51
|
Grogan SP, Duffy SF, Pauli C, Lotz MK, D’Lima DD. Gene expression profiles of the meniscus avascular phenotype: A guide for meniscus tissue engineering. J Orthop Res 2018; 36:1947-1958. [PMID: 29411909 PMCID: PMC6326361 DOI: 10.1002/jor.23864] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 02/05/2018] [Indexed: 02/04/2023]
Abstract
Avascular (Avas) meniscus regeneration remains a challenge, which is partly a consequence of our limited knowledge of the cells that maintain this tissue region. In this study, we utilized microarrays to characterize gene expression profiles of intact human Avas meniscus tissue and of cells following culture expansion. Using these data, we examined various 3D culture conditions to redifferentiate Avas cells toward the tissue phenotype. RNA was isolated from either the tissue directly or following cell isolation and 2 weeks in monolayer culture. RNA was hybridized on human genome arrays. Differentially expressed (DE) genes were identified by ranking analysis. DAVID pathway analysis was performed and visualized using STRING analysis. Quantitative PCR (qPCR) on additional donor menisci (tissues and cells) were used to validate array data. Avas cells cultured in 3D were subjected to qPCR to compare with the array-generated data. A total of 387 genes were DE based on differentiation state (>3-fold change; p < 0.01). In Avas-cultured cells, the upregulated pathways included focal adhesion, ECM-receptor interaction, regulation of actin cytoskeleton, and PDGF Signaling. In 3D-cultured Avas cells, TGFβ1 or combinations of TGFβ1 and BMP6 were most effective to promote an Avas tissue phenotype. THBS2 and THBS4 expression levels were identified as a means to denote meniscus cell phenotype status. We identified the key gene expression profiles, new markers and pathways involved in characterizing the Avas meniscus phenotype in the native state and during in vitro dedifferentiation and redifferentiation. These data served to screen 3D conditions to generate meniscus-like neotissues. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1947-1958, 2018.
Collapse
Affiliation(s)
- Shawn P Grogan
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA
| | - Stuart F. Duffy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Chantal Pauli
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Martin K Lotz
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA
| | - Darryl D D’Lima
- Shiley Center for Orthopaedic Research and Education at Scripps Clinic, La Jolla, CA,Corresponding author: Darryl D D’Lima, MD, PhD, Shiley Center for Orthopaedic Research and Education at Scripps Clinic, 11025 North Torrey Pines Road, Suite 200, La Jolla, CA 92037, Tel 858 332 0166 Fax 858 332 0669,
| |
Collapse
|
52
|
Costa JB, Silva-Correia J, Reis RL, Oliveira JM. Recent advances on 3D printing of patient-specific implants for fibrocartilage tissue regeneration. ACTA ACUST UNITED AC 2018. [DOI: 10.2217/3dp-2018-0006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Fibrocartilage is a very peculiar type of tissue found in intervertebral disc and meniscus. It is characterized by its avascular nature and for the shear and compressive forces that it can be subjected to. The number of individuals affected by the degeneration of fibrocartilaginous tissues has been growing and the poor outcomes of current treatments have led to an increased interest in new alternative approaches. Therefore, the combination of reverse engineering with 3D printing has been extensively explored in order to produce patient-specific implants capable of improving the current clinical outcomes. This review outlines the recent advances achieved in the tissue engineering field, especially focusing on fibrocartilaginous tissue.
Collapse
Affiliation(s)
- João B Costa
- 3B’s Research Group – Biomaterials, Biodegradables & Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering & Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative & Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal
| | - Joana Silva-Correia
- 3B’s Research Group – Biomaterials, Biodegradables & Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering & Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B’s Research Group – Biomaterials, Biodegradables & Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering & Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative & Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal
| | - Joaquim M Oliveira
- 3B’s Research Group – Biomaterials, Biodegradables & Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering & Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative & Precision Medicine, Headquarters at University of Minho, Avepark, 4805-017 Barco, Guimarães, Portugal
| |
Collapse
|
53
|
Coluccino L, Gottardi R, Ayadi F, Athanassiou A, Tuan RS, Ceseracciu L. Porous Poly(vinyl alcohol)-Based Hydrogel for Knee Meniscus Functional Repair. ACS Biomater Sci Eng 2018; 4:1518-1527. [PMID: 33445309 DOI: 10.1021/acsbiomaterials.7b00879] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The meniscus has a key role within the knee joint, conferring stability, absorbing and redistributing loads, and influencing the overall movement proprioception. Recent developments in the treatment of meniscal injury have progressively shifted the focus from general resection to functional repair, with the recognition that restoring the biomechanical meniscal function helps to prevent degenerative changes in the knee joint and the insurgence of osteoarthritis. To address this clinical need, we have developed a biomimetic implant based on a porous poly(vinyl alcohol) (PVA) hydrogel. Such hydrogels are stable, biocompatible, and suitable to surgical translation, and their mechanical properties can be tuned to reduce the mismatch in the case of partial meniscectomy. The PVA implant structure is porous and permeable, allowing fluid flows and facilitating anatomical integration in situ. Here, we present a chemo-physical characterization of PVA porous hydrogels, focusing on their tunable morphology and associated viscoelastic properties. Biocompatibility was evaluated using primary bovine meniscal fibrochondrocytes, and integration with native tissues was assessed in an ex vivo model. Overall, our results suggest that a synthetic meniscal implant based on a porous PVA hydrogel could restore the physiological function of the meniscus and represent a promising clinical alternative to current resection treatments.
Collapse
Affiliation(s)
- Luca Coluccino
- Department of Orthopaedic Surgery, Department of Chemical Engineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | - Riccardo Gottardi
- Department of Orthopaedic Surgery, Department of Chemical Engineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States.,Ri.MED Foundation, Palermo 90133, Italy
| | - Farouk Ayadi
- UNIROUEN, INSA Rouen, CNRS, PBS, Normandie Universite, 76000 Rouen, France
| | | | - Rocky S Tuan
- Department of Orthopaedic Surgery, Department of Chemical Engineering, and the McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15213, United States
| | | |
Collapse
|
54
|
Xie J, Zhang D, Lin Y, Yuan Q, Zhou X. Anterior Cruciate Ligament Transection-Induced Cellular and Extracellular Events in Menisci: Implications for Osteoarthritis. Am J Sports Med 2018. [PMID: 29513553 DOI: 10.1177/0363546518756087] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND The meniscus plays an important role in knee joint diseases such as osteoarthritis (OA). Meniscal injuries can be accompanied by joint catabolic events initiated by inflammation, leading to articular cartilage destruction, but the cellular events responsible for intrinsic meniscal injury and the extracellular matrix changes necessary for meniscal degradation are not well known. PURPOSE To explore the cellular and matrix-related changes of menisci based on a mouse OA model of anterior cruciate ligament transection (ACLT). STUDY DESIGN Controlled laboratory study. METHODS A mouse ACLT OA model was established by transection of anterior cruciate ligaments on the right knee joints of 8-week-old male (n = 34) and female (n = 34) C57 mice. The knee joints were collected at 1, 2, 4, and 8 weeks after ACLT surgery, and the meniscal changes were analyzed by radiography, histology, immunohistochemistry, immunoblot, and quantitative real-time polymerase chain reaction. RESULTS The deterioration of menisci was more extensive than that of articular cartilage and subchondral bone at 4 weeks after ACLT surgery. The rapid loss of collagen II and Sox9 in chondrocyte-like cells in the white-white zone of menisci was confirmed, and the activation of potential meniscus progenitor cells and chondroblasts was identified based on the increase of CD90, CD105, and Runx2. Further, the intrinsic inflammation in the bone marrow-like zone of menisci was activated by enhancement of dendritic cells (CD11c+), T cells (CD3+), and macrophages (F4/80+) with the increase of the inflammatory factors interleukin 1β and tumor necrosis factor α. Finally, the extracellular matrix events involving changes in chemokines, increases of matrix proteases (matrix metalloproteinases and ADAMTS5), and decreases of lysyl oxidase family were elucidated. CONCLUSION ACLT-induced meniscal changes not only could explain the contribution of the meniscus to the progress of OA but also could provide a cue for initiation of preventive treatments in the early stages of OA. CLINICAL RELEVANCE This study provides support for better protection of menisci in ACL injury-induced conditions such as OA and indicates that menisci should be considered in the development of clinical pharmacological interventions.
Collapse
Affiliation(s)
- Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
55
|
Huang H, Xu H, Zhao J. A Novel Approach for Meniscal Regeneration Using Kartogenin-Treated Autologous Tendon Graft. Am J Sports Med 2017; 45:3289-3297. [PMID: 28859517 DOI: 10.1177/0363546517721192] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND The meniscus is one of the most commonly injured parts of the body, and meniscal healing is difficult. HYPOTHESIS Kartogenin (KGN) induces tendon stem cells (TSCs) to differentiate into cartilage cells in vitro and form meniscus-like tissue in vivo. A damaged meniscus can be replaced with a KGN-treated autologous tendon graft. STUDY DESIGN Controlled laboratory study. METHODS In the in vitro experiments, TSCs were isolated from rabbit patellar tendons and cultured with various concentrations of KGN, from 0 to 1000 µM. The effect of KGN on the chondrogenesis of TSCs in vitro was investigated by histochemical staining and quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR). The in vivo experiments were carried out on 6 New Zealand White rabbits by removing a meniscus from the rabbit knee and implanting an autologous tendon graft treated with KGN or saline. The meniscus formation in vivo was examined by histological analysis and immune staining. RESULTS The proliferation of TSCs was promoted by KGN in a concentration-dependent manner. Both histochemical staining and qRT-PCR showed that the chondrogenic differentiation of TSCs was increased with KGN concentration. After 3 months of implantation, the tendon graft treated with KGN formed a meniscus-like tissue with a white and glistening appearance, while the saline-treated tendon graft retained tendon-like tissue and appeared yellowish and unhealthy. Histochemical staining showed that after 3 months of implantation, the KGN-treated tendon graft had a structure similar to that of normal meniscus. Many cartilage-like cells and fibrocartilage-like tissues were found in the KGN-treated tendon graft. However, no cartilage-like cells were found in the saline-treated tendon graft after 3 months of implantation. Furthermore, the KGN-treated tendon graft was positively stained by both anti-collagen type I and type II antibodies, but the saline-treated tendon graft was not stained by collagen type II. CONCLUSION The findings indicated that KGN can induce the differentiation of TSCs into cartilage-like cells in vitro and in vivo. The results suggest that KGN-treated tendon graft may be a good substitute for meniscal repair and regeneration. CLINICAL RELEVANCE This study revealed the direct effects of KGN on the chondrogenic differentiation of TSCs in vitro and in vivo. A KGN-treated autologous tendon graft induced formation of a meniscus-like tissue in vivo. This study provides a new cartilage regenerating technology for the treatment of damaged meniscus.
