1
|
Lin J, Huang J, Jiao Z, Nian M, Li C, Dai Y, Jia S, Zhang X. Mesenchymal stem cells for osteoarthritis: Recent advances in related cell therapy. Bioeng Transl Med 2025; 10:e10701. [PMID: 39801757 PMCID: PMC11711223 DOI: 10.1002/btm2.10701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/20/2024] [Accepted: 07/04/2024] [Indexed: 01/06/2025] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease that affects the entire joint and has been a huge burden on the health care system worldwide. Although traditional therapy and targeted cartilage cell therapy have made significant progress in the treatment of OA and cartilage regeneration, there are still many problems. Mesenchymal stem cells from various tissues are the most studied cell type and have been used in preclinical and clinical studies of OA, because they are more widely available, have a greater capacity for in vitro expansion, and have anti-inflammatory and immunomodulatory properties compared to autologous chondrocytes. This article will systematically review the latest developments in these areas. It may provide new insights for improving OA and cartilage regeneration.
Collapse
Affiliation(s)
- Jianjing Lin
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
| | - Jingtao Huang
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Zilu Jiao
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
| | - Mengyuan Nian
- Cardre Health Care DepartmentPeking University Shenzhen HospitalShenzhenChina
| | - Canfeng Li
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
| | - Yali Dai
- Cardre Health Care DepartmentPeking University Shenzhen HospitalShenzhenChina
| | - Shicheng Jia
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
- Shantou University Medical CollegeShantouChina
| | - Xintao Zhang
- Department of Sports Medicine and RehabilitationPeking University Shenzhen HospitalShenzhenChina
| |
Collapse
|
2
|
Josephson TO, Morgan EF. Harnessing mechanical cues in the cellular microenvironment for bone regeneration. Front Physiol 2023; 14:1232698. [PMID: 37877097 PMCID: PMC10591087 DOI: 10.3389/fphys.2023.1232698] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
At the macroscale, bones experience a variety of compressive and tensile loads, and these loads cause deformations of the cortical and trabecular microstructure. These deformations produce a variety of stimuli in the cellular microenvironment that can influence the differentiation of marrow stromal cells (MSCs) and the activity of cells of the MSC lineage, including osteoblasts, osteocytes, and chondrocytes. Mechanotransduction, or conversion of mechanical stimuli to biochemical and biological signals, is thus part of a multiscale mechanobiological process that drives bone modeling, remodeling, fracture healing, and implant osseointegration. Despite strong evidence of the influence of a variety of mechanical cues, and multiple paradigms proposed to explain the influence of these cues on tissue growth and differentiation, even a working understanding of how skeletal cells respond to the complex combinations of stimuli in their microenvironments remains elusive. This review covers the current understanding of what types of microenvironmental mechanical cues MSCs respond to and what is known about how they respond in the presence of multiple such cues. We argue that in order to realize the vast potential for harnessing the cellular microenvironment for the enhancement of bone regeneration, additional investigations of how combinations of mechanical cues influence bone regeneration are needed.
Collapse
Affiliation(s)
- Timothy O. Josephson
- Biomedical Engineering, Boston University, Boston, MA, United States
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States
| | - Elise F. Morgan
- Biomedical Engineering, Boston University, Boston, MA, United States
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States
- Mechanical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
3
|
Jeyachandran D, Murshed M, Haglund L, Cerruti M. A Bioglass-Poly(lactic-co-glycolic Acid) Scaffold@Fibrin Hydrogel Construct to Support Endochondral Bone Formation. Adv Healthc Mater 2023; 12:e2300211. [PMID: 37462089 PMCID: PMC11468889 DOI: 10.1002/adhm.202300211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/21/2023] [Accepted: 07/05/2023] [Indexed: 07/29/2023]
Abstract
Bone tissue engineering using stem cells to build bone directly on a scaffold matrix often fails due to lack of oxygen at the injury site. This may be avoided by following the endochondral ossification route; herein, a cartilage template is promoted first, which can survive hypoxic environments, followed by its hypertrophy and ossification. However, hypertrophy is so far only achieved using biological factors. This work introduces a Bioglass-Poly(lactic-co-glycolic acid@fibrin (Bg-PLGA@fibrin) construct where a fibrin hydrogel infiltrates and encapsulates a porous Bg-PLGA. The hypothesis is that mesenchymal stem cells (MSCs) loaded in the fibrin gel and induced into chondrogenesis degrade the gel and become hypertrophic upon reaching the stiffer, bioactive Bg-PLGA core, without external induction factors. Results show that Bg-PLGA@fibrin induces hypertrophy, as well as matrix mineralization and osteogenesis; it also promotes a change in morphology of the MSCs at the gel/scaffold interface, possibly a sign of osteoblast-like differentiation of hypertrophic chondrocytes. Thus, the Bg-PLGA@fibrin construct can sequentially support the different phases of endochondral ossification purely based on material cues. This may facilitate clinical translation by decreasing in-vitro cell culture time pre-implantation and the complexity associated with the use of external induction factors.
Collapse
Affiliation(s)
| | - Monzur Murshed
- Faculty of DentistryDepartment of Medicineand Shriners Hospital for ChildrenMcGill UniversityMontrealQuebecH4A 0A9Canada
| | - Lisbet Haglund
- Experimental SurgeryMcGill UniversityMontrealH3G 2M1Canada
| | - Marta Cerruti
- Department of Mining and Materials EngineeringMcGill UniversityMontrealH3A 0C1Canada
| |
Collapse
|
4
|
Lu C, Zheng J, Yoshitomi T, Kawazoe N, Yang Y, Chen G. How Hydrogel Stiffness Affects Adipogenic Differentiation of Mesenchymal Stem Cells under Controlled Morphology. ACS APPLIED BIO MATERIALS 2023; 6:3441-3450. [PMID: 37061939 DOI: 10.1021/acsabm.3c00159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2023]
Abstract
Matrix stiffness has been disclosed as an essential regulator of cell fate. However, it is barely studied how the matrix stiffness affects stem cell functions when cell morphology changes. Thus, in this study, the effect of hydrogel stiffness on adipogenic differentiation of human bone-marrow-derived mesenchymal stem cells (hMSCs) with controlled morphology was investigated. Micropatterns of different size and elongation were prepared by a photolithographical micropatterning technique. The hMSCs were cultured on the micropatterns and showed a different spreading area and elongation following the geometry of the underlying micropatterns. The cells with controlled morphology were embedded in agarose hydrogels of different stiffnesses. The cells showed a different level of adipogenic differentiation that was dependent on both hydrogel stiffness and cell morphology. Adipogenic differentiation became strong when the cell spreading area decreased and hydrogel stiffness increased. Adipogenic differentiation did not change with cell elongation. Therefore, cell spreading area and hydrogel stiffness could synergistically affect adipogenic differentiation of hMSCs, while cell elongation did not affect adipogenic differentiation. A change of cell morphology and hydrogel stiffness was accompanied by actin filament alignment that was strongly related to adipogenic differentiation. The results indicated that cell morphology could affect cellular sensitivity to hydrogel stiffness. The results will provide useful information for the elucidation of the interaction of stem cells and their microenvironmental biomechanical cues.
Collapse
Affiliation(s)
- Chengyu Lu
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Jing Zheng
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Toru Yoshitomi
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Naoki Kawazoe
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Yingnan Yang
- Graduate School of Life and Environment Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8572, Japan
| | - Guoping Chen
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
- Department of Materials Science and Engineering, Graduate School of Pure and Applied Science, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
5
|
Petitjean N, Canadas P, Jorgensen C, Royer P, Le Floc'h S, Noël D. Complex deformation of cartilage micropellets following mechanical stimulation promotes chondrocyte gene expression. Stem Cell Res Ther 2023; 14:226. [PMID: 37649121 PMCID: PMC10469822 DOI: 10.1186/s13287-023-03459-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND Articular cartilage (AC)'s main function is to resist to a stressful mechanical environment, and chondrocytes are responding to mechanical stress for the development and homeostasis of this tissue. However, current knowledge on processes involved in response to mechanical stimulation is still limited. These mechanisms are commonly investigated in engineered cartilage models where the chondrocytes are included in an exogeneous biomaterial different from their natural extracellular matrix. The aim of the present study is to better understand the impact of mechanical stimulation on mesenchymal stromal cells (MSCs)-derived chondrocytes generated in their own extracellular matrix. METHODS A fluidic custom-made device was used for the mechanical stimulation of cartilage micropellets obtained from human MSCs by culture in a chondrogenic medium for 21 days. Six micropellets were positioned into the conical wells of the device chamber and stimulated with different signals of positive pressure (amplitude, frequency and duration). A camera was used to record the sinking of each micropellet into their cone, and micropellet deformation was analyzed using a finite element model. Micropellets were harvested at different time points after stimulation for RT-qPCR and histology analysis. RESULTS Moderate micropellet deformation was observed during stimulation with square pressure signals as mean von Mises strains between 6.39 and 14.35% were estimated for amplitudes of 1.75-14 kPa superimposed on a base pressure of 50% of the amplitude. The compression, tension and shear observed during deformation did not alter micropellet microstructure as shown by histological staining. A rapid and transient increase in the expression of chondrocyte markers (SOX9, AGG and COL2B) was measured after a single 30-min stimulation with a square pressure signal of 3.5 kPa amplitude superimposed on a minimum pressure of 1.75 kPa, at 1 Hz. A small change of 1% of cyclical deformations when using a square pressure signal instead of a constant pressure signal induced a fold change of 2 to 3 of chondrogenic gene expression. Moreover, the expression of fibrocartilage (COL I) or hypertrophic cartilage (COL X, MMP13 and ADAMTS5) was not significantly regulated, except for COL X. CONCLUSIONS Our data demonstrate that the dynamic deformation of cartilage micropellets by fluidic-based compression modulates the expression of chondrocyte genes responsible for the production of a cartilage-like extracellular matrix. This lays the foundations for further investigating the chondrocyte mechanobiology and the cartilage growth under mechanical stimulation.
Collapse
Affiliation(s)
- Noémie Petitjean
- IRMB, University of Montpellier, INSERM, Montpellier, France
- LMGC, CNRS, University of Montpellier, Montpellier, France
| | | | - Christian Jorgensen
- IRMB, University of Montpellier, INSERM, Montpellier, France
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France
| | - Pascale Royer
- LMGC, CNRS, University of Montpellier, Montpellier, France
| | | | - Danièle Noël
- IRMB, University of Montpellier, INSERM, Montpellier, France.
- Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, Montpellier, France.
