the progress of differentiation was tested via histological staining and monitoring of specific gene expression. Results Blood products enhance ex vivo cell metabolism. Chondrogenesis is enhanced by EDTA-PRP and osteogenesis by citrate PRP, whereas hyperacute serum enhances both differentiations comparably. This finding was consistent in histological analysis as well as in gene expression. Lower blood product concentrations and shorter differentiation periods lead to superior histological results for chondrogenesis. Both PRP types had a different biological effect depending upon concentration, whereas hyperacute serum seemed to have a more consistent effect, independent of the used concentration. Conclusion (i) Blood product preparation method, (ii) type of anticoagulant, (iii) differentiation time, and (iv) blood product concentration have a significant influence on stem cell viability and the differentiation potential, favouring no use of anticoagulation, shorter differentiation time, and lower blood product concentrations. Cell-free blood products like hyperacute serum may be considered as an alternative supplementation in regenerative medicine, especially for stem cell therapies.
Collapse
|
31
|
Stocco E, Barbon S, Piccione M, Belluzzi E, Petrelli L, Pozzuoli A, Ramonda R, Rossato M, Favero M, Ruggieri P, Porzionato A, Di Liddo R, De Caro R, Macchi V. Infrapatellar Fat Pad Stem Cells Responsiveness to Microenvironment in Osteoarthritis: From Morphology to Function. Front Cell Dev Biol 2019; 7:323. [PMID: 31921840 PMCID: PMC6914674 DOI: 10.3389/fcell.2019.00323] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/25/2019] [Indexed: 12/16/2022] Open
Abstract
Recently, infrapatellar fat pad (IFP) has been considered as a source of stem cells for cartilage regeneration in osteoarthritis (OA) due to their ability for differentiation into chondrocytes. However, stressful conditions, like that related to OA, may induce a pathogenic reprograming. The aim of this study was to characterize the structural and functional properties of a new population of stem cells isolated from osteoarthritic infrapatellar fat pad (OA-IFP). Nine OA patients undergoing total knee arthroplasty (TKA) were enrolled in this study [median age = 74 years, interquartile range (IQR) = 78.25-67.7; median body mass index = 29.4 Kg/m2, IQR = 31.7-27.4]. OA-IFP stem cells were isolated and characterized for morphology, stemness, metabolic profile and multi-differentiative potential by transmission electron microscopy, flow cytometric analysis, gene expression study and cytochemistry. OA-IFP stem cells displayed a spindle-like morphology, self-renewal potential and responsiveness (CD44, CD105, VEGFR2, FGFR2, IL1R, and IL6R) to microenvironmental stimuli. Characterized by high grade of stemness (STAT3, NOTCH1, c-Myc, OCT-4, KLF4, and NANOG), the cells showed peculiar immunophenotypic properties (CD73+/CD39+/CD90+/CD105+/CD44–/+/CD45–). The expression of HLA-DR, CD34, Fas and FasL was indicative of a possible phenotypic reprograming induced by inflammation. Moreover, the response to mechanical stimuli together with high expression level of COL1A1 gene, suggested their possible protective response against in vivo mechanical overloading. Conversely, the low expression of CD38/NADase was indicative of their inability to counteract NAD+-mediated OA inflammation. Based on the ultrastructural, immunophenotypic and functional characterization, OA-IFP stem cells were hypothesized to be primed by the pathological environment and to exert incomplete protective activity from OA inflammation.
Collapse
Affiliation(s)
- Elena Stocco
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padua, Italy.,LifeLab Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy.,Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling ONLUS, Padua, Italy
| | - Silvia Barbon
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padua, Italy.,LifeLab Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy.,Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling ONLUS, Padua, Italy
| | - Monica Piccione
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Elisa Belluzzi
- Musculoskeletal Pathology and Oncology Laboratory, Department of Surgery, Oncology and Gastroenterology DiSCOG, University of Padova, Padua, Italy.,Department of Orthopaedics and Orthopaedic Oncology, University of Padova, Padua, Italy
| | - Lucia Petrelli
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padua, Italy
| | - Assunta Pozzuoli
- Musculoskeletal Pathology and Oncology Laboratory, Department of Surgery, Oncology and Gastroenterology DiSCOG, University of Padova, Padua, Italy.,Department of Orthopaedics and Orthopaedic Oncology, University of Padova, Padua, Italy
| | - Roberta Ramonda
- Rheumatology Unit, Department of Medicine - DIMED, University Hospital of Padova, Padua, Italy
| | - Marco Rossato
- Clinica Medica 3, Department of Medicine - DIMED, University of Padova, Padua, Italy
| | - Marta Favero
- Rheumatology Unit, Department of Medicine - DIMED, University Hospital of Padova, Padua, Italy
| | - Pietro Ruggieri
- Department of Orthopaedics and Orthopaedic Oncology, University of Padova, Padua, Italy
| | - Andrea Porzionato
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padua, Italy.,LifeLab Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy
| | - Rosa Di Liddo
- Foundation for Biology and Regenerative Medicine, Tissue Engineering and Signaling ONLUS, Padua, Italy.,Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padua, Italy
| | - Raffaele De Caro
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padua, Italy.,LifeLab Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy
| | - Veronica Macchi
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, Padua, Italy.,LifeLab Program, Consorzio per la Ricerca Sanitaria (CORIS), Padua, Italy
| |
Collapse
|
32
|