Collapse
Affiliation(s)
- He Huang
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Hongyao Xu
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Jianning Zhao
- Department of Orthopaedics, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.,Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
56
|
Terai S, Hashimoto Y, Orita K, Yamasaki S, Takigami J, Shinkuma T, Teraoka T, Nishida Y, Takahashi M, Nakamura H. The origin and distribution of CD68, CD163, and αSMA + cells in the early phase after meniscal resection in a parabiotic rat model. Connect Tissue Res 2017; 58:562-572. [PMID: 28165810 DOI: 10.1080/03008207.2017.1284825] [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: 02/03/2023]
Abstract
We previously reported that circulating peripheral blood-borne cells (PBCs) contribute to early-phase meniscal reparative change. Because macrophages and myofibroblasts are important contributors of tissue regeneration, we examined their origin and distribution in the reparative meniscus. Reparative menisci were evaluated at 1, 2, and 4 weeks post-meniscectomy by immunohistochemistry to locate monocytes and macrophages (stained positive for CD68 and CD163), and myofibroblasts (stained positive for αSMA). Of the total number of cells, 13% were CD68+ at 1 week post-meniscectomy, which decreased to 1% by 4 weeks post-meniscectomy; of these, almost half of CD68+ cells (49.4%: 98.8% as PBCs) were green fluorescent protein (GFP)-positive post-meniscectomy (1, 2, and 4 weeks), indicating that the majority of CD68+ cells were derived from PBCs. Of the total cells, 6% were CD163+ at 1 week post-meniscectomy, which decreased to 1% by week 4. Of the CD163+ cells, the majority were GFP-positive (42.5%: 85.0% as PBCs) after 1 week; however, this decreased significantly over time, which indicates that the majority of CD163+ cells are derived from PBCs during the early phase of meniscal reparative change, but are derived from resident cells at later time points. Of the total cells, 38% were αSMA+ at 1 week post-meniscectomy, which decreased to 3% by 4 weeks. The proportion of GFP-positive αSMA+ cells was 2.8% after 1 week, with no significant change over time, which indicates that the majority of αSMA+ cells originated from resident cells. Here, we describe the origin and distribution of macrophages and myofibroblasts during meniscal reparative change.
Collapse
Affiliation(s)
- Shozaburo Terai
- a Department of Orthopaedic Surgery , Osaka City University Graduate School of Medicine , Osaka , Japan
| | - Yusuke Hashimoto
- a Department of Orthopaedic Surgery , Osaka City University Graduate School of Medicine , Osaka , Japan
| | - Kumi Orita
- a Department of Orthopaedic Surgery , Osaka City University Graduate School of Medicine , Osaka , Japan
| | - Shinya Yamasaki
- b Department of Orthopaedic Surgery , Osaka City General Hospital , Osaka , Japan
| | - Junsei Takigami
- c Department of Orthopaedic Surgery , Shimada Hospital , Habikino , Japan
| | - Takafumi Shinkuma
- a Department of Orthopaedic Surgery , Osaka City University Graduate School of Medicine , Osaka , Japan
| | - Takanori Teraoka
- a Department of Orthopaedic Surgery , Osaka City University Graduate School of Medicine , Osaka , Japan
| | - Yohei Nishida
- a Department of Orthopaedic Surgery , Osaka City University Graduate School of Medicine , Osaka , Japan
| | - Masafumi Takahashi
- d Division of Inflammation Research, Centre for Molecular Medicine , Jichi Medical University , Shimotsuke , Japan
| | - Hiroaki Nakamura
- a Department of Orthopaedic Surgery , Osaka City University Graduate School of Medicine , Osaka , Japan
| |
Collapse
|
57
|
Critchley SE, Kelly DJ. Bioinks for bioprinting functional meniscus and articular cartilage. ACTA ACUST UNITED AC 2017. [DOI: 10.2217/3dp-2017-0012] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
3D bioprinting can potentially enable the engineering of biological constructs mimicking the complex geometry, composition, architecture and mechanical properties of different tissues and organs. Integral to the successful bioprinting of functional articular cartilage and meniscus is the identification of suitable bioinks and cell sources to support chondrogenesis or fibrochondrogenesis, respectively. Such bioinks must also possess the appropriate rheological properties to be printable and support the generation of complex geometries. This review will outline the parameters required to develop bioinks for such applications and the current recent advances in 3D bioprinting of functional meniscus and articular cartilage. The paper will conclude by discussing key scientific and technical hurdles in this field and by defining future research directions for cartilage and meniscus bioprinting.
Collapse
Affiliation(s)
- Susan E Critchley
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical & Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
| | - Daniel J Kelly
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
- Department of Mechanical & Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin, Ireland
- Department of Anatomy, Royal College of Surgeons in Ireland, Dublin, Ireland
- Advanced Materials & Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland & Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
58
|
Liang Y, Idrees E, Andrews SHJ, Labib K, Szojka A, Kunze M, Burbank AD, Mulet-Sierra A, Jomha NM, Adesida AB. Plasticity of Human Meniscus Fibrochondrocytes: A Study on Effects of Mitotic Divisions and Oxygen Tension. Sci Rep 2017; 7:12148. [PMID: 28939894 PMCID: PMC5610182 DOI: 10.1038/s41598-017-12096-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 09/04/2017] [Indexed: 02/07/2023] Open
Abstract
Meniscus fibrochondrocytes (MFCs) may be the optimal cell source to repair non-healing meniscus injuries using tissue engineering strategies. In this study, we investigated the effects of mitotic divisions and oxygen tension on the plasticity of adult human MFCs. Our assessment techniques included gene expression, biochemical, histological, and immunofluorescence assays. MFCs were expanded in monolayer culture with combined growth factors TGFβ1 and FGF-2 (T1F2) under normoxia (21% O2). Trilineage (adipogenesis, chondrogenesis and osteogenesis) differentiation was performed under both normoxic (21% O2) and hypoxic (3% O2) conditions. The data demonstrated that MFCs with a mean total population doubling of 10 can undergo adipogenesis and chondrogenesis. This capability was enhanced under hypoxic conditions. The MFCs did not undergo osteogenesis. In conclusion, our findings suggest that extensively expanded human MFCs have the capacity to generate tissues with the functional matrix characteristics of avascular meniscus. To this end, expanded MFCs may be an ideal cell source for engineering functional constructs for the replacement or repair of avascular meniscus.
Collapse
Affiliation(s)
- Yan Liang
- University of Alberta, Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Edmonton, T6G 2E1, Canada
- Division of Burn and Reconstructive Surgery, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, People's Republic of China
| | - Enaam Idrees
- University of Alberta, Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Edmonton, T6G 2E1, Canada
| | - Stephen H J Andrews
- University of Alberta, Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Edmonton, T6G 2E1, Canada
| | - Kirollos Labib
- University of Alberta, Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Edmonton, T6G 2E1, Canada
| | - Alexander Szojka
- University of Alberta, Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Edmonton, T6G 2E1, Canada
| | - Melanie Kunze
- University of Alberta, Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Edmonton, T6G 2E1, Canada
| | - Andrea D Burbank
- University of Alberta, Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Edmonton, T6G 2E1, Canada
| | - Aillette Mulet-Sierra
- University of Alberta, Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Edmonton, T6G 2E1, Canada
| | - Nadr M Jomha
- University of Alberta, Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Edmonton, T6G 2E1, Canada
| | - Adetola B Adesida
- University of Alberta, Department of Surgery, Divisions of Orthopaedic Surgery and Surgical Research, Edmonton, T6G 2E1, Canada.
| |
Collapse
|
59
|
Seol D, Zhou C, Brouillette MJ, Song I, Yu Y, Choe HH, Lehman AD, Jang KW, Fredericks DC, Laughlin BJ, Martin JA. Characteristics of meniscus progenitor cells migrated from injured meniscus. J Orthop Res 2017; 35:1966-1972. [PMID: 27813166 PMCID: PMC6354255 DOI: 10.1002/jor.23472] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 10/28/2016] [Indexed: 02/04/2023]
Abstract
Serious meniscus injuries seldom heal and increase the risk for knee osteoarthritis; thus, there is a need to develop new reparative therapies. In that regard, stimulating tissue regeneration by autologous stem/progenitor cells has emerged as a promising new strategy. We showed previously that migratory chondrogenic progenitor cells (CPCs) were recruited to injured cartilage, where they showed a capability in situ tissue repair. Here, we tested the hypothesis that the meniscus contains a similar population of regenerative cells. Explant studies revealed that migrating cells were mainly confined to the red zone in normal menisci: However, these cells were capable of repopulating defects made in the white zone. In vivo, migrating cell numbers increased dramatically in damaged meniscus. Relative to non-migrating meniscus cells, migrating cells were more clonogenic, overexpressed progenitor cell markers, and included a larger side population. Gene expression profiling showed that the migrating population was more similar to CPCs than other meniscus cells. Finally, migrating cells equaled CPCs in chondrogenic potential, indicating a capacity for repair of the cartilaginous white zone of the meniscus. These findings demonstrate that, much as in articular cartilage, injuries to the meniscus mobilize an intrinsic progenitor cell population with strong reparative potential. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1966-1972, 2017.
Collapse
Affiliation(s)
- Dongrim Seol
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
| | - Cheng Zhou
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA,Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Marc J. Brouillette
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
| | - Ino Song
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA,Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA
| | - Yin Yu
- Center for Engineering in Medicine, Massachusetts General Hospital, Boston, MA 02114, USA,Harvard Medical School, Boston, MA 02114, USA
| | - Hyeong Hun Choe
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
| | - Abigail D. Lehman
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
| | - Kee W. Jang
- Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD 20892, USA
| | - Douglas C. Fredericks
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA,Department of Bone Healing Research Laboratory and Iowa Spine Research Laboratory, University of Iowa, Iowa City, IA 52242, USA
| | - Barbara J. Laughlin
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA
| | - James A. Martin
- Department of Orthopaedics and Rehabilitation, University of Iowa, Iowa City, IA 52242, USA,Department of Biomedical Engineering, University of Iowa, Iowa City, IA 52242, USA,Correspondence to James A. Martin (Address: 1182 ML, University of Iowa, Iowa City, IA 52242; T: +1-319-335-5810; F: +1-319-335-5631; )
| |
Collapse
|
60
|
Wu Q, Huang JH. Ectopic expression of Smurf2 and acceleration of age-related intervertebral disc degeneration in a mouse model. J Neurosurg Spine 2017; 27:116-126. [DOI: 10.3171/2016.11.spine16901] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVELumbar intervertebral disc degeneration, an age-related process, is a major cause of low-back pain. Although low-back pain is a very common clinical problem in the aging population, no effective treatment is available, largely owing to lack of understanding of the molecular mechanisms underlying disc degeneration. The goal of this study was to characterize how ectopic expression of Smurf2 driven by the collagen Type II alpha 1 (Col2a1) promoter alters disc cell phenotype and associated cellular events, matrix synthesis, and gene expression during disc degeneration in mice.METHODSTo characterize how ectopic expression of Smurf2 in Col2a1-promoter working cells affects the disc degeneration process, the authors performed histological and immunohistochemical analysis of lumbar spine specimens harvested from wild-type (WT) and Col2a1-Smurf2 transgenic mice at various ages (n ≥ 6 in each age group). To elucidate the molecular mechanism underlying Smurf2-mediated disc degeneration, the authors isolated cells from WT and Col2a1-Smurf2 transgenic lumbar intervertebral discs and performed Western blot and real-time RT-PCR (reverse transcription polymerase chain reaction) to examine the protein and mRNA levels of interesting targets.RESULTSThe authors demonstrated that approximately 30% of WT mice at 10–12 months of age had started to show disc degeneration and that the disc degeneration process was accelerated by 3–6 months in Col2a1-Smurf2 transgenic mice. Chondrocyte-like cell proliferation, maturation, and fibrotic tissue formation in the inner annulus were often accompanied by fibroblast-to-chondrocyte differentiation in the outer annulus in transgenic discs. The chondrocyte-like cells in transgenic discs expressed higher levels of connective tissue growth factor (CTGF) than were expressed in WT counterparts.CONCLUSIONSThe findings that ectopic expression of Smurf2 driven by the Col2a1 promoter accelerated disc degeneration in Col2a1-Smurf2 transgenic mice, and that higher levels of CTGF protein and mRNA were present in Col2a1-Smurf2 transgenic discs, indicate that Smurf2 accelerates disc degeneration via upregulation of CTGF.