- Inserm U1183, IRMB, Hôpital Saint-Eloi, 80 Avenue Augustin Fliche, 34295, Montpellier Cedex 5, France.
| |
Collapse
|
6
|
Lazaro-Pacheco D, Mohseni M, Rudd S, Cooper-White J, Holsgrove TP. The role of biomechanical factors in models of intervertebral disc degeneration across multiple length scales. APL Bioeng 2023; 7:021501. [PMID: 37180733 PMCID: PMC10168717 DOI: 10.1063/5.0137698] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/24/2023] [Indexed: 05/16/2023] Open
Abstract
Low back pain is the leading cause of disability, producing a substantial socio-economic burden on healthcare systems worldwide. Intervertebral disc (IVD) degeneration is a primary cause of lower back pain, and while regenerative therapies aimed at full functional recovery of the disc have been developed in recent years, no commercially available, approved devices or therapies for the regeneration of the IVD currently exist. In the development of these new approaches, numerous models for mechanical stimulation and preclinical assessment, including in vitro cell studies using microfluidics, ex vivo organ studies coupled with bioreactors and mechanical testing rigs, and in vivo testing in a variety of large and small animals, have emerged. These approaches have provided different capabilities, certainly improving the preclinical evaluation of these regenerative therapies, but challenges within the research environment, and compromises relating to non-representative mechanical stimulation and unrealistic test conditions, remain to be resolved. In this review, insights into the ideal characteristics of a disc model for the testing of IVD regenerative approaches are first assessed. Key learnings from in vivo, ex vivo, and in vitro IVD models under mechanical loading stimulation to date are presented alongside the merits and limitations of each model based on the physiological resemblance to the human IVD environment (biological and mechanical) as well as the possible feedback and output measurements for each approach. When moving from simplified in vitro models to ex vivo and in vivo approaches, the complexity increases resulting in less controllable models but providing a better representation of the physiological environment. Although cost, time, and ethical constraints are dependent on each approach, they escalate with the model complexity. These constraints are discussed and weighted as part of the characteristics of each model.
Collapse
Affiliation(s)
- Daniela Lazaro-Pacheco
- Department of Engineering, University of Exeter, Harrison Building, Streatham Campus, North Park Road, Exeter EX4 4QF, United Kingdom
| | - Mina Mohseni
- School of Chemical Engineering, The University of Queensland, St. Lucia QLD 4072, Australia
| | - Samuel Rudd
- School of Chemical Engineering, The University of Queensland, St. Lucia QLD 4072, Australia
| | | | - Timothy Patrick Holsgrove
- Department of Engineering, University of Exeter, Harrison Building, Streatham Campus, North Park Road, Exeter EX4 4QF, United Kingdom
| |
Collapse
|
7
|
Li T, Ma Z, Zhang Y, Yang Z, Li W, Lu D, Liu Y, Qiang L, Wang T, Ren Y, Wang W, He H, Zhou X, Mao Y, Zhu J, Wang J, Chen X, Dai K. Regeneration of Humeral Head Using a 3D Bioprinted Anisotropic Scaffold with Dual Modulation of Endochondral Ossification. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205059. [PMID: 36755334 PMCID: PMC10131811 DOI: 10.1002/advs.202205059] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 11/06/2022] [Indexed: 06/18/2023]
Abstract
Tissue engineering is theoretically thought to be a promising method for the reconstruction of biological joints, and thus, offers a potential treatment alternative for advanced osteoarthritis. However, to date, no significant progress is made in the regeneration of large biological joints. In the current study, a biomimetic scaffold for rabbit humeral head regeneration consisting of heterogeneous porous architecture, various bioinks, and different hard supporting materials in the cartilage and bone regions is designed and fabricated in one step using 3D bioprinting technology. Furthermore, orchestrated dynamic mechanical stimulus combined with different biochemical cues (parathyroid hormone [PTH] and chemical component hydroxyapatite [HA] in the outer and inner region, respectively) are used for dual regulation of endochondral ossification. Specifically, dynamic mechanical stimulus combined with growth factor PTH in the outer region inhibits endochondral ossification and results in cartilage regeneration, whereas dynamic mechanical stimulus combined with HA in the inner region promotes endochondral ossification and results in efficient subchondral bone regeneration. The strategy established in this study with the dual modulation of endochondral ossification for 3D bioprinted anisotropic scaffolds represents a versatile and scalable approach for repairing large joints.
Collapse
Affiliation(s)
- Tao Li
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
- Department of OrthopaedicsXinhua Hospital affiliated to Shanghai Jiaotong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092P. R. China
| | - Zhengjiang Ma
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Yuxin Zhang
- Department of Oral SurgeryShanghai Ninth People's HospitalShanghai Jiao Tong University School of MedicineCollege of StomatologyShanghai Jiao Tong UniversityNational Center for StomatologyNational Clinical Research Center for Oral DiseasesShanghai Key Laboratory of StomatologyShanghai200011China
| | - Zezheng Yang
- Department of OrthopedicsThe Fifth People's Hospital of ShanghaiFudan UniversityMinhang DistrictShanghai200240P. R. China
| | - Wentao Li
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Dezhi Lu
- School of MedicineShanghai UniversityJing An DistrictShanghai200444China
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Lei Qiang
- Southwest JiaoTong University College of MedicineNo. 111 North 1st Section of Second Ring RoadChengdu610036China
| | - Tianchang Wang
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Ya Ren
- Southwest JiaoTong University College of MedicineNo. 111 North 1st Section of Second Ring RoadChengdu610036China
| | - Wenhao Wang
- Southwest JiaoTong University College of MedicineNo. 111 North 1st Section of Second Ring RoadChengdu610036China
| | - Hongtao He
- The Third Ward of Department of OrthopedicsThe Second Hospital of Dalian Medical UniversityNo. 467, Zhongshan Road, Shahekou DistrictDalianLiaoning Province116000P. R. China
| | - Xiaojun Zhou
- College of Biological Science and Medical EngineeringState Key Laboratory for Modification of Chemical Fibers and Polymer MaterialsDonghua UniversityShanghai201620P. R. China
| | - Yuanqing Mao
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Junfeng Zhu
- Department of OrthopaedicsXinhua Hospital affiliated to Shanghai Jiaotong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092P. R. China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| | - Xiaodong Chen
- Department of OrthopaedicsXinhua Hospital affiliated to Shanghai Jiaotong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092P. R. China
| | - Kerong Dai
- Shanghai Key Laboratory of Orthopaedic ImplantDepartment of Orthopaedic SurgeryShanghai Ninth People's Hospital Affiliated Shanghai Jiao Tong University School of Medicine639 Zhizaoju RdShanghai200011China
| |
Collapse
|
8
|
Kinugasa K, Shimomura K, Tachibana Y, Hiramatsu K, Horibe S, Shino K, Tanaka Y. Posterior Ankle Impingement Caused by Hyaline-Like Cartilage Generation in Ballet Dancers-A Report of 2 Cases. J Foot Ankle Surg 2022; 61:e9-e14. [PMID: 34801380 DOI: 10.1053/j.jfas.2021.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 04/03/2020] [Accepted: 10/11/2021] [Indexed: 02/03/2023]
Abstract
Posterior ankle impingement syndrome is mainly seen in ballet dancers and frequently associated with specific movements in ballet such as pointe and demi pointe in which the whole-body weight is applied to the maximally plantar flexed ankle. We performed arthroscopic debridement for 2 dedicated ballet dancers on the intervening soft tissue causing posterior ankle impingement syndrome (PAIS). In both cases, T2-weighted magnetic resonance imaging (MRI) revealed low-signal intensity of meniscus-like soft tissue without abnormal osseous findings, connecting from the posterior side of the talus to Kager's fat pad. To examine the intervening soft tissue in detail, we performed histological evaluation by hematoxylin and eosin staining, Safranin O fast green staining, and immunohistochemistry for type I collagen and type II collagen. Hematoxylin and eosin staining showed that there was cartilage-like tissue including chondrocyte-like cells in contact with fibrous tissue. The extracellular matrix in the cartilage zone was consistently stained by Safranin O staining and type II collagen without any staining with type I collagen. These findings suggested that the meniscus-like soft tissue appearing as low-signal intensity on MRI at the posterior side of talus included hyaline-like cartilage. To the extent of our knowledge, these were rare cases of hyaline-like cartilage generation causing PAIS in ballet dancers, which might be associated with ballet specific movements resulting in chondrogenesis.
Collapse
Affiliation(s)
- Kazutaka Kinugasa
- Department of Orthopaedic Sports Medicine, Osaka Rosai Hospital, Sakai, Osaka, Japan.
| | - Kazunori Shimomura
- Department of Orthopaedic Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yuta Tachibana
- Department of Orthopaedic Sports Medicine, Osaka Rosai Hospital, Sakai, Osaka, Japan
| | - Kunihiko Hiramatsu
- Department of Orthopaedic Surgery, Tamai Orthopaedic Hospital, Hannan, Osaka, Japan
| | - Shuji Horibe
- Faculty of Comprehensive Rehabilitation, Osaka Prefecture University, Habikino, Osaka, Japan
| | - Konsei Shino
- Department of Sports Orthopaedics, Yukioka Hospital, Kita-ku, Osaka, Osaka, Japan
| | - Yoshinari Tanaka
- Department of Orthopaedic Sports Medicine, Osaka Rosai Hospital, Sakai, Osaka, Japan
| |
Collapse
|
9
|
Volz M, Wyse-Sookoo KR, Travascio F, Huang CY, Best TM. MECHANOBIOLOGICAL APPROACHES FOR STIMULATING CHONDROGENESIS OF STEM CELLS. Stem Cells Dev 2022; 31:460-487. [PMID: 35615879 DOI: 10.1089/scd.2022.0049] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Chondrogenesis is the process of differentiation of stem cells into mature chondrocytes. Such a process consists of chemical, functional, and structural changes which are initiated and mediated by the host environment of the cells. To date, the mechanobiology of chondrogenesis has not been fully elucidated. Hence, experimental activity is focused on recreating specific environmental conditions for stimulating chondrogenesis, and to look for a mechanistic interpretation of the mechanobiological response of cells in the cartilaginous tissues. There are a large number of studies on the topic that vary considerably in their experimental protocols used for providing environmental cues to cells for differentiation, making generalizable conclusions difficult to ascertain. The main objective of this contribution is to review the mechanobiological stimulation of stem cell chondrogenesis and methodological approaches utilized to date to promote chondrogenesis of stem cells in-vitro. In-vivo models will also be explored, but this area is currently limited. An overview of the experimental approaches used by different research groups may help the development of unified testing methods that could be used to overcome existing knowledge gaps, leading to an accelerated translation of experimental findings to clinical practice.