Zhang S, Hu B, Liu W, Wang P, Lv X, Chen S, Liu H, Shao Z. Articular cartilage regeneration: The role of endogenous mesenchymal stem/progenitor cell recruitment and migration. Semin Arthritis Rheum 2019; 50:198-208. [PMID: 31767195 DOI: 10.1016/j.semarthrit.2019.11.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 10/04/2019] [Accepted: 11/01/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Trauma- or osteoarthritis-related cartilage damage resulted in functional decline of joints and heavy burden of public health. Recently, the reparative role of mesenchymal stem/progenitor cells (MSCs) in articular cartilage (AC) reconstruction is drawing more and more attention. OBJECTIVE To provide a review on (1) the locations and categories of joint-resident MSCs, (2) the regulation of chondrogenic capacities of MSCs, (3) the migratory approaches of MSCs to diseased AC and regulatory mechanisms. METHODS PubMed and Web of Science were searched for English-language articles related to MSC recruitment and migration for AC repair until June 2019. The presence of various MSCs in or around joints, the potential approaches to diseased AC` and the regenerative capacities of MSCs were reviewed. RESULTS Various intra- and peri-articular MSCs, with inherent migratory potentials, are present in multiple stem cell niches in or around joints. The recruitment and migration of joint-resident MSCs play crucial roles in endogenous AC repair. Multiple recruiting signals, such as chemokines, growth factors, etc., emerge during the development of AC diseases and participate in the regulation of MSC mobilization. Motivated MSCs could migrate into cartilage lesions and then exert multiple reparative potentials, including extracellular matrix (ECM) reconstruction and microenvironment modulation. CONCLUSION In general, AC repair based on endogenous MSC recruitment and migration is a feasible strategy, and a promising research field. Furthermore, endogenous AC repair mediated by native MSCs would provide new opportunities to efficient preventative or therapeutic options for AC diseases.
Collapse
Affiliation(s)
- Shuo Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Binwu Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Peng Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| | - Songfeng Chen
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| | - Hongjian Liu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, Hubei Province, China.
| |
Collapse
|
33
|
Jiang LF, Fang JH, Wu LD. Role of infrapatellar fat pad in pathological process of knee osteoarthritis: Future applications in treatment. World J Clin Cases 2019; 7:2134-2142. [PMID: 31531309 PMCID: PMC6718789 DOI: 10.12998/wjcc.v7.i16.2134] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 06/11/2019] [Accepted: 06/27/2019] [Indexed: 02/05/2023] Open
Abstract
It has been found that obese people have a higher proportion in suffering from osteoarthritis (OA), not only in the weight-bearing joints like knee and hip joints, even in non-weight-bearing joints such as hand joints. One of the reasons is because the large amount of adipose tissue secretes some factors, which can promote the occurrence of arthritis. As an important structure of the knee joint, the infrapatellar fat pad (IPFP) is actually a piece of adipose tissue. The aim of this review is to offer a comprehensive view of the anatomy and physiological characteristics of IPFP and its relationship with the pathological process of OA, indicating the important function of IPFP in OA. At the same time, with the development of adipose derived stem cells in the treatment of OA, owing to its special advantages, the IPFP is becoming a kind of important, minimally invasive fat stem cell source, providing a new approach for the treatment of OA. We hope that this review will offer an overview of all published data regarding the IPFP and will indicate novel directions for future research.
Collapse
Affiliation(s)
- Li-Feng Jiang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Jing-Hua Fang
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| | - Li-Dong Wu
- Department of Orthopedics Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
34
|
Browe DC, Mahon OR, Díaz‐Payno PJ, Cassidy N, Dudurych I, Dunne A, Buckley CT, Kelly DJ. Glyoxal cross‐linking of solubilized extracellular matrix to produce highly porous, elastic, and chondro‐permissive scaffolds for orthopedic tissue engineering. J Biomed Mater Res A 2019; 107:2222-2234. [DOI: 10.1002/jbm.a.36731] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 05/03/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Affiliation(s)
- David C. Browe
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Olwyn R. Mahon
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- School of Biochemistry and Immunology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin Ireland
| | - Pedro J. Díaz‐Payno
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Nina Cassidy
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
| | - Ivan Dudurych
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
| | - Aisling Dunne
- School of Biochemistry and Immunology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin Dublin Ireland
| | - Conor T. Buckley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| | - Daniel J. Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences InstituteTrinity College Dublin Dublin Ireland
- Department of Mechanical and Manufacturing Engineering, School of EngineeringTrinity College Dublin Dublin Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER)Royal College of Surgeons in Ireland and Trinity College Dublin Dublin Ireland
| |
Collapse
|
35
|
Sasaki A, Mizuno M, Mochizuki M, Sekiya I. Mesenchymal stem cells for cartilage regeneration in dogs. World J Stem Cells 2019; 11:254-269. [PMID: 31171954 PMCID: PMC6545524 DOI: 10.4252/wjsc.v11.i5.254] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/29/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
Articular cartilage damage and osteoarthritis (OA) are common orthopedic diseases in both humans and dogs. Once damaged, the articular cartilage seldom undergoes spontaneous repair because of its avascular, aneural, and alymphatic state, and the damage progresses to a chronic and painful situation. Dogs have distinctive characteristics compared to other laboratory animal species in that they share an OA pathology with humans. Dogs can also require treatment for naturally developed OA; therefore, effective treatment methods for OA are desired in veterinary medicine as well as in human medicine. Recently, interest has grown in regenerative medicine that includes the use of mesenchymal stem cells (MSCs). In cartilage repair, MSCs are a promising therapeutic tool due to their self-renewal capacity, ability to differentiate into cartilage, potential for trophic factor production, and capacity for immunomodulation. The MSCs from dogs (canine MSCs; cMSCs) share various characteristics with MSCs from other animal species, but they show some deviations, particularly in their differentiation ability and surface epitope expression. In vivo studies of cMSCs have demonstrated that intraarticular cMSC injection into cartilage lesions results in excellent hyaline cartilage regeneration. In clinical situations, cMSCs have shown great therapeutic effects, including amelioration of pain and lameness in dogs suffering from OA. However, some issues remain, such as a lack of regulations or guidelines and a need for unified methods for the use of cMSCs. This review summarizes what is known about cMSCs, including their in vitro characteristics, their therapeutic effects in cartilage lesion treatment in preclinical in vivo studies, their clinical efficacy for treatment of naturally developed OA in dogs, and the current limitations of cMSC studies.