Collapse
|
61
|
Chen S, Fu P, Wu H, Pei M. Meniscus, articular cartilage and nucleus pulposus: a comparative review of cartilage-like tissues in anatomy, development and function. Cell Tissue Res 2017; 370:53-70. [PMID: 28413859 DOI: 10.1007/s00441-017-2613-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/17/2017] [Indexed: 01/07/2023]
Abstract
The degradation of cartilage in the human body is impacted by aging, disease, genetic predisposition and continued insults resulting from daily activity. The burden of cartilage defects (osteoarthritis, rheumatoid arthritis, intervertebral disc damage, knee replacement surgeries, etc.) is daunting in light of substantial economic and social stresses. This review strives to broaden the scope of regenerative medicine and tissue engineering approaches used for cartilage repair by comparing and contrasting the anatomical and functional nature of the meniscus, articular cartilage (AC) and nucleus pulposus (NP). Many review papers have provided detailed evaluations of these cartilages and cartilage-like tissues individually but none have comprehensively examined the parallels and inconsistencies in signaling, genetic expression and extracellular matrix composition between tissues. For the first time, this review outlines the importance of understanding these three tissues as unique entities, providing a comparative analysis of anatomy, ultrastructure, biochemistry and function for each tissue. This novel approach highlights the similarities and differences between tissues, progressing research toward an understanding of what defines each tissue as distinctive. The goal of this paper is to provide researchers with the fundamental knowledge to correctly engineer the meniscus, AC and NP without inadvertently developing the wrong tissue function or biochemistry.
Collapse
Affiliation(s)
- Song Chen
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, One Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Peiliang Fu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Haishan Wu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, One Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
| |
Collapse
|
62
|
Treatments of Meniscus Lesions of the Knee: Current Concepts and Future Perspectives. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2017. [DOI: 10.1007/s40883-017-0025-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
63
|
Rey-Rico A, Cucchiarini M, Madry H. Hydrogels for precision meniscus tissue engineering: a comprehensive review. Connect Tissue Res 2017; 58:317-328. [PMID: 28051883 DOI: 10.1080/03008207.2016.1276576] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The meniscus plays a pivotal role to preserve the knee joint homeostasis. Lesions to the meniscus are frequent, have a reduced ability to heal, and may induce tibiofemoral osteoarthritis. Current reconstructive therapeutic options mainly focus on the treatment of lesions in the peripheral vascularized region. In contrast, few approaches are capable of stimulating repair of damaged meniscal tissue in the central, avascular portion. Tissue engineering approaches are of high interest to repair or replace damaged meniscus tissue in this area. Hydrogel-based biomaterials are of special interest for meniscus repair as its inner part contains relatively high proportions of proteoglycans which are responsible for the viscoelastic compressive properties and hydration grade. Hydrogels exhibiting high water content and providing a specific three-dimensional (3D) microenvironment may be engineered to precisely resemble this topographical composition of the meniscal tissue. Different polymers of both natural and synthetic origins have been manipulated to produce hydrogels hosting relevant cell populations for meniscus regeneration and provide platforms for meniscus tissue replacement. So far, these compounds have been employed to design controlled delivery systems of bioactive molecules involved in meniscal reparative processes or to host genetically modified cells as a means to enhance meniscus repair. This review describes the most recent advances on the use of hydrogels as platforms for precision meniscus tissue engineering.
Collapse
Affiliation(s)
- Ana Rey-Rico
- a Center of Experimental Orthopaedics , Saarland University Medical Center , Homburg/Saar , Germany
| | - Magali Cucchiarini
- a Center of Experimental Orthopaedics , Saarland University Medical Center , Homburg/Saar , Germany
| | - Henning Madry
- a Center of Experimental Orthopaedics , Saarland University Medical Center , Homburg/Saar , Germany.,b Department of Orthopaedic Surgery , Saarland University Medical Center , Homburg/Saar , Germany
| |
Collapse
|
64
|
Furumatsu T, Ozaki T. An Analysis of Pathological Activities of CCN Proteins in Joint Disorders: Mechanical Stretch-Mediated CCN2 Expression in Cultured Meniscus Cells. Methods Mol Biol 2017; 1489:533-542. [PMID: 27734404 DOI: 10.1007/978-1-4939-6430-7_45] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The multifunctional growth factor CYR61/CTGF/NOV (CCN) 2, also known as connective tissue growth factor, regulates cellular proliferation, differentiation, and tissue regeneration. Recent literatures have described important roles of CCN2 in the meniscus metabolism. However, the mechanical stress-mediated transcriptional regulation of CCN2 in the meniscus remains unclear. The meniscus is a fibrocartilaginous tissue that controls complex biomechanics of the knee joint. Therefore, the injured unstable meniscus has a poor healing potential especially in the avascular inner region. In addition, dysfunction of the meniscus correlates with the progression of degenerative knee joint disorders and joint space narrowing. Here, we describe an experimental approach that investigates the distinct cellular behavior of inner and outer meniscus cells in response to mechanical stretch. Our experimental model can analyze the relationships between stretch-induced CCN2 expression and its functional role in the meniscus homeostasis.
Collapse
Affiliation(s)
- Takayuki Furumatsu
- Department of Orthopaedic Surgery, Okayama University Graduate School, 2-5-1 Shikatacho, Kitaku, Okayama, 700-8558, Japan.
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School, 2-5-1 Shikatacho, Kitaku, Okayama, 700-8558, Japan
| |
Collapse
|
65
|
Narayanan G, Vernekar VN, Kuyinu EL, Laurencin CT. Poly (lactic acid)-based biomaterials for orthopaedic regenerative engineering. Adv Drug Deliv Rev 2016; 107:247-276. [PMID: 27125191 PMCID: PMC5482531 DOI: 10.1016/j.addr.2016.04.015] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 03/09/2016] [Accepted: 04/17/2016] [Indexed: 02/07/2023]
Abstract
Regenerative engineering converges tissue engineering, advanced materials science, stem cell science, and developmental biology to regenerate complex tissues such as whole limbs. Regenerative engineering scaffolds provide mechanical support and nanoscale control over architecture, topography, and biochemical cues to influence cellular outcome. In this regard, poly (lactic acid) (PLA)-based biomaterials may be considered as a gold standard for many orthopaedic regenerative engineering applications because of their versatility in fabrication, biodegradability, and compatibility with biomolecules and cells. Here we discuss recent developments in PLA-based biomaterials with respect to processability and current applications in the clinical and research settings for bone, ligament, meniscus, and cartilage regeneration.
Collapse
Affiliation(s)
- Ganesh Narayanan
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Varadraj N Vernekar
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Emmanuel L Kuyinu
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Cato T Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA; Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA; School of Medicine, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Reconstructive Sciences, University of Connecticut Health Center, Farmington, CT 06030, USA; Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA; Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA.
| |
Collapse
|
66
|
Numpaisal PO, Rothrauff BB, Gottardi R, Chien CL, Tuan RS. Rapidly dissociated autologous meniscus tissue enhances meniscus healing: An in vitro study. Connect Tissue Res 2016; 58:355-365. [PMID: 27726454 DOI: 10.1080/03008207.2016.1245727] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Treatment of meniscus tears is a persistent challenge in orthopedics. Although cell therapies have shown promise in promoting fibrocartilage formation in in vitro and preclinical studies, clinical application has been limited by the paucity of autologous tissue and the need for ex vivo cell expansion. Rapid dissociation of the free edges of the anterior and posterior meniscus with subsequent implantation in a meniscus lesion may overcome these limitations. The purpose of this study was to explore the effect of rapidly dissociated meniscus tissue in enhancing neotissue formation in a radial meniscus tear, as simulated in an in vitro explant model. MATERIALS AND METHODS All experiments in this study, performed at minimum with biological triplicates, utilized meniscal tissues from hind limbs of young cows. The effect of varying collagenase concentration (0.1%, 0.2% and 0.5% w/v) and treatment duration (overnight and 30 minutes) on meniscus cell viability, organization of the extracellular matrix (ECM), and gene expression was assessed through a cell metabolism assay, microscopic examination, and quantitative real-time reverse transcription polymerase chain reaction analysis, respectively. Thereafter, an explant model of a radial meniscus tear was used to evaluate the effect of a fibrin gel seeded with one of the following: (1) fibrin alone, (2) isolated and passaged (P2) meniscus cells, (3) overnight digested tissue, and (4) rapidly dissociated tissue. The quality of in vitro healing was determined through histological analysis and derivation of an adhesion index. RESULTS Rapid dissociation in 0.2% collagenase yielded cells with higher levels of metabolism than either 0.1% or 0.5% collagenase. When seeded in a three-dimensional fibrin hydrogel, both overnight digested and rapidly dissociated cells expressed greater levels of collagens type I and II than P2 meniscal cells at 1 week. At 4 and 8 weeks, collagen type II expression remained elevated only in the rapid dissociation group. Histological examination revealed enhanced healing in all cell-seeded treatment groups over cell-free fibrin controls at weeks 1, 4, and 8, but there were no significant differences across the treatment groups. CONCLUSIONS Rapid dissociation of meniscus tissue may provide a single-step approach to augment regenerative healing of meniscus repairs.
Collapse
Affiliation(s)
- Piya-On Numpaisal
- a Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,b Department of Anatomy and Cell Biology , College of Medicine, National Taiwan University , Taipei , Taiwan.,c Institute of Medicine, Suranaree University of Technology , Nakhon Ratchasima , Thailand
| | - Benjamin B Rothrauff
- a Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,d McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh , PA , USA
| | - Riccardo Gottardi
- a Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,e Ri.MED Foundation , Palermo , Italy
| | - Chung-Liang Chien
- b Department of Anatomy and Cell Biology , College of Medicine, National Taiwan University , Taipei , Taiwan
| | - Rocky S Tuan
- a Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery , University of Pittsburgh School of Medicine , Pittsburgh , PA , USA.,d McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh , PA , USA
| |
Collapse
|
67
|
Abstract
The knee is a fascinating yet complex joint. Researchers and clinicians agree that the joint is an organ comprised of highly specialized intrinsic and extrinsic tissues contributing to both health and disease. Key to the function and movement of the knee are the menisci, exquisite fibrocartilage structures that are critical structures for maintaining biological and biomechanical integrity of the joint. The biological/physiological functions of the menisci must be understood at the tissue, cellular and even molecular levels in order to determine clinically relevant methods for assessing it and influencing it. By investigating normal and pathological functions at the basic science level, we can begin to translate data to patients. The objective of this article is to provide an overview of this translational pathway so that progression toward improved diagnostic, preventative, and therapeutic strategies can be effectively pursued. We have thoroughly examined the pathobiological, biomarker, and imaging aspects of meniscus research. This translational approach can be effective toward optimal diagnosis, prevention, and treatment for the millions of patients who suffer from meniscal disorders each year.