Collapse
Affiliation(s)
- Mallory Volz
- University of Miami, 5452, Biomedical Engineering, Coral Gables, Florida, United States;
| | | | - Francesco Travascio
- University of Miami, 5452, Mechanical and Aerospace Engineering, 1251 Memorial Drive, MEB 217B, Coral Gables, Florida, United States, 33146;
| | - Chun-Yuh Huang
- University of Miami, 5452, Biomedical Engineering, Coral Gables, Florida, United States;
| | - Thomas M Best
- University of Miami Miller School of Medicine, 12235, School of Medicine, Miami, Florida, United States;
| |
Collapse
|
10
|
Lückgen J, Raqué E, Reiner T, Diederichs S, Richter W. NFκB inhibition to lift the mechano-competence of mesenchymal stromal cell-derived neocartilage toward articular chondrocyte levels. Stem Cell Res Ther 2022; 13:168. [PMID: 35477424 PMCID: PMC9044876 DOI: 10.1186/s13287-022-02843-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/07/2022] [Indexed: 11/10/2022] Open
Abstract
Background Fully functional regeneration of skeletal defects by multipotent progenitor cells requires that differentiating cells gain the specific mechano-competence needed in the target tissue. Using cartilage neogenesis as an example, we asked whether proper phenotypic differentiation of mesenchymal stromal cells (MSC) into chondrocytes in vitro will install the adequate biological mechano-competence of native articular chondrocytes (AC). Methods The mechano-competence of human MSC- and AC-derived neocartilage was compared during differentiation for up to 35 days. The neocartilage layer was subjected to physiologic dynamic loading in a custom-designed bioreactor and assayed for mechano-sensitive gene and pathway activation, extracellular matrix (ECM) synthesis by radiolabel incorporation, nitric oxide (NO) and prostaglandin E2 (PGE2) production. Input from different pathways was tested by application of agonists or antagonists. Results MSC and AC formed neocartilage of similar proteoglycan content with a hardness close to native tissue. Mechano-stimulation on day 21 and 35 induced a similar upregulation of mechano-response genes, ERK phosphorylation, NO production and PGE2 release in both groups, indicating an overall similar transduction of external mechanical signals. However, while AC maintained or enhanced proteoglycan synthesis after loading dependent on tissue maturity, ECM synthesis was always significantly disturbed by loading in MSC-derived neocartilage. This was accompanied by significantly higher COX2 and BMP2 background expression, > 100-fold higher PGE2 production and a weaker SOX9 stimulation in response to loading in MSC-derived neocartilage. Anabolic BMP-pathway activity was not rate limiting for ECM synthesis after loading in both groups. However, NFκB activation mimicked the negative loading effects and enhanced PGE2 production while inhibition of catabolic NFκB signaling rescued the load-induced negative effects on ECM synthesis in MSC-derived neocartilage. Conclusions MSC-derived chondrocytes showed a higher vulnerability to be disturbed by loading despite proper differentiation and did not acquire an AC-like mechano-competence to cope with the mechanical stress of a physiologic loading protocol. Managing catabolic NFκB influences was one important adaptation to install a mechano-resistance closer to AC-derived neocartilage. This new knowledge asks for a more functional adaptation of MSC chondrogenesis, novel pharmacologic co-treatment strategies for MSC-based clinical cartilage repair strategies and may aid a more rational design of physical rehabilitation therapy after AC- versus MSC-based surgical cartilage intervention. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-02843-x.
Collapse
Affiliation(s)
- Janine Lückgen
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Elisabeth Raqué
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Tobias Reiner
- Department of Orthopaedic and Trauma Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Solvig Diederichs
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany
| | - Wiltrud Richter
- Research Centre for Experimental Orthopaedics, Heidelberg University Hospital, Schlierbacher Landstrasse 200a, 69118, Heidelberg, Germany.
| |
Collapse
|
11
|
Alizadeh Sardroud H, Wanlin T, Chen X, Eames BF. Cartilage Tissue Engineering Approaches Need to Assess Fibrocartilage When Hydrogel Constructs Are Mechanically Loaded. Front Bioeng Biotechnol 2022; 9:787538. [PMID: 35096790 PMCID: PMC8790514 DOI: 10.3389/fbioe.2021.787538] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/10/2021] [Indexed: 12/19/2022] Open
Abstract
Chondrocytes that are impregnated within hydrogel constructs sense applied mechanical force and can respond by expressing collagens, which are deposited into the extracellular matrix (ECM). The intention of most cartilage tissue engineering is to form hyaline cartilage, but if mechanical stimulation pushes the ratio of collagen type I (Col1) to collagen type II (Col2) in the ECM too high, then fibrocartilage can form instead. With a focus on Col1 and Col2 expression, the first part of this article reviews the latest studies on hyaline cartilage regeneration within hydrogel constructs that are subjected to compression forces (one of the major types of the forces within joints) in vitro. Since the mechanical loading conditions involving compression and other forces in joints are difficult to reproduce in vitro, implantation of hydrogel constructs in vivo is also reviewed, again with a focus on Col1 and Col2 production within the newly formed cartilage. Furthermore, mechanotransduction pathways that may be related to the expression of Col1 and Col2 within chondrocytes are reviewed and examined. Also, two recently-emerged, novel approaches of load-shielding and synchrotron radiation (SR)–based imaging techniques are discussed and highlighted for future applications to the regeneration of hyaline cartilage. Going forward, all cartilage tissue engineering experiments should assess thoroughly whether fibrocartilage or hyaline cartilage is formed.
Collapse
Affiliation(s)
- Hamed Alizadeh Sardroud
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- *Correspondence: Hamed Alizadeh Sardroud,
| | - Tasker Wanlin
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Mechanical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - B. Frank Eames
- Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
12
|
Uzieliene I, Bironaite D, Bernotas P, Sobolev A, Bernotiene E. Mechanotransducive Biomimetic Systems for Chondrogenic Differentiation In Vitro. Int J Mol Sci 2021; 22:9690. [PMID: 34575847 PMCID: PMC8469886 DOI: 10.3390/ijms22189690] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoarthritis (OA) is a long-term chronic joint disease characterized by the deterioration of bones and cartilage, which results in rubbing of bones which causes joint stiffness, pain, and restriction of movement. Tissue engineering strategies for repairing damaged and diseased cartilage tissue have been widely studied with various types of stem cells, chondrocytes, and extracellular matrices being on the lead of new discoveries. The application of natural or synthetic compound-based scaffolds for the improvement of chondrogenic differentiation efficiency and cartilage tissue engineering is of great interest in regenerative medicine. However, the properties of such constructs under conditions of mechanical load, which is one of the most important factors for the successful cartilage regeneration and functioning in vivo is poorly understood. In this review, we have primarily focused on natural compounds, particularly extracellular matrix macromolecule-based scaffolds and their combinations for the chondrogenic differentiation of stem cells and chondrocytes. We also discuss different mechanical forces and compression models that are used for In Vitro studies to improve chondrogenic differentiation. Summary of provided mechanical stimulation models In Vitro reviews the current state of the cartilage tissue regeneration technologies and to the potential for more efficient application of cell- and scaffold-based technologies for osteoarthritis or other cartilage disorders.
Collapse
Affiliation(s)
- Ilona Uzieliene
- State Research Institute Centre for Innovative Medicine, Department of Regenerative Medicine, LT-08406 Vilnius, Lithuania; (I.U.); (D.B.); (P.B.)
| | - Daiva Bironaite
- State Research Institute Centre for Innovative Medicine, Department of Regenerative Medicine, LT-08406 Vilnius, Lithuania; (I.U.); (D.B.); (P.B.)
| | - Paulius Bernotas
- State Research Institute Centre for Innovative Medicine, Department of Regenerative Medicine, LT-08406 Vilnius, Lithuania; (I.U.); (D.B.); (P.B.)
| | - Arkadij Sobolev
- Latvian Institute of Organic Synthesis, 21 Aizkraukles Str., LV-1006 Riga, Latvia;
| | - Eiva Bernotiene
- State Research Institute Centre for Innovative Medicine, Department of Regenerative Medicine, LT-08406 Vilnius, Lithuania; (I.U.); (D.B.); (P.B.)
| |
Collapse
|
13
|
Das P, Mishra R, Devi B, Rajesh K, Basak P, Roy M, Roy P, Lahiri D, Nandi SK. Decellularized xenogenic cartilage extracellular matrix (ECM) scaffolds for the reconstruction of osteochondral defects in rabbits. J Mater Chem B 2021; 9:4873-4894. [PMID: 34095925 DOI: 10.1039/d1tb00314c] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The use of decellularized native allogenic or xenogenic cartilaginous extracellular matrix (ECM) biomaterials is widely expanding in the fields of tissue engineering and regenerative medicine. In this study, we aimed to develop an acellular, affordable, biodegradable, easily available goat conchal cartilaginous ECM derived scaffolding biomaterial for repair and regeneration of osteochondral defects in rabbits. Cartilages harvested from freshly collected goat ears were decellularized using chemical agents, namely, hypotonic-hypertonic (HH) buffer and Triton X-100 solution, separately. The morphologies and ultrastructure orientations of the decellularized cartilages remained unaltered in spite of complete cellular loss. Furthermore, when the acellular cartilaginous ECMs were cultured with murine mesenchymal stem cells (MSCs) (C3H10T1/2 cells), cellular infiltration and proliferation were thoroughly monitored using SEM, DAPI and FDA stained images, whereas the MTT assay proved the biocompatibility of the matrices. The increasing amounts of secreted ECM proteins (collagen and sGAG) indicated successful chondrogenic differentiation of the MSCs in the presence of the treated cartilage samples. In vivo biocompatibility studies showed no significant immune response or tissue rejection in the treated samples but tissue necrosis in control samples after 3 months. Upon implantation of the constructs in rabbits' osteochondral defects for 3 months, the histological and micro-CT evaluation revealed significant enhancement and regeneration of neocartilage and subchondral bony tissues. The IGF-1 loaded cartilaginous constructs showed comparatively better healing response after 3 months. Our results showed that decellularized xenogenic cartilaginous biomaterials preserved the bioactivity and integrity of the matrices that also favored in vitro stem cell proliferation and chondrogenic differentiation and enabled osteochondral regeneration, thus paving a new way for articular cartilage reconstruction.
Collapse
Affiliation(s)
- Piyali Das
- School of Bioscience and Engineering, Jadavpur University, Kolkata, India
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Jo YK, Lee D. Biopolymer Microparticles Prepared by Microfluidics for Biomedical Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e1903736. [PMID: 31559690 DOI: 10.1002/smll.201903736] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/31/2019] [Indexed: 06/10/2023]
Abstract
Biopolymers are macromolecules that are derived from natural sources and have attractive properties for a plethora of biomedical applications due to their biocompatibility, biodegradability, low antigenicity, and high bioactivity. Microfluidics has emerged as a powerful approach for fabricating polymeric microparticles (MPs) with designed structures and compositions through precise manipulation of multiphasic flows at the microscale. The synergistic combination of materials chemistry afforded by biopolymers and precision provided by microfluidic capabilities make it possible to design engineered biopolymer-based MPs with well-defined physicochemical properties that are capable of enabling an efficient delivery of therapeutics, 3D culture of cells, and sensing of biomolecules. Here, an overview of microfluidic approaches is provided for the design and fabrication of functional MPs from three classes of biopolymers including polysaccharides, proteins, and microbial polymers, and their advances for biomedical applications are highlighted. An outlook into the future research on microfluidically-produced biopolymer MPs for biomedical applications is also provided.