Collapse
Affiliation(s)
- Akari Sasaki
- Laboratory of Veterinary Emergency Medicine, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Mitsuru Mizuno
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Manabu Mochizuki
- Laboratory of Veterinary Emergency Medicine, Graduate School of Agricultural and Life Sciences, the University of Tokyo, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Ichiro Sekiya
- Center for Stem Cell and Regenerative Medicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8519, Japan
| |
Collapse
|
36
|
Isolation and Characterization of an Adult Stem Cell Population from Human Epidural Fat. Stem Cells Int 2019; 2019:2175273. [PMID: 31007686 PMCID: PMC6441529 DOI: 10.1155/2019/2175273] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/06/2018] [Indexed: 12/20/2022] Open
Abstract
Study Design Isolation and characterization of human epidural fat (HEF) stem/progenitor cells. Objective To identify a progenitor population within HEF and to determine if they meet the minimal criteria of a mesenchymal stem cell (MSC). Summary of Background Data The biological function, if any, has yet to be determined for HEF. The presence of MSCs within HEF may indicate a regenerative potential within the HEF. Methods HEF was isolated from 10 patients during elective spinal surgery. HEF cells were differentiated along osteo-, adipo-, and chondrogenic lineages, with differentiation analyzed via qPCR and histology. The cell surface receptor profile of HEF cells was examined by flow cytometry. HEF cells were also assayed through the collagen contraction assay. Prx1CreERT2GFP:R26RTdTomato MSC lineage-tracking mice were employed to identify EF MSCs in vivo. Results HEF cell lines were obtained from all 10 patients in the study. Cells from 2/10 patients demonstrated full MSC potential, while cells from 6/10 patients demonstrated progenitor potential; 2/10 patients presented with cells that retained only adipogenic potential. HEF cells demonstrated MSC surface marker expression. All patient cell lines contracted collagen gels. A Prx1-positive population in mouse epidural fat that appeared to contribute to the dura of the spinal cord was observed in vivo. Conclusions MSC and progenitor populations are present within HEF. MSCs were not identified in all patients examined in the current study. Furthermore, all patient lines demonstrated collagen contraction capacity, suggesting either a contaminating activated fibroblast population or HEF MSCs/progenitors also demonstrating a fibroblast-like phenotype. In vivo analysis suggests that these cell populations may contribute to the dura. Overall, these results suggest that cells within epidural fat may play a biological role within the local environment above providing a mechanical buffer.
Collapse
|
37
|
Sun Y, Chen S, Pei M. Comparative advantages of infrapatellar fat pad: an emerging stem cell source for regenerative medicine. Rheumatology (Oxford) 2018; 57:2072-2086. [PMID: 29373763 PMCID: PMC6256334 DOI: 10.1093/rheumatology/kex487] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 11/13/2017] [Indexed: 12/15/2022] Open
Abstract
Growing evidence indicates that infrapatellar fat pad (IPFP)-derived stem cells (IPFSCs) exert robust proliferation capacities and multilineage differentiation potentials. However, few papers summarize the advantages that the IPFP and IPFSCs have in regenerative medicine. In this review we delineate the development and anatomy of the IPFP by comparing it with an adjacent fibrous tissue, synovium, and a more frequently harvested fat depot, subcutaneous adipose tissue. Furthermore, we explore the similarities and differences of stem cells from these three tissues in terms of IPFSCs, synovium-derived stem cells and subcutaneous adipose tissue-derived stem cells in proliferation capacity and tri-lineage differentiation potentials, including chondrogenesis, osteogenesis and adipogenesis. Finally, we highlight the advantages of IPFSCs in regenerative medicine, such as the abundant accessibility and the ability to resist inflammation and senescence, two hurdles for cell-based tissue regeneration. Considering the comparative advantages of IPFSCs, the IPFP can serve as an excellent stem cell source for regenerative medicine, particularly for cartilage regeneration.