Collapse
Affiliation(s)
- James L Cook
- a University of Missouri Department of Orthopaedic Surgery and Thompson Laboratory for Regenerative Orthopaedics , Columbia , MO , USA
| | - Keiichi Kuroki
- a University of Missouri Department of Orthopaedic Surgery and Thompson Laboratory for Regenerative Orthopaedics , Columbia , MO , USA
| | - Aaron M Stoker
- a University of Missouri Department of Orthopaedic Surgery and Thompson Laboratory for Regenerative Orthopaedics , Columbia , MO , USA
| | - Farrah A Monibi
- a University of Missouri Department of Orthopaedic Surgery and Thompson Laboratory for Regenerative Orthopaedics , Columbia , MO , USA
| | - Brandon L Roller
- b Department of Radiology , Wake Forest Baptist Medical Center , Winston-Salem , NC , USA
| |
Collapse
|
68
|
|
69
|
Inoue H, Furumatsu T, Maehara A, Tanaka T, Ozaki T. Histological and biological comparisons between complete and incomplete discoid lateral meniscus. Connect Tissue Res 2016; 57:408-16. [PMID: 27267748 DOI: 10.1080/03008207.2016.1195827] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The discoid lateral meniscus (DLM) is an anatomically abnormal meniscus that covers a greater area of the tibial plateau than the normal meniscus. The DLM is classified into two types: complete (CDLM) and incomplete (ICDLM) types. In this study, we investigated the histological and cell biological characteristics of CDLM and ICDLM. The number of blood vessels, proteoglycan deposition, and collagen distribution were assessed using meniscal tissues. Collagen production was also investigated in CDLM and ICDLM cells. The intercondylar region of the CDLM had a higher number of blood vessels than the inner region of the ICDLM. Safranin O staining density and type II collagen deposition in ICDLM were higher than those in CDLM. Type II collagen-positive cells were higher in ICLDM than in CDLM. CDLM cells showed slender fibroblastic morphology, while ICDLM cells were triangular chondrocytic in shape. This study demonstrated that the intercondylar region of the CDLM showed similar properties to the outer region of the meniscus. The inner region of the ICDLM, on the other hand, differed from the intercondylar region of the CDLM. Our results suggest that the intercondylar region of the CDLM may have a high healing potential like the outer meniscus.
Collapse
Affiliation(s)
- Hiroto Inoue
- a Department of Orthopaedic Surgery , Okayama University Graduate School , Okayama , Japan
| | - Takayuki Furumatsu
- a Department of Orthopaedic Surgery , Okayama University Graduate School , Okayama , Japan
| | - Ami Maehara
- a Department of Orthopaedic Surgery , Okayama University Graduate School , Okayama , Japan
| | - Takaaki Tanaka
- a Department of Orthopaedic Surgery , Okayama University Graduate School , Okayama , Japan
| | - Toshifumi Ozaki
- a Department of Orthopaedic Surgery , Okayama University Graduate School , Okayama , Japan
| |
Collapse
|
70
|
Advances in combining gene therapy with cell and tissue engineering-based approaches to enhance healing of the meniscus. Osteoarthritis Cartilage 2016; 24:1330-9. [PMID: 27063441 PMCID: PMC5298218 DOI: 10.1016/j.joca.2016.03.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/17/2016] [Accepted: 03/25/2016] [Indexed: 02/02/2023]
Abstract
Meniscal lesions are common problems in orthopaedic surgery and sports medicine, and injury or loss of the meniscus accelerates the onset of knee osteoarthritis (OA). Despite a variety of therapeutic options in the clinics, there is a critical need for improved treatments to enhance meniscal repair. In this regard, combining gene-, cell-, and tissue engineering-based approaches is an attractive strategy to generate novel, effective therapies to treat meniscal lesions. In the present work, we provide an overview of the tools currently available to improve meniscal repair and discuss the progress and remaining challenges for potential future translation in patients.
Collapse
|
71
|
Dyrna F, Herbst E, Hoberman A, Imhoff AB, Schmitt A. Stem cell procedures in arthroscopic surgery. Eur J Med Res 2016; 21:29. [PMID: 27411303 PMCID: PMC4944463 DOI: 10.1186/s40001-016-0224-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 07/05/2016] [Indexed: 12/13/2022] Open
Abstract
The stem cell as the building block necessary for tissue reparation and homeostasis plays a major role in regenerative medicine. Their unique property of being pluripotent, able to control immune process and even secrete a whole army of anabolic mediators, draws interest. While new arthroscopic procedures and techniques involving stem cells have been established over the last decade with improved outcomes, failures and dissatisfaction still occur. Therefore, there is increasing interest in ways to improve the healing response. MSCs are particularly promising for this task given their regenerative potential. While methods of isolating those cells are no longer poses a challenge, the best way of application is not clear. Several experiments in the realm of basic science and animal models have recently been published, addressing this issue, yet the application in clinical practice has lagged. This review provides an overview addressing the current standing of MSCs in the field of arthroscopic surgery.
Collapse
Affiliation(s)
- Felix Dyrna
- Department of Sports Orthopedics Klinikum rechts der Isar, Technical University, Ismaninger Str. 22, 81675, Munich, Germany
| | - Elmar Herbst
- Department of Sports Orthopedics Klinikum rechts der Isar, Technical University, Ismaninger Str. 22, 81675, Munich, Germany
| | - Alexander Hoberman
- Department of Orthopaedic Surgery, University of Connecticut, Farmington, CT, USA
| | - Andreas B Imhoff
- Department of Sports Orthopedics Klinikum rechts der Isar, Technical University, Ismaninger Str. 22, 81675, Munich, Germany
| | - Andreas Schmitt
- Department of Sports Orthopedics Klinikum rechts der Isar, Technical University, Ismaninger Str. 22, 81675, Munich, Germany.
| |
Collapse
|
72
|
Rey-Rico A, Klich A, Cucchiarini M, Madry H. Biomedical-grade, high mannuronic acid content (BioMVM) alginate enhances the proteoglycan production of primary human meniscal fibrochondrocytes in a 3-D microenvironment. Sci Rep 2016; 6:28170. [PMID: 27302206 PMCID: PMC4908386 DOI: 10.1038/srep28170] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 05/31/2016] [Indexed: 02/06/2023] Open
Abstract
Alginates are important hydrogels for meniscus tissue engineering as they support the meniscal fibrochondrocyte phenotype and proteoglycan production, the extracellular matrix (ECM) component chiefly responsible for its viscoelastic properties. Here, we systematically evaluated four biomedical- and two nonbiomedical-grade alginates for their capacity to provide the best three-dimensional (3-D) microenvironment and to support proteoglycan synthesis of encapsulated human meniscal fibrochondrocytes in vitro. Biomedical-grade, high mannuronic acid alginate spheres (BioLVM, BioMVM) were the most uniform in size, indicating an effect of the purity of alginate on the shape of the spheres. Interestingly, the purity of alginates did not affect cell viability. Of note, only fibrochondrocytes encapsulated in BioMVM alginate produced and retained significant amounts of proteoglycans. Following transplantation in an explant culture model, the alginate spheres containing fibrochondrocytes remained in close proximity with the meniscal tissue adjacent to the defect. The results reveal a promising role of BioMVM alginate to enhance the proteoglycan production of primary human meniscal fibrochondrocytes in a 3-D hydrogel microenvironment. These findings have significant implications for cell-based translational studies aiming at restoring lost meniscal tissue in regions containing high amounts of proteoglycans.
Collapse
Affiliation(s)
- Ana Rey-Rico
- Center of Experimental Orthopaedics, Saarland University, D-66421 Homburg, Germany
| | - Angelique Klich
- Center of Experimental Orthopaedics, Saarland University, D-66421 Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University, D-66421 Homburg, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University, D-66421 Homburg, Germany.,Department of Orthopaedic Surgery, Saarland University Medical Center, Saarland University, D-66421 Homburg, Germany
| |
Collapse
|
73
|
Abstract
OBJECTIVE the menisci are easily injured and difficult to repair. The aim of this study was to analyze the current state of meniscal surgery aimed at preserving morphology and conserving the biomechanics of the knee to prevent joint degeneration. METHODOLOGY a search of the electronic medical literature database Medline was conducted, from http://www.ncbi.nlm.nih.gov/pubmed. The search was not limited by language. Candidate articles were identified by searching for those that included the keywords meniscus, surgery, suture, implant, allograft. The limits were included for clinical research and clinical trials. Basic research was not included. The studies selected were evaluated and classified in three different categories: basic science, reconstruction (suture and meniscectomy) and implants (scaffolds and allograft). RESULTS the consequences of meniscectomy performed at a young age can lead to a joint cartilage degeneration twenty years later. There are few surgical options for the repair of meniscal injuries in order both to preserve the meniscus and to ensure the long term survival of the knee joint, meniscectomy, repair, suturing the tear, or reconstruction, when a meniscal allograft or synthetic substitute is used to replace the meniscus, but the biomechanical properties of the native meniscus are not reproduced entirely by the scaffolds that exist today. CONCLUSION therapies that successfully repair or replace the meniscus are therefore likely to prevent or delay osteoarthritis progression.
Collapse
Affiliation(s)
| | - Francisco Forriol
- Department of Clínica Sciences, University San Pablo - CEU, Boadilla del Monte, Spain
| |
Collapse
|
74
|
Cell-Based Strategies for Meniscus Tissue Engineering. Stem Cells Int 2016; 2016:4717184. [PMID: 27274735 PMCID: PMC4871968 DOI: 10.1155/2016/4717184] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/06/2016] [Accepted: 02/11/2016] [Indexed: 12/14/2022] Open
Abstract
Meniscus injuries remain a significant challenge due to the poor healing potential of the inner avascular zone. Following a series of studies and clinical trials, tissue engineering is considered a promising prospect for meniscus repair and regeneration. As one of the key factors in tissue engineering, cells are believed to be highly beneficial in generating bionic meniscus structures to replace injured ones in patients. Therefore, cell-based strategies for meniscus tissue engineering play a fundamental role in meniscal regeneration. According to current studies, the main cell-based strategies for meniscus tissue engineering are single cell type strategies; cell coculture strategies also were applied to meniscus tissue engineering. Likewise, on the one side, the zonal recapitulation strategies based on mimicking meniscal differing cells and internal architectures have received wide attentions. On the other side, cell self-assembling strategies without any scaffolds may be a better way to build a bionic meniscus. In this review, we primarily discuss cell seeds for meniscus tissue engineering and their application strategies. We also discuss recent advances and achievements in meniscus repair experiments that further improve our understanding of meniscus tissue engineering.
Collapse
|
75
|
Shi YY, Chen LX, Xu Y, Hu XQ, Ao YF, Wang JQ. Acetabular Labral Reconstruction With Autologous Tendon Tissue in a Porcine Model: In Vivo Histological Assessment and Gene Expression Analysis of the Healing Tissue. Am J Sports Med 2016; 44:1031-9. [PMID: 26838935 DOI: 10.1177/0363546515623784] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Acetabular labral reconstruction with autologous tendons is the preferred method for treating a severely damaged labrum. However, the healing process of implants remains unknown. Similar to the human acetabular labrum, the porcine acetabular labrum is a fibrocartilage-like tissue. PURPOSE This study aimed to characterize the histological healing process and gene expression profile of implants in a porcine model of acetabular labral reconstruction. STUDY DESIGN Descriptive laboratory study. METHODS Eighteen pigs were included in this study. The pigs underwent unilateral acetabular labral reconstruction. A 1.0 cm-long defect was made at the site of the anterior (cranial) dorsal labrum, which was repaired using an autologous mesogluteus tendon. The pigs were sacrificed at 12 and 24 weeks postoperatively. The implants were subjected to histological assessment and gene expression analysis. The cell phenotype of the implants was visualized using paraffin-embedded sections. RESULTS Macroscopic observations revealed that at 12 weeks, 8 of 9 implants partially filled the labral defect; by contrast, at 24 weeks, 6 of 9 implants fully filled and 3 implants partially filled the labral defects. Oval- or round-shaped fibrochondrocytes were found in the implants at 12 and 24 weeks. The matrix staining results showed that proteoglycan and collagen types 1 and 2 were more evident in the implants at 24 weeks than at 12 weeks. Gene expression analysis results revealed that COL2A1 and COL3A1 were expressed by the implants to a higher extent at 24 weeks than at 12 weeks; COL2A1 and COL3A1 were also expressed to a higher extent in the implants than in the native tendon. CONCLUSION On the basis of the results of histological assessment and gene expression analysis, autologous tendon tissue for acetabular labral reconstruction can fully or partially fill labral defects and converts to fibrocartilage, which is rich in proteoglycan and collagen types 1 and 2, at 24 weeks in a porcine model. CLINICAL RELEVANCE Autologous tendon tissue can be considered as a viable option for acetabular labral reconstruction.