Collapse
Affiliation(s)
- Yun Kee Jo
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Daeyeon Lee
- Department of Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
15
|
Park K, Lee Y, Seo J. Recent Advances in High-throughput Platforms with Engineered Biomaterial Microarrays for Screening of Cell and Tissue Behavior. Curr Pharm Des 2019; 24:5458-5470. [DOI: 10.2174/1381612825666190207093438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/02/2019] [Indexed: 02/06/2023]
Abstract
In the last decades, bioengineers have developed myriad biomaterials for regenerative medicine. Development of screening techniques is essential for understanding complex behavior of cells in the biological microenvironments. Conventional approaches to the screening of cellular behavior in vitro have limitations in terms of accuracy, reusability, labor-intensive screening, and versatility. Thus, drug screening and toxicology test through in vitro screening platforms have been underwhelming. Recent advances in the high-throughput screening platforms somewhat overcome the limitations of in vitro screening platforms via repopulating human tissues’ biophysical and biomchemical microenvironments with the ability to continuous monitoring of miniaturized human tissue behavior. Herein, we review current trends in the screening platform in which a high-throughput system composed of engineered microarray devices is developed to investigate cell-biomaterial interaction. Furthermore, diverse methods to achieve continuous monitoring of cell behavior via developments of biosensor integrated high-throughput platforms, and future perspectives on high-throughput screening will be provided.
Collapse
Affiliation(s)
- Kijun Park
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| | - Yeontaek Lee
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| | - Jungmok Seo
- School of Electrical and Electronic Engineering, Yonsei University, Seoul, 03722, Korea
| |
Collapse
|
16
|
Baumgartner W, Otto L, Hess SC, Stark WJ, Märsmann S, Bürgisser GM, Calcagni M, Cinelli P, Buschmann J. Cartilage/bone interface fabricated under perfusion: Spatially organized commitment of adipose‐derived stem cells without medium supplementation. J Biomed Mater Res B Appl Biomater 2018; 107:1833-1843. [DOI: 10.1002/jbm.b.34276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 09/27/2018] [Accepted: 10/17/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Walter Baumgartner
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Lukas Otto
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Samuel C. Hess
- Institute for Chemical‐ and BioengineeringDepartment of Chemistry and Applied Biosciences ETH Zürich, Zürich Switzerland
| | - Wendelin J. Stark
- Institute for Chemical‐ and BioengineeringDepartment of Chemistry and Applied Biosciences ETH Zürich, Zürich Switzerland
| | - Sonja Märsmann
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
- Division of Trauma SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | | | - Maurizio Calcagni
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Paolo Cinelli
- Division of Trauma SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| | - Johanna Buschmann
- Division of Plastic and Hand SurgeryUniversity Hospital Zürich ZKF, Zürich Switzerland
| |
Collapse
|
17
|
Lipphaus A, Witzel U. Finite-Element Syntheses of Callus and Bone Remodeling: Biomechanical Study of Fracture Healing in Long Bones. Anat Rec (Hoboken) 2018; 301:2112-2121. [PMID: 30290071 DOI: 10.1002/ar.23893] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 01/26/2018] [Accepted: 02/21/2018] [Indexed: 12/11/2022]
Abstract
Computational simulations of fracture healing are a valuable tool to improve fracture treatment and implants. Several finite-element models have been established to predict callus formation due to mechanobiological rules. This work provides a comprehensive simulation for virtual implantation through the combination of callus simulation and finite-element structural synthesis (FESS) of (re-)modeling during and after healing based on Pauwel's theory of histogenesis and Wolff's law. The simulation is based on a linear elastic material model and includes generation of fracture hematoma and initial mesenchymal stem cell concentration out of an unspecified solid, cell proliferation, migration, and differentiation due to mechanical stimuli and time-dependent axial loading. Three nondisplaced femoral shaft fractures with initial interfragmentary movement of 0.2, 0.6, and 1 mm and one fracture with 4 mm translation are modeled. The predictions of interfragmentary movement during fracture healing, healing success, and healing time agree with observed clinical outcome, animal models, and other numerical models. Initial interfragmentary movement between 0.2 and 1 mm leads to healing success, with the fastest healing occurring at 0.2 mm. The model of the dislocated fractures shows no further bending after remodeling and is loaded with physiological stress of -13 MPa. Ideal load-time graphs may give insight into the bone's ability to withstand loads as healing time progresses, and thus holds potential for applications in rehabilitation planning. Better knowledge of the forces present during fracture healing is needed to deploy simulations for surgical planning and manufacturing of patient individualized implants. Anat Rec, 301:2112-2121, 2018. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Andreas Lipphaus
- Research Group of Biomechanics, Chair of Product Development, Department of Mechanical Engineering, Ruhr-University Bochum, Universitätstr. 150, Bochum, DE 44801, Germany
| | - Ulrich Witzel
- Research Group of Biomechanics, Chair of Product Development, Department of Mechanical Engineering, Ruhr-University Bochum, Universitätstr. 150, Bochum, DE 44801, Germany
| |
Collapse
|
18
|
Wang Z, Li Z, Li Z, Wu B, Liu Y, Wu W. Cartilaginous extracellular matrix derived from decellularized chondrocyte sheets for the reconstruction of osteochondral defects in rabbits. Acta Biomater 2018; 81:129-145. [PMID: 30300711 DOI: 10.1016/j.actbio.2018.10.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/20/2018] [Accepted: 10/04/2018] [Indexed: 12/28/2022]
Abstract
Cartilaginous extracellular matrix (ECM) materials derived from decellularized native articular cartilage are widely used in cartilage regeneration. However, it is difficult for endogenous cells to migrate into ECM derived from native cartilage owing to its nonporous structure and dense nature. Moreover, current decellularization approaches frequently lead to architectural breakdown and potential loss of surface composition of ECM. To solve this problem, we aimed to establish a novel biological ECM scaffold from chondrocyte sheets for cartilage regeneration. We cultured chondrocytes harvested from the auricular cartilage of 4-week-old New Zealand rabbits and enabled them to form cell sheets. These sheets were decellularized using sodium dodecyl sulfate (SDS) with three different concentrations, namely, 1%, 5%, and 10%, followed by 1% Triton X-100 and deoxyribonuclease enzyme solution. In vitro microstructural examination and mechanical tests demonstrated that 1% SDS not only removed chondrocytes completely but also maintained the native architecture and composition of ECM, thus avoiding the use of high-concentration SDS. Application of decellularized chondrocyte sheets for osteochondral defects in rabbits resulted in substantial host remodeling and variant regeneration of osteochondral tissues. One percent SDS-treated decellularized chondrocyte sheets contributed to the superior reconstruction of osteochondral defects as compared with 5% and 10% SDS groups, which includes vascularized subchondral bone, articular cartilage with adequate thickness, and integration with host tissues. Furthermore, ECM from 1% SDS significantly increased the migrating potential of bone marrow mesenchymal stem cells (BMSCs) in vitro. RT-PCR and western blot also revealed that ECM increased the expression of SOX-9 in BMSCs, whereas it decreased COL-X expression. In conclusion, our results suggested that the chondrocyte sheets decellularized with 1% SDS preserved the integrity and bioactivity, which favored cell recruitment and enabled osteochondral regeneration in the knee joints of rabbits, thus offering a promising approach for articular cartilage reconstruction without cell transplantation. STATEMENT OF SIGNIFICANCE: Although biological extracellular matrix (ECM) derived from decellularized native cartilage has been widely used in cartilage regeneration, it is difficult for endogenous cells to migrate into ECM owing to its dense nature. Moreover, current decellularization approaches lead to architectural breakdown of ECM. This study established a novel biological ECM from decellularized chondrocyte sheets for cartilage regeneration. Our results suggested that cartilaginous ECM favored cell recruitment and enabled osteochondral regeneration in rabbits, thus offering a promising approach for articular cartilage reconstruction without cell transplantation. SDS 1% adequately decellularized the chondrocytes in cell sheets, whereas it maintained the native architecture and composition of ECM, thereby avoiding the use of high-concentration SDS and providing a new way to acquire cartilaginous ECM.
Collapse
|
19
|
Augustine R. Skin bioprinting: a novel approach for creating artificial skin from synthetic and natural building blocks. Prog Biomater 2018; 7:77-92. [PMID: 29754201 PMCID: PMC6068049 DOI: 10.1007/s40204-018-0087-0] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/29/2018] [Indexed: 12/12/2022] Open
Abstract
Significant progress has been made over the past few decades in the development of in vitro-engineered substitutes that mimic human skin, either as grafts for the replacement of lost skin, or for the establishment of in vitro human skin models. Tissue engineering has been developing as a novel strategy by employing the recent advances in various fields such as polymer engineering, bioengineering, stem cell research and nanomedicine. Recently, an advancement of 3D printing technology referred as bioprinting was exploited to make cell loaded scaffolds to produce constructs which are more matching with the native tissue. Bioprinting facilitates the simultaneous and highly specific deposition of multiple types of skin cells and biomaterials, a process that is lacking in conventional skin tissue-engineering approaches. Bioprinted skin substitutes or equivalents containing dermal and epidermal components offer a promising approach in skin bioengineering. Various materials including synthetic and natural biopolymers and cells with or without signalling molecules like growth factors are being utilized to produce functional skin constructs. This technology emerging as a novel strategy to overcome the current bottle-necks in skin tissue engineering such as poor vascularization, absence of hair follicles and sweat glands in the construct.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha, 2713, Qatar.