Collapse
Affiliation(s)
- Yu Sun
- Department of Orthopaedics, Orthopaedics Institute, Subei People’s Hospital of Jiangsu Province, Yangzhou, Jiangsu, China
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
| | - Song Chen
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, USA
- Exercise Physiology, West Virginia University, Morgantown, WV, USA
- Mary Babb Randolph Cancer Center, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
38
|
Martin-Pena A, Porter R, Plumton G, McCarrel T, Morton A, Guijarro M, Ghivizzani S, Sharma B, Palmer G. Lentiviral-based reporter constructs for profiling chondrogenic activity in primary equine cell populations. Eur Cell Mater 2018; 36:156-170. [PMID: 30311630 PMCID: PMC6788286 DOI: 10.22203/ecm.v036a12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Successful clinical translation of mesenchymal stem cell (MSC)-based therapies for cartilage repair will likely require the implementation of standardised protocols and broadly applicable tools to facilitate the comparisons among cell types and chondroinduction methods. The present study investigated the utility of recombinant lentiviral reporter vectors as reliable tools for comparing chondrogenic potential among primary cell populations and distinguishing cellular-level variations of chondrogenic activity in widely used three-dimensional (3D) culture systems. Primary equine MSCs and chondrocytes were transduced with vectors containing combinations of fluorescent and luciferase reporter genes under constitutive cytomeglavirus (CMV) or chondrocyte-lineage (Col2) promoters. Reporter activity was measured by fluorescence imaging and luciferase assay. In 3D cultures of MSC aggregates and polyethylene glycol-hyaluronic acid (PEG-HA) hydrogels, transforming growth factor beta 3 (TGF-β3)-mediated chondroinduction increased Col2 reporter activity, demonstrating close correlation with histology and mRNA expression levels of COL2A1 and SOX9. Comparison of chondrogenic activities among MSC populations using a secretable luciferase reporter revealed enhanced chondrogenesis in bone-marrow-derived MSCs relative to MSC populations from synovium and adipose tissues. A dual fluorescence reporter - enabling discrimination of highly chondrogenic (Col2-GFP) cells within an MSC population (CMV-tdTomato) - revealed marked heterogeneity in differentiating aggregate cultures and identified chondrogenic cells in chondrocyte-seeded PEG-HA hydrogels after 6 weeks in a subcutaneous implant model - indicating stable, long-term reporter expression in vivo. These results suggested that lentiviral reporter vectors may be used to address fundamental questions regarding chondrogenic activity in chondroprogenitor cell populations and accelerate clinical translation of cell-based cartilage repair strategies.
Collapse
Affiliation(s)
- A. Martin-Pena
- Department of Orthopaedics and Rehabilitation, University
of Florida, Gainesville, Florida USA
| | - R.M. Porter
- Department of Orthopaedics, University of Arkansas, Little
Rock, Arkansas, USA
| | - G Plumton
- Department of Biomedical Engineering, University of
Florida, Gainesville, Florida USA
| | - T.M. McCarrel
- College of Veterinary Sciences, University of Florida,
Gainesville, Florida USA
| | - A.J. Morton
- College of Veterinary Sciences, University of Florida,
Gainesville, Florida USA
| | - M.V. Guijarro
- Department of Anatomy and Cell Biology, University of
Florida, Gainesville, Florida USA
| | - S.C. Ghivizzani
- Department of Orthopaedics and Rehabilitation, University
of Florida, Gainesville, Florida USA
| | - B. Sharma
- Department of Orthopaedics, University of Arkansas, Little
Rock, Arkansas, USA
| | - G.D. Palmer
- Department of Orthopaedics and Rehabilitation, University
of Florida, Gainesville, Florida USA,Address for correspondence: Glyn Palmer, Ph.D,
Dept of Orthopaedics and Rehabilitation, University of Florida, 1600 SW Archer
Rd, MSB, M2-235, Gainesville, FL 32610, Telephone: +1 352 273 7087, Fax: +1 352
273 7427,
| |
Collapse
|
39
|
Ye K, Traianedes K, Robins SA, Choong PFM, Myers DE. Osteochondral repair using an acellular dermal matrix-pilot in vivo study in a rabbit osteochondral defect model. J Orthop Res 2018; 36:1919-1928. [PMID: 29244224 DOI: 10.1002/jor.23837] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 12/04/2017] [Indexed: 02/04/2023]
Abstract
UNLABELLED The aim of this pilot project was to introduce a novel use of acellular dermal matrix (ADM) in combination with infrapatellar fat pad mesenchymal stromal cells (IPFP-MSCs) to effect repair in a rabbit osteochondral defect model. ADM, in a range of surgical procedures, has been shown to promote remodelling of tissue at the site of implantation. Rabbit-derived ADM (rabADM) was prepared from the skin of donor rabbits. Autologous IPFP-MSCs were obtained at the time of knee surgery. Osteochondral defects (4 mm cartilage outer/2 mm central bone defect) were drilled into distal femoral condyles of 12 New Zealand White rabbits. Treatments groups: (i) defect only; (ii) rabADM alone; (iii) IPFP-MSCs alone; and (iv) rabADM with IPFP-MSCs. Condyles were harvested at 12 weeks, and analyzed using histology, immunohistochemistry (types I and II collagen) and histomorphometry to evaluate osteochondral repair. The rabADM only group achieved the highest ratio of type II to non-type II collagen (77.3%) using areal measures (similar to normal cartilage), which indicated a higher quality of cartilage repair. The addition of IPFP-MSCs, with or without rabADM, formed a fibrous collagen cap above the lesion site not seen with rabADM alone. Macroscopically, there was no joint erosion, inflammation, swelling or deformity, and all animals maintained full range of motion. CONCLUSIONS RabADM alone resulted in neocartilage formation similar to native cartilage. IPFP-MSCs limited osteochondral repair and contributed to fibrosis, even in combination with the rabADM. Further studies using ADM for osteochondral repair are warranted in a more appropriate pre-clinical model of osteochondral repair. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1919-1928, 2018.