Collapse
Affiliation(s)
- Yuan-Yuan Shi
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Lin-Xin Chen
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Yan Xu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Xiao-Qing Hu
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Ying-Fang Ao
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| | - Jian-Quan Wang
- Institute of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, China
| |
Collapse
|
76
|
Kuyinu EL, Narayanan G, Nair LS, Laurencin CT. Animal models of osteoarthritis: classification, update, and measurement of outcomes. J Orthop Surg Res 2016; 11:19. [PMID: 26837951 PMCID: PMC4738796 DOI: 10.1186/s13018-016-0346-5] [Citation(s) in RCA: 347] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 01/11/2016] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is one of the most commonly occurring forms of arthritis in the world today. It is a debilitating chronic illness causing pain and immense discomfort to the affected individual. Significant research is currently ongoing to understand its pathophysiology and develop successful treatment regimens based on this knowledge. Animal models have played a key role in achieving this goal. Animal models currently used to study osteoarthritis can be classified based on the etiology under investigation, primary osteoarthritis, and post-traumatic osteoarthritis, to better clarify the relationship between these models and the pathogenesis of the disease. Non-invasive animal models have shown significant promise in understanding early osteoarthritic changes. Imaging modalities play a pivotal role in understanding the pathogenesis of OA and the correlation with pain. These imaging studies would also allow in vivo surveillance of the disease as a function of time in the animal model. This review summarizes the current understanding of the disease pathogenesis, invasive and non-invasive animal models, imaging modalities, and pain assessment techniques in the animals.
Collapse
Affiliation(s)
- Emmanuel L Kuyinu
- Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, USA. .,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA. .,Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA.
| | - Ganesh Narayanan
- Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, USA. .,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA. .,Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA.
| | - Lakshmi S Nair
- Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, USA. .,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA. .,Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA. .,Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA. .,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, USA. .,Institute of Materials Science, University of Connecticut, Storrs, CT, USA.
| | - Cato T Laurencin
- Institute for Regenerative Engineering, University of Connecticut Health, Farmington, CT, USA. .,Raymond and Beverly Sackler Center for Biomedical, Biological, Physical and Engineering Sciences, University of Connecticut Health, Farmington, CT, USA. .,Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA. .,Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA. .,Department of Materials Science and Engineering, University of Connecticut, Storrs, CT, USA. .,Institute of Materials Science, University of Connecticut, Storrs, CT, USA. .,Department of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health, Farmington, CT, USA. .,Department of Chemical and Biomolecular Engineering, University of Connecticut, Storrs, CT, USA.
| |
Collapse
|
77
|
Huang H, Wang S, Gui J, Shen H. A study to identify and characterize the stem/progenitor cell in rabbit meniscus. Cytotechnology 2016; 68:2083-103. [PMID: 26820973 DOI: 10.1007/s10616-016-9949-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 01/18/2016] [Indexed: 12/27/2022] Open
Abstract
The repair of meniscus in the avascular zone remains a great challenge, largely owing to their limited healing capacity. Stem cells based tissue engineering provides a promising treatment option for damaged meniscus because of their multiple differentiation potential. We hypothesized that meniscus-derived stromal cells (MMSCs) may be present in meniscal tissue, and if their pluripotency and character can be established, they may play a role in meniscal healing. To test our hypothesis, we isolated MMSCs, bone marrow-derived stromal cells (BMSCs) and fibrochondrocytes from rabbits. In order to avoid bone marrow mesenchymal stromal cell contamination, the parameniscal tissues and vascular zone of meniscus were removed. The characters of these three types of cells were identified by evaluating morphology, colony formation, proliferation, immunocytochemistry and multi-differentiation. Moreover, a wound in the center of rabbit meniscus was created and used to analyze the effect of BMSCs and MMSCs on wounded meniscus healing. BMSCs & MMSCs expressed the stem cell markers SSEA-4, Nanog, nucleostemin and STRO-1, while fibrochondrocytes expressed none of these markers. Morphologically, MMSCs displayed smaller cell bodies and larger nuclei than ordinary fibrochondrocytes. Moreover, it was certified that MMSCs and BMSCs were all able to differentiate into adipocytes, osteocytes, and chondrocytes in vitro. However, more cartilage formation was found in wounded meniscus filled with MMSCs than that filled with BMSCs. We showed that rabbit menisci harbor the unique cell population MMSCs that has universal stem cell characteristics and posses a tendency to differentiate into chondrocytes. Future research should investigate the mechanobiology of MMSCs and explore the possibility of using MMSCs to more effectively repair or regenerate injured meniscus.
Collapse
Affiliation(s)
- He Huang
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Shukui Wang
- Central Laboratory, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Jianchao Gui
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, China
| | - Haiqi Shen
- Department of Orthopaedic Surgery, Nanjing First Hospital, Nanjing Medical University, 68 Changle Road, Nanjing, 210006, Jiangsu, China.
| |
Collapse
|
78
|
Lee HR, Shon OJ, Park SI, Kim HJ, Kim S, Ahn MW, Do SH. Platelet-Rich Plasma Increases the Levels of Catabolic Molecules and Cellular Dedifferentiation in the Meniscus of a Rabbit Model. Int J Mol Sci 2016; 17:ijms17010120. [PMID: 26784189 PMCID: PMC4730361 DOI: 10.3390/ijms17010120] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 11/24/2015] [Accepted: 01/11/2016] [Indexed: 01/10/2023] Open
Abstract
Despite the susceptibility to frequent intrinsic and extrinsic injuries, especially in the inner zone, the meniscus does not heal spontaneously owing to its poor vascularity. In this study, the effect of platelet-rich plasma (PRP), containing various growth factors, on meniscal mechanisms was examined under normal and post-traumatic inflammatory conditions. Isolated primary meniscal cells of New Zealand white (NZW) rabbits were incubated for 3, 10, 14 and 21 days with PRP(−), 10% PRP (PRP(+)), IL(+) or IL(+)PRP(+). The meniscal cells were collected and examined using reverse-transcription polymerase chain reaction (RT-PCR). Culture media were examined by immunoblot analyses for matrix metalloproteinases (MMP) catabolic molecules. PRP containing growth factors improved the cellular viability of meniscal cells in a concentration-dependent manner at Days 1, 4 and 7. However, based on RT-PCR, meniscal cells demonstrated dedifferentiation, along with an increase in type I collagen in the PRP(+) and in IL(+)PRP(+). In PRP(+), the aggrecan expression levels were lower than in the PRP(−) until Day 21. The protein levels of MMP-1 and MMP-3 were higher in each PRP group, i.e., PRP(+) and IL(+)PRP(+), at each culture time. A reproducible 2-mm circular defect on the meniscus of NZW rabbit was used to implant fibrin glue (control) or PRP in vivo. After eight weeks, the lesions in the control and PRP groups were occupied with fibrous tissue, but not with meniscal cells. This study shows that PRP treatment of the meniscus results in an increase of catabolic molecules, especially those related to IL-1α-induced inflammation, and that PRP treatment for an in vivo meniscus injury accelerates fibrosis, instead of meniscal cartilage.
Collapse
Affiliation(s)
- Hye-Rim Lee
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Korea.
| | - Oog-Jin Shon
- Department of Orthopedic Surgery, College of Medicine, Yeungnam University, Daegu 705-717, Korea.
| | - Se-Il Park
- Cardiovascular Product Evaluation Center, College of Medicine, Yonsei University, Seoul 120-752, Korea.
| | - Han-Jun Kim
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Korea.
| | - Sukyoung Kim
- School of Materials Science and Engineering, Yeungnam University, Gyeongsan 712-749, Korea.
| | - Myun-Whan Ahn
- Department of Orthopedic Surgery, College of Medicine, Yeungnam University, Daegu 705-717, Korea.
| | - Sun Hee Do
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine, Konkuk University, Seoul 143-701, Korea.
| |
Collapse
|
79
|
A combination of biomolecules enhances expression of E-cadherin and peroxisome proliferator-activated receptor gene leading to increased cell proliferation in primary human meniscal cells: an in vitro study. Cytotechnology 2015; 68:1747-61. [PMID: 26511364 DOI: 10.1007/s10616-015-9926-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/15/2015] [Indexed: 12/29/2022] Open
Abstract
The present study investigates the impact of biomolecules (biotin, glucose, chondroitin sulphate, proline) as supplement, (individual and in combination) on primary human meniscus cell proliferation. Primary human meniscus cells isolated from patients undergoing meniscectomy were maintained in Dulbecco's Modified Eagle's Medium (DMEM). The isolated cells were treated with above mentioned biomolecules as individual (0-100 µg/ml) and in combinations, as a supplement to DMEM. Based on the individual biomolecule study, a unique combination of biomolecules (UCM) was finalized using one way ANOVA analysis. With the addition of UCM as supplement to DMEM, meniscal cells reached 100 % confluency within 4 days in 60 mm culture plate; whereas the cells in medium devoid of UCM, required 36 days for reaching confluency. The impact of UCM on cell viability, doubling time, histology, gene expression, biomarkers expression, extra cellular matrix synthesis, meniscus cell proliferation with respect to passages and donor's age were investigated. The gene expression studies for E-cadherin and peroxisome proliferator-activated receptor (PPAR∆) using RT-qPCR and immunohistochemical analysis for Ki67, CD34 and Vimentin confirmed that UCM has significant impact on cell proliferation. The extracellular collagen and glycosaminoglycan secretion in cells supplemented with UCM were found to increase by 31 and 37 fold respectively, when compared to control on the 4th day. The cell doubling time was reduced significantly when supplemented with UCM. The addition of UCM showed positive influence on different passages and age groups. Hence, this optimized UCM can be used as an effective supplement for meniscal tissue engineering.
Collapse
|
80
|
Molinos M, Almeida CR, Gonçalves RM, Barbosa MA. Improvement of Bovine Nucleus Pulposus Cells Isolation Leads to Identification of Three Phenotypically Distinct Cell Subpopulations. Tissue Eng Part A 2015; 21:2216-27. [DOI: 10.1089/ten.tea.2014.0461] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Maria Molinos
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Catarina R. Almeida
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Raquel M. Gonçalves
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Mário A. Barbosa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
81
|
Cucchiarini M, Schmidt K, Frisch J, Kohn D, Madry H. Overexpression of TGF-β via rAAV-Mediated Gene Transfer Promotes the Healing of Human Meniscal Lesions Ex Vivo on Explanted Menisci. Am J Sports Med 2015; 43:1197-205. [PMID: 25646364 DOI: 10.1177/0363546514567063] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Direct application of therapeutic gene vectors derived from the adeno-associated virus (AAV) might be beneficial to improve the healing of meniscal tears. PURPOSE To test the ability of recombinant AAV (rAAV) to overexpress the potent transforming growth factor-β (TGF-β) in primary cultures of human meniscal fibrochondrocytes, in human meniscal explants, and in experimental human meniscal lesions as a new tool to enhance meniscal repair. STUDY DESIGN Controlled laboratory study. METHODS The effects of the candidate treatment on the proliferative and metabolic activities of meniscal cells were monitored in vitro for up to 21 days and in situ in intact and injured human menisci for up to 15 days using biochemical, immunohistochemical, histological, and histomorphometric analyses. RESULTS Efficient production of TGF-β via rAAV was achieved in vitro and in situ, both in the intact and injured meniscus. Application of the rAAV TGF-β vector stimulated the levels of cell proliferation and matrix synthesis (type I collagen) compared with control gene transfer in all systems tested, especially in situ in regions of poor healing capacity and in sites of meniscal injury. No adverse effects of the candidate treatment were observed at the level of osseous differentiation, as tested by immunodetection of type X collagen. Most remarkably, a significant reduction of the amplitude of meniscal tears was noted after TGF-β treatment, an effect that was associated with increased expression levels of the α-smooth muscle actin contractile marker. CONCLUSION Overexpression of TGF-β via rAAV gene transfer is capable of modulating the reparative activities of human meniscal cells, allowing for the healing of meniscal lesions by convenient injection in sites of injury. CLINICAL RELEVANCE Direct gene-based approaches using rAAV have strong potential to develop new therapeutic options that aim at treating human meniscal defects.