| |
Collapse
|
20
|
Polo-Corrales L, Ramirez-Vick J, Feria-Diaz JJ. Recent Advances in Biophysical stimulation of MSC for bone regeneration. ACTA ACUST UNITED AC 2018. [DOI: 10.17485/ijst/2018/v11i15/121405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
21
|
Sawatjui N, Limpaiboon T, Schrobback K, Klein T. Biomimetic scaffolds and dynamic compression enhance the properties of chondrocyte‐ and
MSC
‐based tissue‐engineered cartilage. J Tissue Eng Regen Med 2018; 12:1220-1229. [DOI: 10.1002/term.2653] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 12/21/2017] [Accepted: 02/17/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Nopporn Sawatjui
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences Khon Kaen University Khon Kaen Thailand
| | - Temduang Limpaiboon
- Centre for Research and Development of Medical Diagnostic Laboratories, Faculty of Associated Medical Sciences Khon Kaen University Khon Kaen Thailand
| | - Karsten Schrobback
- Cartilage Regeneration Laboratory, Institute of Health and Biomedical Innovation Queensland University of Technology Brisbane Queensland Australia
| | - Travis Klein
- Cartilage Regeneration Laboratory, Institute of Health and Biomedical Innovation Queensland University of Technology Brisbane Queensland Australia
| |
Collapse
|
22
|
A microfabricated platform for the study of chondrogenesis under different compressive loads. J Mech Behav Biomed Mater 2018; 78:404-413. [DOI: 10.1016/j.jmbbm.2017.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 11/29/2017] [Accepted: 12/01/2017] [Indexed: 01/09/2023]
|
23
|
Boehme KA, Schleicher SB, Traub F, Rolauffs B. Chondrosarcoma: A Rare Misfortune in Aging Human Cartilage? The Role of Stem and Progenitor Cells in Proliferation, Malignant Degeneration and Therapeutic Resistance. Int J Mol Sci 2018; 19:ijms19010311. [PMID: 29361725 PMCID: PMC5796255 DOI: 10.3390/ijms19010311] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 01/07/2018] [Accepted: 01/18/2018] [Indexed: 02/07/2023] Open
Abstract
Unlike other malignant bone tumors including osteosarcomas and Ewing sarcomas with a peak incidence in adolescents and young adults, conventional and dedifferentiated chondrosarcomas mainly affect people in the 4th to 7th decade of life. To date, the cell type of chondrosarcoma origin is not clearly defined. However, it seems that mesenchymal stem and progenitor cells (MSPC) in the bone marrow facing a pro-proliferative as well as predominantly chondrogenic differentiation milieu, as is implicated in early stage osteoarthritis (OA) at that age, are the source of chondrosarcoma genesis. But how can MSPC become malignant? Indeed, only one person in 1,000,000 will develop a chondrosarcoma, whereas the incidence of OA is a thousandfold higher. This means a rare coincidence of factors allowing escape from senescence and apoptosis together with induction of angiogenesis and migration is needed to generate a chondrosarcoma. At early stages, chondrosarcomas are still assumed to be an intermediate type of tumor which rarely metastasizes. Unfortunately, advanced stages show a pronounced resistance both against chemo- and radiation-therapy and frequently metastasize. In this review, we elucidate signaling pathways involved in the genesis and therapeutic resistance of chondrosarcomas with a focus on MSPC compared to signaling in articular cartilage (AC).
Collapse
Affiliation(s)
- Karen A Boehme
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79108 Freiburg, Germany.
| | - Sabine B Schleicher
- Department of Hematology and Oncology, Eberhard Karls University Tuebingen, Children's Hospital, 72076 Tuebingen, Germany.
| | - Frank Traub
- Department of Orthopedic Surgery, Eberhard Karls University Tuebingen, 72076 Tuebingen, Germany.
| | - Bernd Rolauffs
- G.E.R.N. Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Medical Center-Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79108 Freiburg, Germany.
| |
Collapse
|
24
|
Palit S, Khajehpour Tadavani S, Yethiraj A. Realization of a stable, monodisperse water-in-oil droplet system with micro-scale and nano-scale confinement for tandem microscopy and diffusion NMR studies. SOFT MATTER 2018; 14:448-459. [PMID: 29261208 DOI: 10.1039/c7sm01508a] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
In this work we generate stable and monodisperse water-in-oil emulsions using a co-flowing geometry that produced droplet sizes between 13 μm and 250 μm. The drops survived transfer to NMR tubes and were stable for at least 26 hours, enabling the performance of pulsed-field-gradient NMR experiments in addition to microscopy. The drops sizes achieved as a function of flow rate agree well with a simple model for droplet generation: this yields a precise measure of the interfacial tension. The design of a cell mimetic environment with nano-scale confinement has also been demonstrated with diffusion measurements on macromolecules (PEG and Ficoll70) within droplets that are further structured internally using agarose gel networks. Containing the agarose gel in droplets appears to provide very reproducible and homogeneous network environments, enabling quantitative agreement of Ficoll70 dynamics with a theoretical model, with no fit parameters, and, with PEG, yielding a systematic polymer-size dependent slowing down in the network. This is in contrast with bulk agarose, where identical macromolecular diffusion measurements indicate the presence of heterogeneities with water pockets.
Collapse
Affiliation(s)
- Swomitra Palit
- Physics and Physical Oceanography, Memorial University of Newfoundland, St. John's, NL A1B 3X7, Canada.
| | | | | |
Collapse
|
25
|
Seo J, Shin JY, Leijten J, Jeon O, Camci-Unal G, Dikina AD, Brinegar K, Ghaemmaghami AM, Alsberg E, Khademhosseini A. High-throughput approaches for screening and analysis of cell behaviors. Biomaterials 2018; 153:85-101. [PMID: 29079207 PMCID: PMC5702937 DOI: 10.1016/j.biomaterials.2017.06.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 06/17/2017] [Accepted: 06/19/2017] [Indexed: 02/06/2023]
Abstract
The rapid development of new biomaterials and techniques to modify them challenge our capability to characterize them using conventional methods. In response, numerous high-throughput (HT) strategies are being developed to analyze biomaterials and their interactions with cells using combinatorial approaches. Moreover, these systematic analyses have the power to uncover effects of delivered soluble bioactive molecules on cell responses. In this review, we describe the recent developments in HT approaches that help identify cellular microenvironments affecting cell behaviors and highlight HT screening of biochemical libraries for gene delivery, drug discovery, and toxicological studies. We also discuss HT techniques for the analyses of cell secreted biomolecules and provide perspectives on the future utility of HT approaches in biomedical engineering.
Collapse
Affiliation(s)
- Jungmok Seo
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Center for Biomaterials, Korea Institute of Science and Technology, 14 Hwarang-ro, Seongbuk-gu, Seoul, 02792, South Korea
| | - Jung-Youn Shin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Jeroen Leijten
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Department of Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Oju Jeon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Gulden Camci-Unal
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Department of Chemical Engineering, University of Massachusetts Lowell, 1 University Ave, Lowell, MA, 01854-2827, USA
| | - Anna D Dikina
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Katelyn Brinegar
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA
| | - Amir M Ghaemmaghami
- Division of Immunology, School of Life Sciences, Faculty of Medicine and Health Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, 44106, USA; Department of Orthopaedic Surgery, Case Western Reserve University, Cleveland, OH, 44106, USA; National Center for Regenerative Medicine, Division of General Medical Sciences, Case Western Reserve University, Cleveland, OH, 44106, USA.
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA; Department of Bioindustrial Technologies, College of Animal Bioscience and Technology, Konkuk University, Hwayang-dong, Gwangjin-gu, Seoul, 143-701, Republic of Korea; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA; Department of Physics, King Abdulaziz University, Jeddah, 21569, Saudi Arabia.
| |
Collapse
|
26
|
Du V, Luciani N, Richard S, Mary G, Gay C, Mazuel F, Reffay M, Menasché P, Agbulut O, Wilhelm C. A 3D magnetic tissue stretcher for remote mechanical control of embryonic stem cell differentiation. Nat Commun 2017; 8:400. [PMID: 28900152 PMCID: PMC5596024 DOI: 10.1038/s41467-017-00543-2] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 07/06/2017] [Indexed: 12/11/2022] Open
Abstract
The ability to create a 3D tissue structure from individual cells and then to stimulate it at will is a major goal for both the biophysics and regenerative medicine communities. Here we show an integrated set of magnetic techniques that meet this challenge using embryonic stem cells (ESCs). We assessed the impact of magnetic nanoparticles internalization on ESCs viability, proliferation, pluripotency and differentiation profiles. We developed magnetic attractors capable of aggregating the cells remotely into a 3D embryoid body. This magnetic approach to embryoid body formation has no discernible impact on ESC differentiation pathways, as compared to the hanging drop method. It is also the base of the final magnetic device, composed of opposing magnetic attractors in order to form embryoid bodies in situ, then stretch them, and mechanically stimulate them at will. These stretched and cyclic purely mechanical stimulations were sufficient to drive ESCs differentiation towards the mesodermal cardiac pathway. The development of embryoid bodies that are responsive to external stimuli is of great interest in tissue engineering. Here, the authors culture embryonic stem cells with magnetic nanoparticles and show that the presence of magnetic fields could affect their aggregation and differentiation.
Collapse
Affiliation(s)
- Vicard Du
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Nathalie Luciani
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Sophie Richard
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Gaëtan Mary
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Cyprien Gay
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - François Mazuel
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Myriam Reffay
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France
| | - Philippe Menasché
- Department of Cardiovascular Surgery, Hôpital Européen Georges Pompidou; Paris Cardiovascular Research Center, INSERM U970, Université Paris Descartes, Paris, 75015, France
| | - Onnik Agbulut
- Sorbonne Universités, UPMC Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, 75005, Paris, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes (MSC), UMR 7057, CNRS and Université Paris Diderot, 75205, Paris Cedex 13, France.
| |
Collapse
|
27
|
Cho W, Kim ES, Kang CM, Ji YH, Kim JI, Park SJ, Son Y, Kim CH. Low-Dose Ionizing γ-Radiation Promotes Proliferation of Human Mesenchymal Stem Cells and Maintains Their Stem Cell Characteristics. Tissue Eng Regen Med 2017; 14:421-432. [PMID: 30603498 PMCID: PMC6171613 DOI: 10.1007/s13770-017-0045-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 10/04/2016] [Accepted: 10/16/2016] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs), which are multipotent and have self-renewal ability, support the regeneration of damaged normal tissue. A number of external stimuli promote migration of MSCs into peripheral blood and support their participation in wound healing. In an attempt to harness the potential beneficial effects of such external stimuli, we exposed human MSCs (hMSCs) to one such stimulus-low-dose ionizing radiation (LDIR)-and examined their biological properties. To this end, we evaluated differences in proliferation, cell cycle, DNA damage, expression of surface markers (CD29, CD34, CD90, and CD105), and differentiation potential of hMSCs before and after irradiation with γ-rays generated using a 137CS irradiator. At doses less than 50 mGy, LDIR had no significant effect on the viability or apoptosis of hMSCs. Interestingly, 10 mGy of LDIR increased hMSC viability by 8% (p < 0.001) compared with non-irradiated hMSCs. At doses less than 50 mGy, LDIR did not induce DNA damage, including DNA strand breaks, or cause cellular senescence or cell-cycle arrest. Surface marker expression and in vitro differentiation potential of hMSCs were maintained after two exposures to LDIR at 10 mGy per dose. In conclusion, a two-dose exposure to LDIR at 10 mGy per dose not only facilitates proliferation of hMSCs, it also maintains the stem cell characteristics of hMSCs without affecting their viability. These results provide evidence for the potential of LDIR as an external stimulus for in vitro expansion of hMSCs and application in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Wheemoon Cho
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812 Korea
- Department of Genetic Engineering, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, 1732, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104 Korea
| | - E Ske Kim
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812 Korea
| | - Chang Mo Kang
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812 Korea
| | - Young-Hoon Ji
- Research Center for Radiotherapy, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812 Korea
| | - Jong-Il Kim
- Department of Food and Microbial Technology, Seoul Women’s University, 621 Hwaran-ro, Nowon-gu, Seoul, 01797 Korea
| | - Sang Jun Park
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812 Korea
| | - Youngsook Son
- Department of Genetic Engineering, College of Life Sciences and Graduate School of Biotechnology, Kyung Hee University, 1732, Deogyeong-daero, Giheung-gu, Yongin-si, Gyeonggi-do 17104 Korea
| | - Chun-Ho Kim
- Laboratory of Tissue Engineering, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812 Korea
| |
Collapse
|
28
|
Scholtes S, Krämer E, Weisser M, Roth W, Luginbühl R, Grossner T, Richter W. Global chondrocyte gene expression after a single anabolic loading period: Time evolution and re-inducibility of mechano-responses. J Cell Physiol 2017; 233:699-711. [PMID: 28369921 DOI: 10.1002/jcp.25933] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 03/24/2017] [Indexed: 11/09/2022]
Abstract
Aim of this study was a genome-wide identification of mechano-regulated genes and candidate pathways in human chondrocytes subjected to a single anabolic loading episode and characterization of time evolution and re-inducibility of the response. Osteochondral constructs consisting of a chondrocyte-seeded collagen-scaffold connected to β-tricalcium-phosphate were pre-cultured for 35 days and subjected to dynamic compression (25% strain, 1 Hz, 9 × 10 min over 3 hr) before microarray-profiling was performed. Proteoglycan synthesis was determined by 35 S-sulfate-incorporation over 24 hr. Cell viability and hardness of constructs were unaltered by dynamic compression while proteoglycan synthesis was significantly stimulated (1.45-fold, p = 0.016). Among 115 significantly regulated genes, 114 were up-regulated, 48 of them ≥ twofold. AP-1-relevant transcription factors FOSB and FOS strongly increased in line with elevated ERK1/2-phosphorylation and rising MAP3K4 expression. Expression of proteoglycan-synthesizing enzymes CHSY1 and GALNT4 was load-responsive as were factors associated with the MAPK-, TGF-β-, calcium-, retinoic-acid-, Wnt-, and Notch-signaling pathway which were significantly upregulated SOX9, and BMP6 levels rose significantly also after multiple loading episodes at daily intervals even at the 14th cycle with no indication for desensitation. Canonical pSmad2/3 and pSmad1/5/9-signaling showed no consistent regulation. This study associates novel genes with mechanoregulation in chondrocytes, raising SOX9 protein levels with anabolic loading and suggests that more pathways than so far anticipated apparently work together in a complex network of stimulators and feedback-regulators. Upregulation of mechanosensitive indicators extending differentially into the resting time provides crucial knowledge to maximize cartilage matrix deposition for the generation of high-level cartilage replacement tissue.