Collapse
Affiliation(s)
- Ken Ye
- Department of Surgery, University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Orthopaedics, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Kathy Traianedes
- Department of Clinical Neurosciences, St Vincent's Hospital Melbourne, Victoria Parade, Fitzroy, 3065, Australia.,Department of Medicine, University of Melbourne, St Vincent's Hospital, Fitzroy, Australia
| | - Shalley A Robins
- Department of Medicine, University of Melbourne, St Vincent's Hospital, Fitzroy, Australia
| | - Peter F M Choong
- Department of Surgery, University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, Australia.,Department of Orthopaedics, St Vincent's Hospital Melbourne, Fitzroy, Australia
| | - Damian E Myers
- Department of Medicine, University of Melbourne, St Vincent's Hospital, Fitzroy, Australia.,Victoria University, Sunshine Hospital, St Albans, Australia.,Australian Institute for Musculoskeletal Science, Victoria University and The University of Melbourne, Western Centre for Health and Research Education, Sunshine Hospital, St Albans, Australia
| |
Collapse
|
40
|
Sun Y, Yan L, Chen S, Pei M. Functionality of decellularized matrix in cartilage regeneration: A comparison of tissue versus cell sources. Acta Biomater 2018; 74:56-73. [PMID: 29702288 PMCID: PMC7307012 DOI: 10.1016/j.actbio.2018.04.048] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 01/12/2023]
Abstract
Increasing evidence indicates that decellularized extracellular matrices (dECMs) derived from cartilage tissues (T-dECMs) or chondrocytes/stem cells (C-dECMs) can support proliferation and chondrogenic differentiation of cartilage-forming cells. However, few review papers compare the differences between these dECMs when they serve as substrates for cartilage regeneration. In this review, after an introduction of cartilage immunogenicity and decellularization methods to prepare T-dECMs and C-dECMs, a comprehensive comparison focuses on the effects of T-dECMs and C-dECMs on proliferation and chondrogenic differentiation of chondrocytes/stem cells in vitro and in vivo. Key factors within dECMs, consisting of microarchitecture characteristics and micromechanical properties as well as retained insoluble and soluble matrix components, are discussed in-depth for potential mechanisms underlying the functionality of these dECMs in regulating chondrogenesis. With this information, we hope to benefit dECM based cartilage engineering and tissue regeneration for future clinical application. STATEMENT OF SIGNIFICANCE The use of decellularized extracellular matrix (dECM) is becoming a promising approach for tissue engineering and regeneration. Compared to dECM derived from cartilage tissue, recently reported dECM from cell sources exhibits a distinct role in cell based cartilage regeneration. In this review paper, for the first time, tissue and cell based dECMs are comprehensively compared for their functionality in cartilage regeneration. This information is expected to provide an update for dECM based cartilage regeneration.
Collapse
Affiliation(s)
- Yu Sun
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Department of Orthopaedics, Orthopaedics Institute, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, China
| | - Lianqi Yan
- Department of Orthopaedics, Orthopaedics Institute, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu 225001, China
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan 610083, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV 26506, USA; Exercise Physiology, West Virginia University, Morgantown, WV 26506, USA; WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV 26506, USA.
| |
Collapse
|
41
|
Chang J, Liao Z, Lu M, Meng T, Han W, Ding C. Systemic and local adipose tissue in knee osteoarthritis. Osteoarthritis Cartilage 2018; 26:864-871. [PMID: 29578044 DOI: 10.1016/j.joca.2018.03.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 02/28/2018] [Accepted: 03/15/2018] [Indexed: 02/08/2023]
Abstract
Knee osteoarthritis (OA) is the most common joint disease. Body adipose tissue has been shown to be related to the development and progression of knee OA. Among systemic adipose tissues, subcutaneous adipose tissue is significantly and negatively associated with muscle mass and forces, and could be related to the presence and progression of knee OA. Visceral adipose tissue is associated with increased cartilage loss and production of pro-inflammatory cytokines. Intra-muscular adipose tissue is associated with knee osteoarthritic changes, but it remains controversial if inter-muscular adipose tissue has a role to play in the pathogenesis for knee OA. Knee local adipose tissue such as infrapatellar fat pad (IPFP) can interact with neighbouring tissues, and may have a biphasic effect in knee OA. The underlying mechanisms for the roles of the systemic and local fat in knee OA could be related to biomechanical, metabolic, inflammatory factors and fat fibrosis, which may have a separated or combined effect on OA. Tissue engineering from systemic or local adipose tissue is a new research direction, and adipose tissue-derived stem cells from systemic or local adipose tissue may be beneficial for OA cartilage repair. Research on systemic and local adipose tissue would provide novel approaches for prevention and treatment of knee OA, but further studies are required to explore the roles of different adipose tissues in knee OA and the effects of stem cells derived from different adipose tissues on knee OA.