Collapse
Affiliation(s)
- Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Katharina Schmidt
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Janina Frisch
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany
| | - Dieter Kohn
- Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| | - Henning Madry
- Center of Experimental Orthopaedics, Saarland University Medical Center, Homburg/Saar, Germany Department of Orthopaedic Surgery, Saarland University Medical Center, Homburg/Saar, Germany
| |
Collapse
|
82
|
Moran CJ, Busilacchi A, Lee CA, Athanasiou KA, Verdonk PC. Biological augmentation and tissue engineering approaches in meniscus surgery. Arthroscopy 2015; 31:944-55. [PMID: 25687715 DOI: 10.1016/j.arthro.2014.11.044] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 11/19/2014] [Accepted: 11/19/2014] [Indexed: 02/02/2023]
Abstract
PURPOSE The purpose of this review was to evaluate the role of biological augmentation and tissue engineering strategies in meniscus surgery. Although clinical (human), preclinical (animal), and in vitro tissue engineering studies are included here, we have placed additional focus on addressing preclinical and clinical studies reported during the 5-year period used in this review in a systematic fashion while also providing a summary review of some important in vitro tissue engineering findings in the field over the past decade. METHODS A search was performed on PubMed for original works published from 2009 to March 31, 2014 using the term "meniscus" with all the following terms: "scaffolds," "constructs," "cells," "growth factors," "implant," "tissue engineering," and "regenerative medicine." Inclusion criteria were the following: English-language articles and original clinical, preclinical (in vivo), and in vitro studies of tissue engineering and regenerative medicine application in knee meniscus lesions published from 2009 to March 31, 2014. RESULTS Three clinical studies and 18 preclinical studies were identified along with 68 tissue engineering in vitro studies. These reports show the increasing promise of biological augmentation and tissue engineering strategies in meniscus surgery. The role of stem cell and growth factor therapy appears to be particularly useful. A review of in vitro tissue engineering studies found a large number of scaffold types to be of promise for meniscus replacement. Limitations include a relatively low number of clinical or preclinical in vivo studies, in addition to the fact there is as yet no report in the literature of a tissue-engineered meniscus construct used clinically. Neither does the literature provide clarity on the optimal meniscus scaffold type or biological augmentation with which meniscus repair or replacement would be best addressed in the future. There is increasing focus on the role of mechanobiology and biomechanical and biochemical cues in this process, however, and it is hoped that this may lead to improvements in this strategy. CONCLUSIONS There appears to be significant potential for biological augmentation and tissue engineering strategies in meniscus surgery to enhance options for repair and replacement. However, there are still relatively few clinical studies being reported in this regard. There is a strong need for improved translational activities and infrastructure to link the large amounts of in vitro and preclinical biological and tissue engineering data to clinical application. LEVEL OF EVIDENCE Level IV, systematic review of Level I-IV studies.
Collapse
Affiliation(s)
- Cathal J Moran
- Sports Surgery Clinic and Trinity College Dublin, Dublin, Ireland.
| | - Alberto Busilacchi
- Clinical Orthopaedics, Università Politecnica delle Marche, Ancona, Italy
| | - Cassandra A Lee
- Departments of Orthopaedic Surgery & Biomedical Engineering, University of California, Davis, California, U.S.A
| | - Kyriacos A Athanasiou
- Departments of Orthopaedic Surgery & Biomedical Engineering, University of California, Davis, California, U.S.A
| | | |
Collapse
|
83
|
Fuhrmann IK, Steinhagen J, Rüther W, Schumacher U. Comparative immunohistochemical evaluation of the zonal distribution of extracellular matrix and inflammation markers in human meniscus in osteoarthritis and rheumatoid arthritis. Acta Histochem 2015; 117:243-54. [PMID: 25827912 DOI: 10.1016/j.acthis.2014.12.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 12/18/2014] [Accepted: 12/22/2014] [Indexed: 12/19/2022]
Abstract
UNLABELLED The purpose of our study was to analyze the distribution of the major extracellular matrix glycosaminoglycan hyaluronan (HA), its receptor CD44 and cells which influence (re)modeling of the extracellular matrix (T- and B-cells, macrophages, endothelial cells) in menisci obtained from patients suffering from rheumatoid arthritis or osteoarthritis in order to analyze whether these markers could be useful to differentiate between both arthropathies. Human menisci were sampled from patients undergoing total knee arthroplasty. Histological staining (H&E, PAS/Alcian Blue for neutral and charged carbohydrate residues) and (immuno)histochemistry were performed for detection of HA, CD44, sphingosine-1-phosphate receptor 1 (EDG-1) as a marker for endothelial cells, CD3 as a marker for T-cells, CD20 as a marker for B-cells and CD68 as a marker for macrophages. The extracellular matrix in the vascularized zone showed higher amounts of HA as well as acid carbohydrate residues in comparison to the poorly vascularized zones of the meniscus in both disease entities. EDG-1 positive endothelial cells were present in all zones, with fewer cells being detected in the inner zones of the rheumatoid menisci than in the osteoarthritic ones. Macrophages, T- and B-cells as well as CD44-positive cells were more prominent in the vascularized zone of the meniscus than in the poorly vascularized central zone. The distribution patterns of the extracellular matrix components as well as the CD44-positive cells and the inflammation markers in the peripheral zone resembled the distribution in synovial tissue, indicating that both synovia and meniscus were involved in pathological changes in osteoarthritis and rheumatoid arthritis. IN CONCLUSION the distribution of extracellular glycoconjugates and of cells modulating their synthesis showed similar results in both arthropathies, not enabling a differentiation between rheumatoid arthritis and osteoarthritis but underlining the role of these markers in inflammation and degradation in human meniscus.
Collapse
|
84
|
Abstract
The meniscus plays a critical biomechanical role in the knee, providing load support, joint stability, and congruity. Importantly, growing evidence indicates that the mechanobiologic response of meniscal cells plays a critical role in the physiologic, pathologic, and repair responses of the meniscus. Here we review experimental and theoretical studies that have begun to directly measure the biomechanical effects of joint loading on the meniscus under physiologic and pathologic conditions, showing that the menisci are exposed to high contact stresses, resulting in a complex and nonuniform stress-strain environment within the tissue. By combining microscale measurements of the mechanical properties of meniscal cells and their pericellular and extracellular matrix regions, theoretical and experimental models indicate that the cells in the meniscus are exposed to a complex and inhomogeneous environment of stress, strain, fluid pressure, fluid flow, and a variety of physicochemical factors. Studies across a range of culture systems from isolated cells to tissues have revealed that the biological response of meniscal cells is directly influenced by physical factors, such as tension, compression, and hydrostatic pressure. In addition, these studies have provided new insights into the mechanotransduction mechanisms by which physical signals are converted into metabolic or pro/anti-inflammatory responses. Taken together, these in vivo and in vitro studies show that mechanical factors play an important role in the health, degeneration, and regeneration of the meniscus. A more thorough understanding of the mechanobiologic responses of the meniscus will hopefully lead to therapeutic approaches to prevent degeneration and enhance repair of the meniscus.
Collapse
|
85
|
Roumazeille T, Klouche S, Rousselin B, Bongiorno V, Graveleau N, Billot N, Hardy P. Arthroscopic meniscal allograft transplantation with two tibia tunnels without bone plugs: evaluation of healing on MR arthrography and functional outcomes. Knee Surg Sports Traumatol Arthrosc 2015; 23:264-9. [PMID: 23508524 DOI: 10.1007/s00167-013-2476-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 03/11/2013] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Meniscal allograft transplantation seems to be a valid therapeutic option to restore the knee function and limit the development of osteoarthritis after menisectomy. No surgical technique has been shown to provide better results than others. The main objective of this study was to assess graft healing after arthroscopic meniscal allograft transplantation without bone plugs. METHODS This retrospective study included all patients who underwent arthroscopic meniscal allograft transplantation during 2005-2010. The meniscal horns were fixed through two tibia tunnels without bone plugs. The primary endpoint was graft healing according to Henning's criteria on MR arthrography (MRA) at 6-month follow-up. The secondary endpoints were the KOOS questionnaire, the IKDC score, measurement of the joint space and meniscal extrusion on both MRA at 6-month and MRI at last follow-up. The series included 22 patients, mean age 37 ± 7.5 years. The allograft was lateral in 20 cases and medial in 2 cases. The mean follow-up was 4.4 ± 1.6 years with one lost to follow-up. RESULTS MR arthrography was performed in 14/21 patients at 6-months of follow-up: 8/14 (57.1%) had total graft healing, 2/14 (14.3%) partial healing and 4/14 (28.6%) no healing. At final follow-up, all functional scores had significantly improved. The average pre- and post-operative joint space thickness was similar. MRI showed meniscal extrusion in 75% of patients. CONCLUSION The meniscal allograft transplantation without bone plugs effectively treats painful and functional sequellae of meniscectomies. The graft healed in most patients at 6-month follow-up. The long-term clinical relevance of meniscal extrusion has to be evaluated. LEVEL OF EVIDENCE Retrospective study, Level IV.
Collapse
Affiliation(s)
- Thibaut Roumazeille
- Hôpitaux Universitaires Paris Ile-de-France Ouest, AP-HP, 92100, Boulogne-Billancourt, France
| | | | | | | | | | | | | |
Collapse
|
86
|
Zhang X, Aoyama T, Ito A, Tajino J, Nagai M, Yamaguchi S, Iijima H, Kuroki H. Regional comparisons of porcine menisci. J Orthop Res 2014; 32:1602-11. [PMID: 25196310 DOI: 10.1002/jor.22687] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 06/10/2014] [Indexed: 02/04/2023]
Abstract
The purpose of this study was to analyze histologic, biochemical, and biomechanical differences between zonal, regional, and anatomic locations of porcine menisci. We evaluated six menisci removed from pigs. Medial and lateral menisci were divided into three regions: anterior, middle, and posterior. In each portion, the central zone (CZ) and peripheral zone (PZ) were examined histologically (hematoxylin & eosin, safranin O/Fast green, and picrosiriusred staining), using scanning electron microscopy, biochemically (hydroxyproline assay for collagen content and dimethylmethylene blue assay for glycosaminoglycan [GAG] content), and biomechanically (compression testing). Collagen content in the CZ was lower than that in the PZ. GAG content in the CZ was higher than that in the PZ. GAG content in the PZ of the posterior portion was significantly higher than that in the anterior and middle portions. Compression strength in the CZ was higher than that in the PZ. The differences in cellular phenotype, vascular penetration, and ECM not only between CZ and PZ but also among the anterior, middle, and posterior portions were clarified in the immature porcine meniscus. This result helps further our understanding of the biological characteristic of the meniscus. © 2014 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 32:1602-1611, 2014.