Collapse
Affiliation(s)
- Simone Scholtes
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | - Elisabeth Krämer
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | - Melanie Weisser
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | - Wolfgang Roth
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | | | - Tobias Grossner
- Department of Orthopaedic and Trauma Surgery, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Wiltrud Richter
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| |
Collapse
|
29
|
Horner CB, Hirota K, Liu J, Maldonado M, Hyle Park B, Nam J. Magnitude‐dependent and inversely‐related osteogenic/chondrogenic differentiation of human mesenchymal stem cells under dynamic compressive strain. J Tissue Eng Regen Med 2017; 12:e637-e647. [DOI: 10.1002/term.2332] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 08/01/2016] [Accepted: 09/26/2016] [Indexed: 01/02/2023]
Affiliation(s)
| | - Koji Hirota
- Department of BioengineeringUniversity of California Riverside CA 92521 USA
| | - Junze Liu
- Department of BioengineeringUniversity of California Riverside CA 92521 USA
| | - Maricela Maldonado
- Department of BioengineeringUniversity of California Riverside CA 92521 USA
| | - B. Hyle Park
- Department of BioengineeringUniversity of California Riverside CA 92521 USA
| | - Jin Nam
- Department of BioengineeringUniversity of California Riverside CA 92521 USA
| |
Collapse
|
30
|
Seyedi F, Farsinejad A, Nematollahi-Mahani SN. Fibrin scaffold enhances function of insulin producing cells differentiated from human umbilical cord matrix-derived stem cells. Tissue Cell 2017; 49:227-232. [DOI: 10.1016/j.tice.2017.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/05/2017] [Indexed: 11/29/2022]
|
31
|
Peroglio M, Douma LS, Caprez TS, Janki M, Benneker LM, Alini M, Grad S. Intervertebral disc response to stem cell treatment is conditioned by disc state and cell carrier: An ex vivo study. J Orthop Translat 2017; 9:43-51. [PMID: 29662798 PMCID: PMC5822953 DOI: 10.1016/j.jot.2017.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 12/26/2022] Open
Abstract
In vitro and in vivo studies evidenced that mesenchymal stem cells (MSCs) contribute to intervertebral disc (IVD) regeneration by differentiation towards the disc phenotype and matrix synthesis and/or by paracrine signalling to endogenous cells, thereby promoting a healthier disc phenotype in degenerative discs. The aim of this study was to investigate IVD response to human MSC (hMSC) treatment based on the disc degenerative state and hMSC carrier. Bovine caudal IVDs with endplates were cultured in a bioreactor under simulated physiological (0.1 Hz load and sufficient glucose) or degenerative (10 Hz load and limited glucose) conditions for 7 days. Discs were partially nucleotomised, restored with hMSCs in either fibrin gel or saline solution and cultured under physiological conditions for 7 days. Controls included fibrin and saline without hMSCs. Cell viability, histology, disc height, and gene expression analyses were performed to evaluate regeneration. hMSCs in fibrin were viable and homogenously distributed following 7 days of culture under dynamic loading in partially nucleotomised discs. IVD response to hMSCs was conditioned by both disc degenerative state and hMSC carrier. The effect of the regenerative treatment was stronger on simulated-degenerative discs than on simulated-physiological discs. hMSCs in fibrin induced a superior anabolic response in degenerative IVDs compared with fibrin alone, thus suggesting an added value of the cellular therapy compared with an acellular solution. When comparing fibrin and saline as a hMSC carrier, a significantly higher anabolic response was observed in IVDs treated with hMSCs in fibrin. Moreover, it was found that the degenerative state of the disc influenced hMSC differentiation. Indeed, a significantly higher expression of specific discogenic markers (ACAN and CA12) was observed in hMSCs implanted into physiological discs than in those implanted into degenerative discs. In conclusion, host disc cells and donor hMSC response depend on the degenerative state of the host disc and carrier used for hMSC delivery, and these two aspects need to be considered for a successful translation of hMSC therapies for the treatment of IVD degeneration.
Collapse
Affiliation(s)
| | | | | | | | | | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland
| | | |
Collapse
|
32
|
|
33
|
Cardiomyogenesis of periodontal ligament-derived stem cells by dynamic tensile strain. Cell Tissue Res 2016; 367:229-241. [DOI: 10.1007/s00441-016-2503-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 09/06/2016] [Indexed: 12/25/2022]
|
34
|
Mechanical Stimulation Increases Knee Meniscus Gene RNA-level Expression in Adipose-derived Stromal Cells. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2016; 4:e864. [PMID: 27757329 PMCID: PMC5054995 DOI: 10.1097/gox.0000000000000854] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 06/15/2016] [Indexed: 01/22/2023]
Abstract
Supplemental Digital Content is available in the text. Efforts have been made to engineer knee meniscus tissue for injury repair, yet most attempts have been unsuccessful. Creating a cell source that resembles the complex, heterogeneous phenotype of the meniscus cell remains difficult. Stem cell differentiation has been investigated, mainly using bone marrow mesenchymal cells and biochemical means for differentiation, resulting in no solution. Mechanical stimulation has been investigated to an extent with no conclusion. Here, we explore the potential for and effectiveness of mechanical stimulation to induce the meniscal phenotype in adipose-derived stromal cells.
Collapse
|
35
|
Popa EG, Reis RL, Gomes ME. Seaweed polysaccharide-based hydrogels used for the regeneration of articular cartilage. Crit Rev Biotechnol 2016; 35:410-24. [PMID: 24646368 DOI: 10.3109/07388551.2014.889079] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This manuscript provides an overview of the in vitro and in vivo studies reported in the literature focusing on seaweed polysaccharides based hydrogels that have been proposed for applications in regenerative medicine, particularly, in the field of cartilage tissue engineering. For a better understanding of the main requisites for these specific applications, the main aspects of the native cartilage structure, as well as recognized diseases that affect this tissue are briefly described. Current available treatments are also presented to emphasize the need for alternative techniques. The following part of this review is centered on the description of the general characteristics of algae polysaccharides, as well as relevant properties required for designing hydrogels for cartilage tissue engineering purposes. An in-depth overview of the most well known seaweed polysaccharide, namely agarose, alginate, carrageenan and ulvan biopolymeric gels, that have been proposed for engineering cartilage is also provided. Finally, this review describes and summarizes the translational aspect for the clinical application of alternative systems emphasizing the importance of cryopreservation and the commercial products currently available for cartilage treatment.
Collapse
Affiliation(s)
- Elena Geta Popa
- a 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine , AvePark , Guimarães , Portugal and
| | | | | |
Collapse
|
36
|
Extracorporeal shockwave therapy promotes chondrogenesis in cartilage tissue engineering: A hypothesis based on previous evidence. Med Hypotheses 2016; 91:9-15. [DOI: 10.1016/j.mehy.2016.03.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 03/17/2016] [Accepted: 03/25/2016] [Indexed: 01/08/2023]
|
37
|
Aisenbrey EA, Bryant SJ. Mechanical loading inhibits hypertrophy in chondrogenically differentiating hMSCs within a biomimetic hydrogel. J Mater Chem B 2016; 4:3562-3574. [PMID: 27499854 PMCID: PMC4972607 DOI: 10.1039/c6tb00006a] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Three dimensional hydrogels are a promising vehicle for delivery of adult human bone-marrow derived mesenchymal stem cells (hMSCs) for cartilage tissue engineering. One of the challenges with using this cell type is the default pathway is terminal differentiation, a hypertrophic phenotype and precursor to endochondral ossification. We hypothesized that a synthetic hydrogel consisting of extracellular matrix (ECM) analogs derived from cartilage when combined with dynamic loading provides physiochemical cues for achieving a stable chondrogenic phenotype. Hydrogels were formed from crosslinked poly(ethylyene glycol) as the base chemistry and to which (meth)acrylate functionalized ECM analogs of RGD (cell adhesion peptide) and chondroitin sulfate (ChS, a negatively charged glycosaminoglycan) were introduced. Bone-marrow derived hMSCs from three donors were encapsulated in the hydrogels and cultured under free swelling conditions or under dynamic com pressive loading with 2.5 ng/ml TGF-β3. hMSC differentiation was assessed by quantitative PCR and immunohistochemistry. Nine hydrogel formulations were initially screened containing 0, 0.1 or 1mM RGD and 0, 1 or 2wt% ChS. After 21 days, the 1% ChS and 0.1 mM RGD hydrogel had the highest collagen II gene expression, but this was accompanied by high collagen X gene expression. At the protein level, collagen II was detected in all formulations with ECM analogs, but minimally detectable in the hydrogel without ECM analogs. Collagen X protein was present in all formulations. The 0.1 mM RGD and 1% ChS formulation was selected and subjected to five loading regimes: no loading, 5% strain 0.3Hz (1.5%/s), 10% strain 0.3 Hz (3%/s), 5% strain 1 Hz (5%/s), and 10% strain 1Hz (10%/s). After 21 days, ~70-90% of cells stained positive for collagen II protein regardless of the culture condition. On the contrary, only ~20-30% of cells stained positive for collagen X protein under 3 and 5%/s loading conditions, which was accompanied by minimal staining for RunX2. The other culture conditions had more cells staining positive for collagen X (40-60%) and was accompanied by positive staining for RunX2. In summary, a cartilage-like biomimetic hydrogel supports chondrogenesis of hMSCs, but dynamic loading only under select strain rates is able to inhibit hypertrophy.