Collapse
Affiliation(s)
- J Chang
- Department of Orthopaedics, 4th Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China; Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Z Liao
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Rheumatology and Immunology Division, 3rd Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - M Lu
- Department of Orthopaedics, 1st Affiliated Hospital, Anhui Medical University, Hefei, Anhui, China
| | - T Meng
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - W Han
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - C Ding
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia; Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
42
|
Bhattacharjee A, Katti DS. Pore Alignment in Gelatin Scaffolds Enhances Chondrogenic Differentiation of Infrapatellar Fat Pad Derived Mesenchymal Stromal Cells. ACS Biomater Sci Eng 2018; 5:114-125. [DOI: 10.1021/acsbiomaterials.8b00246] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Arijit Bhattacharjee
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology—Kanpur, Kanpur 208016, Uttar Pradesh, India
| | - Dhirendra S Katti
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology—Kanpur, Kanpur 208016, Uttar Pradesh, India
| |
Collapse
|
43
|
De Bari C, Roelofs AJ. Stem cell-based therapeutic strategies for cartilage defects and osteoarthritis. Curr Opin Pharmacol 2018; 40:74-80. [PMID: 29625333 DOI: 10.1016/j.coph.2018.03.009] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 03/20/2018] [Indexed: 02/08/2023]
Abstract
The gold standard cell therapy for repair of articular cartilage defects is autologous chondrocyte implantation, with good outcomes long-term. Mesenchymal stromal/stem cells (MSCs) from bone marrow or connective tissues such as fat are being pursued as alternatives for cartilage repair, and are trialled via intra-articular administration in patients with knee osteoarthritis. Early-phase clinical studies concur on safety and provide some promising insight into efficacy, but the mechanism of action remains unclear. Recent studies implicate extracellular vesicles as important mediators of MSC action, offering exciting therapeutic prospects. Our increasing understanding of the mechanisms underlying intrinsic articular cartilage maintenance and repair fosters hope that novel/repurposed therapeutics could elicit repair through activation of endogenous stem/progenitor cells to maintain healthy joints and prevent osteoarthritis.
Collapse
Affiliation(s)
- Cosimo De Bari
- Arthritis & Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, UK.
| | - Anke J Roelofs
- Arthritis & Regenerative Medicine Laboratory, Aberdeen Centre for Arthritis and Musculoskeletal Health, Institute of Medical Sciences, University of Aberdeen, UK
| |
Collapse
|
44
|
Current Therapeutic Strategies for Stem Cell-Based Cartilage Regeneration. Stem Cells Int 2018; 2018:8490489. [PMID: 29765426 PMCID: PMC5889878 DOI: 10.1155/2018/8490489] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 12/14/2017] [Accepted: 01/23/2018] [Indexed: 12/13/2022] Open
Abstract
The process of cartilage destruction in the diarthrodial joint is progressive and irreversible. This destruction is extremely difficult to manage and frustrates researchers, clinicians, and patients. Patients often take medication to control their pain. Surgery is usually performed when pain becomes uncontrollable or joint function completely fails. There is an unmet clinical need for a regenerative strategy to treat cartilage defect without surgery due to the lack of a suitable regenerative strategy. Clinicians and scientists have tried to address this using stem cells, which have a regenerative potential in various tissues. Cartilage may be an ideal target for stem cell treatment because it has a notoriously poor regenerative potential. In this review, we describe past, present, and future strategies to regenerate cartilage in patients. Specifically, this review compares a surgical regenerative technique (microfracture) and cell therapy, cell therapy with and without a scaffold, and therapy with nonaggregated and aggregated cells. We also review the chondrogenic potential of cells according to their origin, including autologous chondrocytes, mesenchymal stem cells, and induced pluripotent stem cells.
Collapse
|
45
|
Combating Osteoarthritis through Stem Cell Therapies by Rejuvenating Cartilage: A Review. Stem Cells Int 2018; 2018:5421019. [PMID: 29765416 PMCID: PMC5885495 DOI: 10.1155/2018/5421019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 02/05/2018] [Indexed: 12/13/2022] Open
Abstract
Knee osteoarthritis (OA) is a chronic degenerative disorder which could be distinguished by erosion of articular cartilage, pain, stiffness, and crepitus. Not only aging-associated alterations but also the metabolic factors such as hyperglycemia, dyslipidemia, and obesity affect articular tissues and may initiate or exacerbate the OA. The poor self-healing ability of articular cartilage due to limited regeneration in chondrocytes further adversely affects the osteoarthritic microenvironment. Traditional and current surgical treatment procedures for OA are limited and incapable to reverse the damage of articular cartilage. To overcome these limitations, cell-based therapies are currently being employed to repair and regenerate the structure and function of articular tissues. These therapies not only depend upon source and type of stem cells but also on environmental conditions, growth factors, and chemical and mechanical stimuli. Recently, the pluripotent and various multipotent mesenchymal stem cells have been employed for OA therapy, due to their differentiation potential towards chondrogenic lineage. Additionally, the stem cells have also been supplemented with growth factors to achieve higher healing response in osteoarthritic cartilage. In this review, we summarized the current status of stem cell therapies in OA pathophysiology and also highlighted the potential areas of further research needed in regenerative medicine.