Collapse
Affiliation(s)
- Xiangkai Zhang
- Department of Motor Function Analysis, Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | | | |
Collapse
|
87
|
Actifit synthetic meniscal substitute: experience with 18 patients in Brest, France. Orthop Traumatol Surg Res 2014; 100:S385-9. [PMID: 25454332 DOI: 10.1016/j.otsr.2014.09.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 09/17/2014] [Indexed: 02/02/2023]
Abstract
BACKGROUND The management of post-meniscectomy pain is poorly standardised. Allogeneic transplantation may be appropriate in some patients after total meniscectomy. After partial meniscectomy, the synthetic meniscal substitute Actifit may constitute a valid option if the knee is stable or stabilised and aligned or re-aligned. The interconnected pore structure of Actifit promotes tissue regeneration from the meniscal wall. Arthroscopy is used to position the implant, which is then sutured to the remaining native meniscus using horizontal stitches and to the meniscal wall using vertical stitches. However, a burdensome programme of rigorous rehabilitation is required after Actifit implantation. HYPOTHESIS We hypothesised that implantation of a meniscal substitute effectively alleviated pain without adversely affecting the knee. OBJECTIVES To assess the intra-articular behaviour of Actifit and the outcomes of Actifit implantation in a prospective case-series of patients monitored using arthroscopy, pathology, and imaging studies, as well as the Lysholm score to assess clinical benefits on daily activities. MATERIALS AND METHODS Between October 2009 and April 2012, 18 patients underwent Actifit implantation at the military hospital in Brest, France. All procedures were performed by the same surgeon, who had extensive experience with meniscal suturing. There were 13 males and 5 females aged 20 to 46 years. The medial meniscus was involved in 13 patients and the lateral meniscus in 5 patients. Actifit implantation was used alone in 6 patients and in combination with anterior cruciate ligament reconstruction and/or realignment osteotomy in 12 patients. All patients were followed-up for at least 2 years. RESULTS The mean Lysholm score after 1 year was 92%, indicating excellent outcomes. Magnetic resonance imaging showed no damage to the implant or degeneration of the neighbouring cartilage. Histological examination of meniscal substitute biopsies taken 1 year after implantation showed polymer ingrowth by normal chondrocytes and fibrochondrocytes. The clinical and radiographic outcomes compared favourably with those seen after isolated procedures on bone or ligaments. DISCUSSION Actifit has no deleterious effects on patients. The implant induces and promotes meniscal regeneration. Actifit constitutes a major addition to our therapeutic armamentarium. We provide convincing evidence that meniscal reconstruction can be highly beneficial in decreasing the risk of progression to knee osteoarthritis. LEVEL OF EVIDENCE IV.
Collapse
|
88
|
Petzold J, Casadonte R, Otto M, Kriegsmann M, Granrath M, Baltzer A, Vogel J, Drees P, Deininger S, Becker M, Kriegsmann J. MALDI-Massenspektrometrie am Meniskus. Z Rheumatol 2014; 74:438-46. [DOI: 10.1007/s00393-014-1468-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
89
|
Di Giancamillo A, Deponti D, Addis A, Domeneghini C, Peretti GM. Meniscus maturation in the swine model: changes occurring along with anterior to posterior and medial to lateral aspect during growth. J Cell Mol Med 2014; 18:1964-74. [PMID: 25216283 PMCID: PMC4244012 DOI: 10.1111/jcmm.12367] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 06/10/2014] [Indexed: 12/27/2022] Open
Abstract
The meniscus plays important roles in knee function and mechanics and is characterized by a heterogeneous matrix composition. The changes in meniscus vascularization observed during growth suggest that the tissue-specific composition may be the result of a maturation process. This study has the aim to characterize the structural and biochemical variations that occur in the swine meniscus with age. To this purpose, menisci were collected from young and adult pigs and divided into different zones. In study 1, both lateral and medial menisci were divided into the anterior horn, the body and the posterior horn for the evaluation of glycosaminoglycans (GAGs), collagen 1 and 2 content. In study 2, the menisci were sectioned into the inner, the intermediate and the outer zones to determine the variations in the cell phenotype along with the inner–outer direction, through gene expression analysis. According to the results, the swine meniscus is characterized by an increasing enrichment in the cartilaginous component with age, with an increasing deposition in the anterior horn (GAGs and collagen 2; P < 0.01 both); moreover, this cartilaginous matrix strongly increases in the inner avascular and intermediate zone, as a consequence of a specific differentiation of meniscal cells towards a cartilaginous phenotype (collagen 2, P < 0.01). The obtained data add new information on the changes that accompany meniscus maturation, suggesting a specific response of meniscal cells to the regional mechanical stimuli in the knee joint.
Collapse
|
90
|
Pereira H, Caridade SG, Frias AM, Silva-Correia J, Pereira DR, Cengiz IF, Mano JF, Oliveira JM, Espregueira-Mendes J, Reis RL. Biomechanical and cellular segmental characterization of human meniscus: building the basis for Tissue Engineering therapies. Osteoarthritis Cartilage 2014; 22:1271-81. [PMID: 25038489 DOI: 10.1016/j.joca.2014.07.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 06/29/2014] [Accepted: 07/05/2014] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To overcome current limitations of Tissue Engineering (TE) strategies, deeper comprehension on meniscus biology is required. This study aims to combine biomechanical segmental analysis of fresh human meniscus tissues and its correlation with architectural and cellular characterization. METHOD Morphologically intact menisci, from 44 live donors were studied after division into three radial segments. Dynamic mechanical analysis (DMA) was performed at physiological-like conditions. Micro-computed tomography (CT) analysis of freeze-dried samples assessed micro-structure. Flow cytometry, histology and histomorphometry were used for cellular study and quantification. RESULTS Anterior segments present significantly higher damping properties. Mid body fresh medial meniscus presents higher values of E' compared to lateral. Cyclic loads influence the viscoelastic behavior of menisci. By increasing the frequency leads to an increase in stiffness. Conversely, with increasing frequencies, the capacity to dissipate energy and damping properties initially decrease and then rise again. Age and gender directly correlate with higher E' and tan δ. Micro-CT analysis revealed that mean porosity was 55.5 (21.2-89.8)% and 64.7 (47.7-81.8)% for freeze-dried lateral and medial meniscus, respectively. Predominant cells are positive for CD44, CD73, CD90 and CD105, and lack CD31, CD34 and CD45 (present in smaller populations). Histomorphometry revealed that cellularity decreases from vascular zone 1 to zone 3. Anterior segments of lateral and medial meniscus have inferior cellularity as compared to mid body and posterior ones. CONCLUSION Menisci are not uniform structures. Anterior segments have lower cellularity and higher damping. Cyclic loads influence viscoelastic characteristics. Future TE therapies should consider segmental architecture, cellularity and biomechanics of fresh tissue.
Collapse
Affiliation(s)
- H Pereira
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio de Barco, 4806-909, Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal; Clínica Espregueira-Mendes F.C. Porto Stadium - FIFA Medical Centre of Excellence, Portugal; Orthopedic Department Centro Hospitalar Póvoa de Varzim - Vila do Conde, Portugal.
| | - S G Caridade
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio de Barco, 4806-909, Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - A M Frias
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio de Barco, 4806-909, Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - J Silva-Correia
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio de Barco, 4806-909, Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - D R Pereira
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio de Barco, 4806-909, Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - I F Cengiz
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio de Barco, 4806-909, Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - J F Mano
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio de Barco, 4806-909, Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| | - J M Oliveira
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio de Barco, 4806-909, Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal.
| | - J Espregueira-Mendes
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio de Barco, 4806-909, Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal; Clínica Espregueira-Mendes F.C. Porto Stadium - FIFA Medical Centre of Excellence, Portugal
| | - R L Reis
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Univ. Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, S. Cláudio de Barco, 4806-909, Taipas, Guimarães, Portugal; ICVS/3B's - PT Government Associated Laboratory, Portugal
| |
Collapse
|
91
|
Fox AJS, Wanivenhaus F, Burge AJ, Warren RF, Rodeo SA. The human meniscus: a review of anatomy, function, injury, and advances in treatment. Clin Anat 2014; 28:269-87. [PMID: 25125315 DOI: 10.1002/ca.22456] [Citation(s) in RCA: 255] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/23/2014] [Indexed: 12/22/2022]
Abstract
Meniscal injuries are recognized as a cause of significant musculoskeletal morbidity. The menisci are vital for the normal function and long-term health of the knee joint. The purpose of this review is to provide current knowledge regarding the anatomy and biomechanical functions of the menisci, incidence, injury patterns and the advancements in treatment options of meniscal injury. A literature search was performed by a review of PubMed, Google Scholar, MEDLINE, and OVID for all relevant articles published between 1897 and 2014. This study highlights the anatomical and biomechanical characteristics of the menisci, which may be relevant to injury patterns and treatment options. An understanding of the normal anatomy and biomechanical functions of the knee menisci is a necessary prerequisite to understanding pathologies associated with the knee.
Collapse
Affiliation(s)
- Alice J S Fox
- Laboratory for Soft Tissue Research, Hospital for Special Surgery, 535 East 70th Street, New York, New York
| | | | | | | | | |
Collapse
|
92
|
Cell distribution and regenerative activity following meniscus replacement. INTERNATIONAL ORTHOPAEDICS 2014; 38:1937-44. [DOI: 10.1007/s00264-014-2426-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 06/12/2014] [Indexed: 01/06/2023]
|
93
|
Nonviral gene transfer into human meniscal cells. Part II: effect of three-dimensional environment and overexpression of human fibroblast growth factor 2. INTERNATIONAL ORTHOPAEDICS 2014; 38:1931-6. [PMID: 24957484 DOI: 10.1007/s00264-014-2405-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 05/31/2014] [Indexed: 10/25/2022]
Abstract
PURPOSE Our aim was to study the effect of three-dimensional (3D) environment and overexpression of human fibroblast growth factor 2 (FGF-2) on meniscal fibrochondrocytes in vitro. METHODS Human meniscal fibrochondrocytes were transfected with expression plasmid vectors carrying the Photinus pyralis luciferase gene, the Escherichia coli β-galactosidase gene or a human FGF-2 cDNA. Modified fibrochondrocytes were cultivated in 3D alginate hydrogel or cell pellets or in 2D monolayer culture. RESULTS The levels of luciferase activity showed a peak at day two and returned to baseline levels by day 11, regardless of the type of cultivation. Both 3D environments supported the secretion of human FGF-2 protein upon FGF-2 transfection. Overexpression of human FGF-2 by genetically modified human meniscal fibrochondrocytes stimulated proliferation but not glycosaminoglycan synthesis only in 3D culture. Culture in alginate spheres resulted in a larger difference in cell numbers compared with pellet cultures. CONCLUSIONS Three-dimensional alginate spheres are well suited for the culture of genetically modified human meniscal fibrochondrocytes. These data are of value for cell-based approaches to meniscal repair using genetically modified human meniscal fibrochondrocytes overexpressing human FGF-2.