Collapse
Affiliation(s)
- E A Aisenbrey
- University of Colorado,Boulder. Chemical and Biological Engineering, UCB 596. Boulder, CO. 80309
| | - S J Bryant
- University of Colorado,Boulder. Chemical and Biological Engineering, UCB 596. Boulder, CO. 80309
| |
Collapse
|
38
|
Teng S, Liu C, Guenther D, Omar M, Neunaber C, Krettek C, Jagodzinski M. Influence of biomechanical and biochemical stimulation on the proliferation and differentiation of bone marrow stromal cells seeded on polyurethane scaffolds. Exp Ther Med 2016; 11:2086-2094. [PMID: 27284290 PMCID: PMC4888012 DOI: 10.3892/etm.2016.3206] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 02/19/2016] [Indexed: 02/06/2023] Open
Abstract
The aim of the present investigation was to compare the effects of cyclic compression, perfusion, dexamethasone (DEX) and bone morphogenetic protein-7 (BMP-7) on the proliferation and differentiation of human bone marrow stromal cells (hBMSCs) in polyurethane scaffolds in a perfusion bioreactor. Polyurethane scaffolds seeded with hBMSCs were cultured under six different conditions, as follows: 10% Cyclic compression at 0.5 and 5 Hz; 10 ml/min perfusion; 100 nM DEX; 100 ng/ml BMP-7; and 1 ml/min perfusion without mechanical and biochemical stimulation (control). On days 7 and 14, samples were tested for the following data: Cell proliferation; mRNA expression of Runx2, COL1A1 and osteocalcin; osteocalcin content; calcium deposition; and the equilibrium modulus of the tissue specimen. The results indicated that BMP-7 and 10 ml/min perfusion promoted cell proliferation, which was inhibited by 5 Hz cyclic compression and DEX. On day 7, the 5 Hz cyclic compression inhibited Runx2 expression, whereas the 0.5 Hz cyclic compression and BMP-7 upregulated the COL1A1 mRNA levels on day 7 and enhanced the osteocalcin expression on day 14. The DEX-treated hBMSCs exhibited downregulated osteocalcin expression. After 14 days, the BMP-7 group exhibited the highest calcium deposition, followed by the 0.5 Hz cyclic compression and the DEX groups. The equilibrium modulus of the engineered constructs significantly increased in the BMP-7, 0.5 Hz cyclic compression and DEX groups. In conclusion, the present results suggest that BMP-7 and perfusion enhance cell proliferation, whereas high frequency cyclic compression inhibits the proliferation and osteogenic differentiation of hBMSCs. Low frequency cyclic compression is more effective than DEX, but less effective compared with BMP-7 on the osteogenic differentiation of hBMSCs seeded on polyurethane scaffolds.
Collapse
Affiliation(s)
- Songsong Teng
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Chaoxu Liu
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Daniel Guenther
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Mohamed Omar
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Claudia Neunaber
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Christian Krettek
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Jagodzinski
- Department of Orthopedic Trauma, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
39
|
Panadero J, Lanceros-Mendez S, Ribelles JG. Differentiation of mesenchymal stem cells for cartilage tissue engineering: Individual and synergetic effects of three-dimensional environment and mechanical loading. Acta Biomater 2016; 33:1-12. [PMID: 26826532 DOI: 10.1016/j.actbio.2016.01.037] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 12/17/2015] [Accepted: 01/25/2016] [Indexed: 12/22/2022]
Abstract
Chondrogenesis of dedifferentiated chondrocytes and mesenchymal stem cells is influenced not only by soluble molecules like growth factors, but also by the cell environment itself. The latter is achieved through both mechanical cues - which act as stimulation factor and influences nutrient transport - and adhesion to extracellular matrix cues - which determine cell shape. Although the effects of soluble molecules and cell environment have been intensively addressed, few observations and conclusions about the interaction between the two have been achieved. In this work, we review the state of the art on the single effects between mechanical and biochemical cues, as well as on the combination of the two. Furthermore, we provide a discussion on the techniques currently used to determine the mechanical properties of materials and tissues generated in vitro, their limitations and the future research needs to properly address the identified problems. STATEMENT OF SIGNIFICANCE The importance of biomechanical cues in chondrogenesis is well known. This paper reviews the existing literature on the effect of mechanical stimulation on chondrogenic differentiation of mesenchymal stem cells in order to regenerate hyaline cartilage. Contradictory results found with respect to the effect of different modes of external loading can be explained by the different properties of the scaffolding system that holds the cells, which determine cell adhesion and morphology and spatial distribution of cells, as well as the stress transmission to the cells. Thus, this review seeks to provide an insight into the interplay between external loading program and scaffold properties during chondrogenic differentiation. The review of the literature reveals an important gap in the knowledge in this field and encourages new experimental studies. The main issue is that in each of the few cases in which the interplay is investigated, just two groups of scaffolds are compared, leaving intermediate adhesion conditions out of study. The authors propose broader studies implementing new high-throughput techniques for mechanical characterization of tissue engineering constructs and the inclusion of fatigue analysis as support methodology to more exhaustive mechanical characterization.
Collapse
|
40
|
Pelaez D, H. Michel J, S. Cheung H. Growth on elastic silicone substrate elicits a partial myogenic response in periodontal ligament derived stem cells. AIMS BIOENGINEERING 2016. [DOI: 10.3934/bioeng.2016.4.515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
41
|
Miao T, Miller EJ, McKenzie C, Oldinski RA. Physically crosslinked polyvinyl alcohol and gelatin interpenetrating polymer network theta-gels for cartilage regeneration. J Mater Chem B 2015; 3:9242-9249. [PMID: 32262923 DOI: 10.1039/c5tb00989h] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Theta-gels are hydrogels that form during the solidification and phase separation of two dislike polymers, in which a low molecular weight polymer behaves as a porogen and is removed through dialysis. For this study, interpenetrating polymer network (IPN) hydrogels were formed between polyvinyl alcohol (PVA) and gelatin using theta-gel fabrication techniques, i.e., in the presence of a porogen. The addition of gelatin to a PVA theta-gel, formed with a porogen, polyethylene glycol (PEG), created macro-porous hydrogels, and increased shear storage moduli and elastic moduli, compared to PVA-gelatin scaffold controls. A reduction in PVA crystallinity was verified by Fourier transform infrared (FTIR) spectroscopy in hydrogels fabricated using a porogen, i.e., PVA-PEG-gelatin, compared to PVA, PVA-PEG, or PVA-gelatin hydrogels alone. Van Geison staining confirmed the retention of gelatin after dialysis. A range of hydrogel moduli was achieved by optimizing PVA concentration, molecular weight, and gelatin concentration. PVA-gelatin hydrogels maintained primary human mesenchymal stem cell (MSC) viability. Soft (∼10 kPa) and stiff (∼100 kPa) PVA-gelatin hydrogels containing type II collagen significantly increased glycosaminoglycan (GAG) production compared to controls. PVA-gelatin hydrogels, formed using theta-gel techniques, warrant further investigation as articular cartilage tissue engineering scaffolds.
Collapse
Affiliation(s)
- Tianxin Miao
- Bioengineering Program, University of Vermont, Burlington, VT 05405, USA.
| | | | | | | |
Collapse
|
42
|
Guo T, Yu L, Lim CG, Goodley AS, Xiao X, Placone JK, Ferlin KM, Nguyen BNB, Hsieh AH, Fisher JP. Effect of Dynamic Culture and Periodic Compression on Human Mesenchymal Stem Cell Proliferation and Chondrogenesis. Ann Biomed Eng 2015; 44:2103-13. [PMID: 26577256 DOI: 10.1007/s10439-015-1510-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/11/2015] [Indexed: 12/27/2022]
Abstract
We have recently developed a bioreactor that can apply both shear and compressive forces to engineered tissues in dynamic culture. In our system, alginate hydrogel beads with encapsulated human mesenchymal stem cells (hMSCs) were cultured under different dynamic conditions while subjected to periodic, compressive force. A customized pressure sensor was developed to track the pressure fluctuations when shear forces and compressive forces were applied. Compared to static culture, dynamic culture can maintain a higher cell population throughout the study. With the application of only shear stress, qRT-PCR and immunohistochemistry revealed that hMSCs experienced less chondrogenic differentiation than the static group. The second study showed that chondrogenic differentiation was enhanced by additional mechanical compression. After 14 days, alcian blue staining showed more extracellular matrix formed in the compression group. The upregulation of the positive chondrogenic markers such as Sox 9, aggrecan, and type II collagen were demonstrated by qPCR. Our bioreactor provides a novel approach to apply mechanical forces to engineered cartilage. Results suggest that a combination of dynamic culture with proper mechanical stimulation may promote efficient progenitor cell expansion in vitro, thereby allowing the culture of clinically relevant articular chondrocytes for the treatment of articular cartilage defects.
Collapse
Affiliation(s)
- Ting Guo
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Li Yu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Casey G Lim
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Addison S Goodley
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Xuan Xiao
- Department of Ophthalmology, Renming Hospital of Wuhan University, Wuhan, China
| | - Jesse K Placone
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Kimberly M Ferlin
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Bao-Ngoc B Nguyen
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - Adam H Hsieh
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA
| | - John P Fisher
- Fischell Department of Bioengineering, University of Maryland, 3238 Jeong H. Kim Engineering Building, College Park, MD, 20742, USA.
| |
Collapse
|
43
|
Sheykhhasan M, Qomi RT, Ghiasi M. Fibrin Scaffolds Designing in order to Human Adipose-derived Mesenchymal Stem Cells Differentiation to Chondrocytes in the Presence of TGF-β3. Int J Stem Cells 2015; 8:219-227. [PMID: 26634070 PMCID: PMC4651286 DOI: 10.15283/ijsc.2015.8.2.219] [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] [Accepted: 05/09/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND OBJECTIVES One of the most cellular source used for cartilage tissue engineering are mesenchymal stem cells (MSCs). In present study, human MSCs were used as cellular source. Since scaffold plays an important role in tissue engineering the aim of this study is to assess fibrin scaffold ability in chondrogenic differentiation of adipose-derived mesenchymal stem cells (ADMSCs). METHODS ADMSCs were isolated and cultured in DMEM medium supplemented with 10% FBS. Also ADMSCs expanded and characterised by flow cytometry. ADMSCs expressed CD44, CD90, CD105 but not CD34. After trypsinization, cells were entered within the fibrin scaffold. Then, chondrogenic medium was added to the scaffold. Seven days after cell culture, cell viability and proliferation were assessed by MTT test. Finally, 14 days after the ending of chondrogenic differentiation, analysis of chondrogenic genes expression was evaluated by RT-PCR and Real time PCR. Also, formation and development of chondrocyte cells was analysed by histological and immunohistochemistry evaluations. RESULTS Viability and proliferation as well as chondrogenic genes expression within fibrin scaffold increased significantly compared with control group (cells free scaffold). Also, histological and immunohistochemistry evaluation showed that chondrocyte cells and collagen type II are formed on fibrin scaffold. CONCLUSIONS Fibrin is a suitable scaffold for chondrogenic differentiation of ADMSCs.