Collapse
|
46
|
Shimomura K, Ando W, Fujie H, Hart DA, Yoshikawa H, Nakamura N. Scaffold-free tissue engineering for injured joint surface restoration. J Exp Orthop 2018; 5:2. [PMID: 29330730 PMCID: PMC5768574 DOI: 10.1186/s40634-017-0118-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 12/20/2017] [Indexed: 12/31/2022] Open
Abstract
Articular cartilage does not heal spontaneously due to its limited healing capacity, and thus effective treatments for cartilage injuries has remained challenging. Since the first report by Brittberg et al. in 1994, autologous chondrocyte implantation (ACI) has been introduced into the clinic. Recently, as an alternative for chondrocyte-based therapy, mesenchymal stem cell (MSC)-based therapy has received considerable research attention because of the relative ease in handling for tissue harvest, and subsequent cell expansion and differentiation. In this review, we discuss the latest developments regarding stem cell-based therapies for cartilage repair, with special focus on recent scaffold-free approaches.
Collapse
Affiliation(s)
- Kazunori Shimomura
- Medicine for Sports and Performing Arts, Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita City, Osaka, 565-0871, Japan.,Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Wataru Ando
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Hiromichi Fujie
- Division of Human Mechatronics Systems, Faculty of System Design, Tokyo Metropolitan University, 6-6 Asahigaoka, Hino City, Tokyo, 191-0065, Japan
| | - David A Hart
- McCaig Institute for Bone & Joint Health, University of Calgary, 3330 Hospital Drive Northwest, Calgary, AB, T2N 4N1, Canada
| | - Hideki Yoshikawa
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita City, Osaka, 565-0871, Japan
| | - Norimasa Nakamura
- Institute for Medical Science in Sports, Osaka Health Science University, 1-9-27 Tenma, Kita-ku, Osaka City, Osaka, 530-0043, Japan. .,Center for Advanced Medical Engineering and Informatics, Osaka University, 2-2 Yamadaoka, Suita City, Osaka, 565-0871, Japan.
| |
Collapse
|
47
|
Armiento AR, Stoddart MJ, Alini M, Eglin D. Biomaterials for articular cartilage tissue engineering: Learning from biology. Acta Biomater 2018; 65:1-20. [PMID: 29128537 DOI: 10.1016/j.actbio.2017.11.021] [Citation(s) in RCA: 363] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/05/2017] [Accepted: 11/07/2017] [Indexed: 12/27/2022]
Abstract
Articular cartilage is commonly described as a tissue that is made of up to 80% water, is devoid of blood vessels, nerves, and lymphatics, and is populated by only one cell type, the chondrocyte. At first glance, an easy tissue for clinicians to repair and for scientists to reproduce in a laboratory. Yet, chondral and osteochondral defects currently remain an open challenge in orthopedics and tissue engineering of the musculoskeletal system, without considering osteoarthritis. Why do we fail in repairing and regenerating articular cartilage? Behind its simple and homogenous appearance, articular cartilage hides a heterogeneous composition, a high level of organisation and specific biomechanical properties that, taken together, make articular cartilage a unique material that we are not yet able to repair or reproduce with high fidelity. This review highlights the available therapies for cartilage repair and retraces the research on different biomaterials developed for tissue engineering strategies. Their potential to recreate the structure, including composition and organisation, as well as the function of articular cartilage, intended as cell microenvironment and mechanically competent replacement, is described. A perspective of the limitations of the current research is given in the light of the emerging technologies supporting tissue engineering of articular cartilage. STATEMENT OF SIGNIFICANCE The mechanical properties of articular tissue reflect its functionally organised composition and the recreation of its structure challenges the success of in vitro and in vivo reproduction of the native cartilage. Tissue engineering and biomaterials science have revolutionised the way scientists approach the challenge of articular cartilage repair and regeneration by introducing the concept of the interdisciplinary approach. The clinical translation of the current approaches are not yet fully successful, but promising results are expected from the emerging and developing new generation technologies.