Collapse
|
94
|
Halili AN, Hasirci N, Hasirci V. A multilayer tissue engineered meniscus substitute. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2014; 25:1195-1209. [PMID: 24452271 DOI: 10.1007/s10856-014-5145-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 01/10/2014] [Indexed: 06/03/2023]
Abstract
Various methods have been tried to treat the main meniscus problem, meniscal tears, for which we believe tissue engineering could be a viable solution. In this study, a three dimensional, collagen-based meniscus substitute was prepared by tissue engineering using human fibrochondrocytes and a collagen based-scaffold. This construct was made with 3 different collagen-based foams interspaced with two electrospun nano/microfibrous mats. The top layer was made of collagen type I-chondroitin sulfate-hyaluronic acid (Coll-CS-HA), and the middle and the bottom layers were made of only collagen type I with different porosities and thus with different mechanical properties. The mats of aligned fibers were a blend of collagen type I and poly(L-lactic acid-co-glycolic acid) (PLGA). After seeding with human fibrochondrocytes, cell attachment, proliferation, and production of extracellular matrix and glucoseaminoglycan were studied. Cell seeding had a positive effect on the compressive properties of foams and the 3D construct. The 3D construct with all its 5 layers had better mechanical properties than the individual foams.
Collapse
|
95
|
Osawa A, Harner CD, Gharaibeh B, Matsumoto T, Mifune Y, Kopf S, Ingham SJM, Schreiber V, Usas A, Huard J. The use of blood vessel-derived stem cells for meniscal regeneration and repair. Med Sci Sports Exerc 2014; 45:813-23. [PMID: 23247715 DOI: 10.1249/mss.0b013e31827d1e06] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Surgical repairs of tears in the vascular region of the meniscus usually heal better than repairs performed in the avascular region; thus, we hypothesized that this region might possess a richer supply of vascular-derived stem cells than the avascular region. METHODS In this study, we analyzed 6 menisci extracted from aborted human fetuses and 12 human lateral menisci extracted from adult human subjects undergoing total knee arthroplasty. Menisci were immunostained for CD34 (a stem cell marker) and CD146 (a pericyte marker) in situ, whereas other menisci were dissected into two regions (peripheral and inner) and used to isolate meniscus-derived cells by flow cytometry. Cell populations expressing CD34 and CD146 were tested for their multilineage differentiation potentials, including chondrogenic, osteogenic, and adipogenic lineages. Fetal peripheral meniscus cells were transplanted by intracapsular injection into the knee joints of an athymic rat meniscal tear model. Rat menisci were extracted and histologically evaluated after 4 wk posttransplantation. RESULTS Immunohistochemistry and flow cytometric analyses demonstrated that a higher number of CD34- and CD146-positive cells were found in the peripheral region compared with the inner region. The CD34- and CD146-positive cells isolated from the vascular region of both fetal and adult menisci demonstrated multilineage differentiation capacities and were more potent than cells isolated from the inner (avascular) region. Fetal CD34- and CD146-positive cells transplanted into the athymic rat knee joint were recruited into the meniscal tear sites and contributed to meniscus repair. CONCLUSIONS The vascularized region of the meniscus contains more stem cells than the avascular region. These meniscal-derived stem cells were multipotent and contributed to meniscal regeneration.
Collapse
Affiliation(s)
- Aki Osawa
- Stem Cell Research Center, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Pro-inflammatory stimulation of meniscus cells increases production of matrix metalloproteinases and additional catabolic factors involved in osteoarthritis pathogenesis. Osteoarthritis Cartilage 2014; 22:264-74. [PMID: 24315792 PMCID: PMC3967850 DOI: 10.1016/j.joca.2013.11.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 10/22/2013] [Accepted: 11/09/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Meniscus injury increases the risk of osteoarthritis; however, the biologic mechanism remains unknown. We hypothesized that pro-inflammatory stimulation of meniscus would increase production of matrix-degrading enzymes, cytokines and chemokines which cause joint tissue destruction and could contribute to osteoarthritis development. DESIGN Meniscus and cartilage tissue from healthy tissue donors and total knee arthroplasties (TKAs) was cultured. Primary cell cultures were stimulated with pro-inflammatory factors [IL-1β, IL-6, or fibronectin fragments (FnF)] and cellular responses were analyzed by real-time PCR, protein arrays and immunoblots. To determine if NF-κB was required for MMP production, meniscus cultures were treated with inflammatory factors with and without the NF-κB inhibitor, hypoestoxide. RESULTS Normal and osteoarthritic meniscus cells increased their MMP secretion in response to stimulation, but specific patterns emerged that were unique to each stimulus with the greatest number of MMPs expressed in response to FnF. Meniscus collagen and connective tissue growth factor (CTGF) gene expression was reduced. Expression of cytokines (IL-1α, IL-1β, IL-6), chemokines (IL-8, CXCL1, CXCL2, CSF1) and components of the NF-κB and tumor necrosis factor (TNF) family were significantly increased. Cytokine and chemokine protein production was also increased by stimulation. When primary cell cultures were treated with hypoestoxide in conjunction with pro-inflammatory stimulation, p65 activation was reduced as were MMP-1 and MMP-3 production. CONCLUSIONS Pro-inflammatory stimulation of meniscus cells increased matrix metalloproteinase production and catabolic gene expression. The meniscus could have an active biologic role in osteoarthritis development following joint injury through increased production of cytokines, chemokines, and matrix-degrading enzymes.
Collapse
|
97
|
Abstract
Lesions in the meniscus occur particularly in young, active patients in the nonvascularized area which, consequently have a bad intrinsic healing capacity. This has a large impact on the mobility and function of the knee joint. Lesions, and partial lesions, lead to the progression of osteoarthritis over time in a large proportion of patients. The only clinical treatment for severe cases so far is an allograft donor meniscus, which is used mostly in patients with severe osteoarthritis with a completely destroyed meniscus. However, this technique still has to be considered as experimental and, thus, is not yet used on a routine basis. Various technical solutions have been advocated to repair meniscus lesions. One solution is to perform a partial meniscectomy and insert a collagen meniscus implant (CMI) at the site of the lesion. However, the initial mechanical properties of the collagen scaffold are inferior to the native meniscus. Therefore, it is only possible to perform a CMI implantation if the peripheral rim of the meniscus is still intact. Histology of preclinical and clinical biopsies of the implanted CMI demonstrated a repopulation of the scaffold by fibrous tissue and in time a remodeling of the fibrous tissue into fibrocartilaginous-like tissue. Based on histology, the ingrowth of new tissue into the CMI might occur by a process of synovial overgrowth, but other mechanisms of revitalization are also possible. Although some clinical studies demonstrated improvement in outcome scores, the number of patients was small in all studies and the positive effect on the prevention of progression of osteoarthritis was not compared with control groups.
Collapse
Affiliation(s)
- Pieter Buma
- Orthopedic Research Laboratory, Radboud University, Nijmegen Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | | | | |
Collapse
|
98
|
Longo UG, Loppini M, Romeo G, Maffulli N, Denaro V. Histological scoring systems for tissue-engineered, ex vivo and degenerative meniscus. Knee Surg Sports Traumatol Arthrosc 2013; 21:1569-76. [PMID: 22829330 DOI: 10.1007/s00167-012-2142-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 07/09/2012] [Indexed: 02/05/2023]
Abstract
PURPOSE Because its function is strictly related to the quality of meniscal tissue, one of the most important outcome measures for the evaluation of meniscal repair effectiveness is the assessment of histological features. Data on the validation and application of the histological scoring systems in research settings and specific fields of meniscal disorders are lacking. The available histological scoring systems to assess meniscal tissue were systematically evaluated. METHODS Histological scoring systems for the analysis of degenerative meniscal changes, ex vivo and tissue-engineered meniscal repair were reviewed. Furthermore, the validity and applicability of the scoring systems were assessed. RESULTS The Copenhaver classification and Mankin score have been modified to classify the degeneration of collagen bundles in the meniscal structure. The Pauli score seems to be a comprehensive and simple scoring system for the evaluation of both macroscopic and histologic meniscal changes related to ageing and osteoarthritic degeneration. The Zhang score may be used for ex vivo gene therapy in meniscus healing. The Ishida score seems to be the most adequate for the evaluation of tissue-engineered meniscal repair. CONCLUSION Although several histological scoring systems are available to assess meniscal structure, only few of them have been validated for specific application in research settings. Validated scores are required to provide a standardized data collection to allow the comparison of results of different research groups. Further experimental and clinical studies are needed to find a comprehensive and validated histological scoring system in the field of meniscus repair. LEVEL OF EVIDENCE Systematic review of Level III studies, Level III.
Collapse
Affiliation(s)
- Umile Giuseppe Longo
- Department of Orthopaedic and Trauma Surgery, Campus Bio-Medico University, Via Alvaro del Portillo, 200, 00128 Trigoria, Rome, Italy
| | | | | | | | | |
Collapse
|
99
|
Deponti D, Di Giancamillo A, Scotti C, Peretti GM, Martin I. Animal models for meniscus repair and regeneration. J Tissue Eng Regen Med 2013; 9:512-27. [PMID: 23712959 DOI: 10.1002/term.1760] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 02/24/2013] [Accepted: 04/02/2013] [Indexed: 12/21/2022]
Abstract
The meniscus plays an important role in knee function and mechanics. Meniscal lesions, however, are common phenomena and this tissue is not able to achieve spontaneous successful repair, particularly in the inner avascular zone. Several animal models have been studied and proposed for testing different reparative approaches, as well as for studying regenerative methods aiming to restore the original shape and function of this structure. This review summarizes the gross anatomy, function, ultrastructure and biochemical composition of the knee meniscus in several animal models in comparison with the human meniscus. The relevance of the models is discussed from the point of view of basic research as well as of clinical translation for meniscal repair, substitution and regeneration. Finally, the advantages and disadvantages of each model for various research directions are critically discussed.
Collapse
|
100
|
Freymann U, Endres M, Goldmann U, Sittinger M, Kaps C. Toward scaffold-based meniscus repair: effect of human serum, hyaluronic acid and TGF-ß3 on cell recruitment and re-differentiation. Osteoarthritis Cartilage 2013; 21:773-81. [PMID: 23473977 DOI: 10.1016/j.joca.2013.02.655] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 01/21/2013] [Accepted: 02/25/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Repair approaches for the non-vascular meniscus are rarely developed. Recent strategies use scaffold-based techniques and inducing factors. The aim of the study was the investigation of cell recruitment and re-differentiation inducing factors for a scaffold-based meniscus repair approach. METHOD 3D cultivation of in vitro expanded human meniscus-derived cells was performed in high-density cultures supplemented with 25% hyaluronic acid (HA), 10% human serum (HS) or 10 ng/ml transforming growth factor (TGF-ß3) compared to untreated controls. The in vitro cell recruitment potential of different HS concentrations was tested by chemotaxis assay. Analysis of chondrocytic markers (type I, II, IX collagen and proteoglycans) was performed on protein and gene expression level. RESULTS Cells were attracted by 1-20% HS. 3D cultures supplemented with 10% HS and 25% HA showed meniscus-like gene expression profiles at day 7 with significantly increased cartilage oligomeric matrix protein (COMP) and aggrecan expression levels in the HS group and a slightly increased profile in the HA group compared to control. The TGF-ß3 group showed an additional induction of gene expression levels for type II and type IX collagen. Histological findings confirmed these results by proteoglycan and type I collagen staining in all groups and type II collagen staining only in the TGF-ß3 group. CONCLUSION This study demonstrates that human meniscus cells are attracted by HS and allow for meniscal matrix formation in 3D culture in the presence of HA and HS, whereas TGF-ß3 additive does not initiate meniscal tissue. Regarding non-vascular meniscus repair, results of this study encourage scaffold-based repair approaches.
Collapse
Affiliation(s)
- U Freymann
- TransTissue Technologies GmbH, Charitéplatz 1/Virchowweg 11, 10117 Berlin, Germany.
| | | | | | | | | |
Collapse
|