Collapse
Affiliation(s)
- Mohsen Sheykhhasan
- Stem Cell laboratory, The Academic Center for Education, Culture and Research, Qom Branch, Qom,
Iran
| | - Reza Tabatabaei Qomi
- Stem Cell laboratory, The Academic Center for Education, Culture and Research, Qom Branch, Qom,
Iran
| | - Mahdieh Ghiasi
- Stem Cell laboratory, The Academic Center for Education, Culture and Research, Qom Branch, Qom,
Iran
| |
Collapse
|
44
|
Ishak MFB, See GB, Hui CK, Abdullah AB, Saim LB, Saim AB, Idrus RBH. The formation of human auricular cartilage from microtic tissue: An in vivo study. Int J Pediatr Otorhinolaryngol 2015; 79:1634-9. [PMID: 26250439 DOI: 10.1016/j.ijporl.2015.06.034] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 06/23/2015] [Accepted: 06/24/2015] [Indexed: 01/28/2023]
Abstract
OBJECTIVES This study aimed to isolate, culture-expand and characterize the chondrocytes isolated from microtic cartilage and evaluate its potential as a cell source for ear cartilage reconstruction. Specific attention was to construct the auricular cartilage tissue by using fibrin as scaffold. STUDY DESIGN Cell culture experiment with the use of microtic chondrocytes. DESIGN Cell culture experiment with the use of microtic chondrocytes. METHODS After ear reconstructive surgery at the Universiti Kebangsaan Malaysia Medical Center, chondrocytes were isolated from microtic cartilage. Chondrocytes isolated from the tissue were cultured expanded until passage 4 (P4). Upon confluency at P4, chondrocytes were harvested and tissue engineered constructs were made with human plasma polymerized to fibrin. Constructs formed later is implanted at the dorsal part of nude mice for 8 weeks, followed by post-implantation evaluation with histology staining (Hematoxylin and Eosin (H&E) and Safranin O), immunohistochemistry and RT-PCR for chondrogenic associated genes expression level. RESULTS Under gross assessment, the construct after 8 weeks of implantation showed similar physical characteristics that of cartilage. Histological staining showed abundant lacunae cells embedded in extracellular matrix similar to that of native cartilage. Safranin O staining showed positive staining which indicates the presence of proteoglycan-rich matrix. Immunohistochemistry analysis showed the strong positive staining for collagen type II, the specific collagen type in the cartilage. Gene expression quantification showed no significant differences in the expression of chondrogenic gene used which is collagen type I, collagen type II, aggrecan core protein (ACP), elastin and sox9 genes when compared to construct formed from normal auricular tissue. CONCLUSION Chondrocytes isolated from microtia cartilage has the potential to be used as an alternative cell source for external ear reconstruction in future clinical application.
Collapse
Affiliation(s)
- Mohamad Fikeri bin Ishak
- Department of Otorhinolaryngology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia; Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Goh Bee See
- Department of Otorhinolaryngology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia.
| | - Chua Kien Hui
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia; Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Asma bt Abdullah
- Department of Otorhinolaryngology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Lokman bin Saim
- Department of Otorhinolaryngology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Aminuddin bin Saim
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia; Ear, Nose and Throat Consultant Clinic, Ampang Puteri Specialist Hospital, Kuala Lumpur, Malaysia
| | - Ruszymah bt Haji Idrus
- Tissue Engineering Centre, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia; Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
45
|
Hosseini MS, Tafazzoli-Shadpour M, Haghighipour N, Aghdami N, Goodarzi A. The synergistic effects of shear stress and cyclic hydrostatic pressure modulate chondrogenic induction of human mesenchymal stem cells. Int J Artif Organs 2015; 38:557-564. [PMID: 26541277 DOI: 10.5301/ijao.5000433] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/03/2015] [Indexed: 01/01/2023]
Abstract
PURPOSE In this study, we examined chondrogenic regulation of 2 types of mesenchymal stem cells seeded on the bioengineered substrate in monolayer cultures under mechanically defined conditions to mimic the in vivo microenvironment of chondrocytes within articular cartilage tissues. METHODS Human adipose-derived mesenchymal stem cells (ASCs) and bone marrow mesenchymal stem cells (BSCs) were exposed to 0.2 Pa shear stress, 3 MPa cyclic hydrostatic pressure, and combined loading with different sequences on chemically designed medical-grade silicone rubber, while no soluble growth factors were added to the culture medium. The expression levels of chondrogenic-specific genes of SOX9, aggrecan, and type II collagen (Col II) were measured. Results were compared to those of cells treated by biological growth factor. RESULTS Gene expression patterns were dependent on the loading regime. Moreover, the source of mesenchymal stem cells (adipose or bone marrow) was influential in gene expression. Overall, enhanced expression of chondrogenic markers was found through application of mechanical stimuli. The response was generally found to be significantly promoted when the 2 loading regimes were superimposed. CONCLUSIONS Differentiation of ASCs was shown by a modest increase in gene expression profiles. In general, BSCs expressed higher levels of chondrogenic gene expression than ASCs after 3 weeks. A greater effect on Col II and SOX9 mRNA expression was observed when combined loadings were applied. Results may be applied in determining the proper loading sequence for obtaining functional target cells in cartilage engineering applications.
Collapse
Affiliation(s)
| | | | | | - Naser Aghdami
- Regenerative Medicine Department, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran - Iran
| | - Alireza Goodarzi
- Regenerative Medicine Department, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran - Iran
| |
Collapse
|
46
|
Baumgartner W, Welti M, Hild N, Hess SC, Stark WJ, Bürgisser GM, Giovanoli P, Buschmann J. Tissue mechanics of piled critical size biomimetic and biominerizable nanocomposites: Formation of bioreactor-induced stem cell gradients under perfusion and compression. J Mech Behav Biomed Mater 2015; 47:124-134. [DOI: 10.1016/j.jmbbm.2015.03.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 03/18/2015] [Accepted: 03/24/2015] [Indexed: 02/08/2023]
|
47
|
Zhang C, Lu Y, Zhang L, Liu Y, Zhou Y, Chen Y, Yu H. Influence of different intensities of vibration on proliferation and differentiation of human periodontal ligament stem cells. Arch Med Sci 2015; 11:638-46. [PMID: 26170859 PMCID: PMC4495159 DOI: 10.5114/aoms.2015.52370] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/02/2014] [Accepted: 03/06/2014] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION To understand the effects of low-magnitude, high-frequency (LMHF) mechanical vibration at different intensities on human periodontal ligament stem cell (hPDLSC) proliferation and osteogenic differentiation. MATERIAL AND METHODS The effect of vibration on hPDLSC proliferation, osteogenic differentiation, tenogenic differentiation and cytoskeleton was assessed at the cellular, genetic and protein level. RESULTS The PDLSC proliferation was decreased after different magnitudes of mechanical vibration; however, there were no obvious senescent cells in the experimental and the static control group. Expression of osteogenesis markers was increased. The expression of alkaline phosphatase (ALP) and osteocalcin (OCN) mRNA was up-regulated at 0.1 g, 0.3 g, 0.6 g and 0.9 g magnitude, with the peak at 0.3 g. The type I collagen (Col-I) level was increased after vibration exposure at 0.1 g, 0.3 g, and 0.6 g, peaking at 0.3 g. The expression levels of both mRNA and protein of Runx2 and osterix (OSX) significantly increased at a magnitude of 0.1 g to 0.9 g, reached a peak at 0.3 g and then decreased slowly. The scleraxis, tenogenic markers, and mRNA expression decreased at 0.05 g, 0.1 g, and 0.3 g, and significantly increased at 0.6 g and 0.9 g. Compared with the static group, the F-actin stress fibers of hPDLSCs became thicker and clearer following vibration. CONCLUSIONS The LMHF mechanical vibration promotes PDLSC osteogenic differentiation and implies the existence of a magnitude-dependent effect of vibration on determining PDLSC commitment to the osteoblast lineage. Changes in the cytoskeleton of hPDLSCs after vibration may be one of the mechanisms of the biological effects.
Collapse
Affiliation(s)
- Chunxiang Zhang
- Tianjin Stomatological Hospital of Nankai University, Tianjin, China
| | - Yanqin Lu
- Xiangya Stomatology Hospital, Central South University, Changsha, Hunan, China
| | - Linkun Zhang
- Tianjin Stomatological Hospital of Nankai University, Tianjin, China
| | - Yang Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yi Zhou
- Affiliated Hospital of Stomatology, Medical College, Zhejiang University, Hangzhou, China
| | - Yangxi Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Haiyang Yu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
|
49
|
Abstract
Mechanical forces, including hydrodynamic shear, hydrostatic pressure, compression, tension, and friction, can have stimulatory effects on cartilage synthesis in tissue engineering systems. Bioreactors capable of exerting forces on cells and tissue constructs within a controlled culture environment are needed to provide appropriate mechanical stimuli. In this chapter, we describe the construction, assembly, and operation of a mechanobioreactor providing simultaneous dynamic shear and compressive loading on developing cartilage tissues to mimic the rolling and squeezing action of articular joints. The device is suitable for studying the effects of mechanical treatment on stem cells and chondrocytes seeded into three-dimensional scaffolds.
Collapse
|
50
|
Abstract
Many technologies that underpin tissue engineering as a research field were developed with the aim of producing functional human cartilage in vitro. Much of our practical experience with three-dimensional cultures, tissue bioreactors, scaffold materials, stem cells, and differentiation protocols was gained using cartilage as a model system. Despite these advances, however, generation of engineered cartilage matrix with the composition, structure, and mechanical properties of mature articular cartilage has not yet been achieved. Currently, the major obstacles to synthesis of clinically useful cartilage constructs are our inability to control differentiation to the extent needed, and the failure of engineered and host tissues to integrate after construct implantation. The aim of this chapter is to distil from the large available body of literature the seminal approaches and experimental techniques developed for cartilage tissue engineering and to identify those specific areas requiring further research effort.
Collapse
Affiliation(s)
- Pauline M Doran
- Faculty of Science, Engineering and Technology, Swinburne University of Technology, 218, Hawthorn, Melbourne, VIC, 3122, Australia.
| |
Collapse
|