Collapse
Affiliation(s)
- A R Armiento
- AO Research Institute Davos, Davos Platz, Switzerland.
| | - M J Stoddart
- AO Research Institute Davos, Davos Platz, Switzerland; University Medical Center, Albert-Ludwigs University, Freiburg, Germany.
| | - M Alini
- AO Research Institute Davos, Davos Platz, Switzerland.
| | - D Eglin
- AO Research Institute Davos, Davos Platz, Switzerland.
| |
Collapse
|
48
|
Ma N, Wang H, Xu X, Wan Y, Liu Y, Wang M, Yu W, Dai Y, Peng J, Guo Q, Yu C, Lu S. Autologous-cell-derived, tissue-engineered cartilage for repairing articular cartilage lesions in the knee: study protocol for a randomized controlled trial. Trials 2017; 18:519. [PMID: 29110690 PMCID: PMC5674846 DOI: 10.1186/s13063-017-2251-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 10/07/2017] [Indexed: 12/22/2022] Open
Abstract
Background Spontaneous recovery from articular cartilage injury is difficult, and the ongoing progression of disease can eventually lead to osteoarthritis. Currently, there is no effective non-surgical treatment for articular cartilage injury. Arthroscopic debridement and microfracture surgery are performed for fibrocartilage repair. But fibrocartilage is different from normal articular cartilage, and functional recovery is not satisfactory. Therefore, it is necessary to develop more effective techniques for articular cartilage repair. Progress in material science, cell biology, biomechanics, and bioreactor technology has allowed the development of biomimetic, tissue-engineered osteochondral composites that have shown potential for the repair of damaged cartilage. We prepared biomimetic, tissue-engineered cartilage scaffolds optimized for biochemical composition and structural characteristics. Based on the experience of our pre-clinical studies on animals, a human articular cartilage acellular matrix scaffold was prepared and is in clinical use. The combination of autologous chondrocytes and scaffolds has shown satisfactory results in repairing cartilage defects in preliminary experiments. Methods This is a prospective randomized controlled trial. One hundred patients with full-thickness cartilage injury of the knee will be randomly divided into two groups to receive treatment with either tissue-engineered cartilage constructed using biomimetic cartilage extracellular-matrix-oriented scaffolds combined with autologous chondrocytes, or arthroscopic debridement and microfracture surgery. There will be five visiting time points: at baseline, then at 3, 6, 12, and 18 months postoperatively. The primary outcome will be therapeutic efficacy as assessed by the Lysholm score at 12 months postoperatively. The secondary outcomes will be the International Knee Documentation Committee score, Visual Analog Scale score, and cartilage injury and repair as assessed by magnetic resonance imaging as well as the incidence of postoperative adverse events. Discussion This trial will attempt to verify the use of tissue-engineered cartilage constructed using autologous chondrocytes combined with allogeneic, acellular cartilage matrix for the repair of cartilage defects, thereby providing favorable evidence for its use in clinical practice. Trial registration ClinicalTrials.gov, identifier: NCT02770209. Registered on 11 May 2016. Electronic supplementary material The online version of this article (doi:10.1186/s13063-017-2251-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ning Ma
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Hongxia Wang
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Xian Xu
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Yiqun Wan
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Yufeng Liu
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Mingjie Wang
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Wen Yu
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Yongjing Dai
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Jiang Peng
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Quanyi Guo
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China.
| | - Changlong Yu
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Shibi Lu
- Institute of Orthopaedics, Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| |
Collapse
|
49
|
Budd E, Waddell S, de Andrés MC, Oreffo ROC. The Potential of microRNAs for Stem Cell-based Therapy for Degenerative Skeletal Diseases. ACTA ACUST UNITED AC 2017; 3:263-275. [PMID: 29214143 PMCID: PMC5700219 DOI: 10.1007/s40610-017-0076-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose of Review Degenerative skeletal disorders including osteoarthritis (OA) and osteoporosis (OP) are the result of attenuation of tissue regeneration and lead to painful conditions with limited treatment options. Preventative measures to limit the onset of OA and OP remain a significant unmet clinical need. MicroRNAs (miRNAs) are known to be involved in the differentiation of stem cells, and in combination with stem cell therapy could induce skeletal regeneration and potentially prevent OA and OP onset. Recent Findings The combination of stem cells and miRNA has been successful at regenerating the bone and cartilage in vivo. MiRNAs, including miR-146b known to be involved in chondrogenic differentiation, could provide innovative targets for stem cell-based therapy, for the repair of articular cartilage defects forestalling the onset of OA or in the generation of a stem cell-based therapy for OP. Summary This review discusses the combination of skeletal stem cells (SSCs) and candidate miRNAs for application in a cell-based therapy approach for skeletal regenerative medicine.
Collapse
Affiliation(s)
- Emma Budd
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - Shona Waddell
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - María C de Andrés
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| | - Richard O C Oreffo
- Bone & Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD UK
| |
Collapse
|
50
|
Physiology of ageing of the musculoskeletal system. Best Pract Res Clin Rheumatol 2017; 31:203-217. [PMID: 29224697 DOI: 10.1016/j.berh.2017.09.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 09/07/2017] [Indexed: 02/06/2023]
Abstract
This review aims to provide a summary of current concepts of ageing in relation to the musculoskeletal system, highlighting recent advances in the understanding of the mechanisms involved in the development of age-related changes in bone, skeletal muscle, chondroid and fibrous tissues. The key components of the musculoskeletal system and their functions are introduced together with a general overview of the molecular hallmarks of ageing. A brief description of the normal architecture of each of these tissue types is followed by a summary of established and developing concepts of mechanisms contributing to the age-related alterations in each. Extensive detailed description of these changes is beyond the scope of this review; instead, we aim to highlight some of the most significant processes and, where possible, the molecular changes underlying these and refer the reader to in-depth, subspecialist reviews of the individual components for further details.
Collapse
|