1
|
Hutcherson CW, Mao M, Thakur B, Dhaher YY. Low-Grade Inflammatory Mediators and Metalloproteinases Yield Synchronous and Delayed Responses to Mechanical Joint Loading. Cartilage 2024; 15:417-427. [PMID: 37614184 PMCID: PMC11526223 DOI: 10.1177/19476035231193089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 06/15/2023] [Accepted: 07/16/2023] [Indexed: 08/25/2023] Open
Abstract
OBJECTIVE Mechanical loading is an essential factor for the maintenance of joint inflammatory homeostasis and the sensitive catabolic-anabolic signaling cascade involved in maintaining cartilage tissue health. However, abnormal mechanical loading of the joint structural tissues can propagate joint metabolic dysfunction in the form of low-grade inflammation. To date, few studies have attempted to delineate the early cascade responsible for the initiation and perpetuation of stress-mediated inflammation and cartilage breakdown in human joints. DESIGN Fifteen healthy human male participants performed a walking paradigm on a cross-tilting treadmill platform. Blood samples were collected before exercise, after 30 minutes of flat walking, after 30 minutes of tilted walking, and after an hour of rest. Serum concentrations of the following biomarkers were measured: interleukin (IL)-1β, IL-6, IL-10, tumor necrosis factor alpha (TNF)-α, matrix metalloproteinase (MMP)-1, MMP-3, MMP-9, MMP-13, transforming growth factor beta (TGF)-β, tissue inhibitor of matrix metalloproteinase 1 (TIMP)-1, and cartilage oligomeric protein (COMP). RESULTS Luminex Multiplex analysis of serum showed increased concentrations of COMP, IL-1β, TNF-α, IL-10, and TGF-β from samples collected after flat and cross-tilted treadmill walking compared to baseline. Serum concentrations of MMP-1 and MMP-13 also increased, but primarily in samples collected after tilted walking. Pearson's correlation analysis showed positive correlations between the expression of COMP, TNF-α, IL-10, and MMP-13 at each study timepoint. CONCLUSION Stress-mediated increases in serum COMP during exercise are associated with acute changes in pro and anti-inflammatory molecular activity and subsequent changes in molecules linked to joint tissue remodeling and repair.
Collapse
Affiliation(s)
- Conner W. Hutcherson
- Department of Orthopaedic Surgery, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michelle Mao
- Department of Orthopaedic Surgery, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bhaskar Thakur
- Department of Physical Medicine and Rehabilitation, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Population and Data sciences, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yasin Y. Dhaher
- Department of Orthopaedic Surgery, The University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Physical Medicine and Rehabilitation, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
2
|
Wang X, Tao J, Zhou J, Shu Y, Xu J. Excessive load promotes temporomandibular joint chondrocyte apoptosis via Piezo1/endoplasmic reticulum stress pathway. J Cell Mol Med 2024; 28:e18472. [PMID: 38842129 PMCID: PMC11154833 DOI: 10.1111/jcmm.18472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024] Open
Abstract
Excessive load on the temporomandibular joint (TMJ) is a significant factor in the development of TMJ osteoarthritis, contributing to cartilage degeneration. The specific mechanism through which excessive load induces TMJ osteoarthritis is not fully understood; however, mechanically-activated (MA) ion channels play a crucial role. Among these channels, Piezo1 has been identified as a mediator of chondrocyte catabolic responses and is markedly increased in osteoarthritis. Our observations indicate that, under excessive load conditions, endoplasmic reticulum stress in chondrocytes results in apoptosis of the TMJ chondrocytes. Importantly, using the Piezo1 inhibitor GsMTx4 demonstrates its potential to alleviate this condition. Furthermore, Piezo1 mediates endoplasmic reticulum stress in chondrocytes by inducing calcium ion influx. Our research substantiates the role of Piezo1 as a pivotal ion channel in mediating chondrocyte overload. It elucidates the link between excessive load, cell apoptosis, and calcium ion influx through Piezo1. The findings underscore Piezo1 as a key player in the pathogenesis of TMJ osteoarthritis, shedding light on potential therapeutic interventions for this condition.
Collapse
Affiliation(s)
- Xiaohui Wang
- College of StomatologyChongqing Medical UniversityChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
- Chongqing Key Laboratory for Oral Diseases and Biomedical SciencesChongqingChina
| | - Junli Tao
- College of StomatologyChongqing Medical UniversityChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
- Chongqing Key Laboratory for Oral Diseases and Biomedical SciencesChongqingChina
| | - Jianping Zhou
- College of StomatologyChongqing Medical UniversityChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
- Chongqing Key Laboratory for Oral Diseases and Biomedical SciencesChongqingChina
| | - Yi Shu
- College of StomatologyChongqing Medical UniversityChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
- Chongqing Key Laboratory for Oral Diseases and Biomedical SciencesChongqingChina
| | - Jie Xu
- College of StomatologyChongqing Medical UniversityChongqingChina
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher EducationChongqingChina
- Chongqing Key Laboratory for Oral Diseases and Biomedical SciencesChongqingChina
- State Key Laboratory of Ultrasound in Medicine and EngineeringChongqing Medical UniversityChongqingChina
| |
Collapse
|
3
|
Du C, Liu J, Liu S, Xiao P, Chen Z, Chen H, Huang W, Lei Y. Bone and Joint-on-Chip Platforms: Construction Strategies and Applications. SMALL METHODS 2024:e2400436. [PMID: 38763918 DOI: 10.1002/smtd.202400436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/28/2024] [Indexed: 05/21/2024]
Abstract
Organ-on-a-chip, also known as "tissue chip," is an advanced platform based on microfluidic systems for constructing miniature organ models in vitro. They can replicate the complex physiological and pathological responses of human organs. In recent years, the development of bone and joint-on-chip platforms aims to simulate the complex physiological and pathological processes occurring in human bones and joints, including cell-cell interactions, the interplay of various biochemical factors, the effects of mechanical stimuli, and the intricate connections between multiple organs. In the future, bone and joint-on-chip platforms will integrate the advantages of multiple disciplines, bringing more possibilities for exploring disease mechanisms, drug screening, and personalized medicine. This review explores the construction and application of Organ-on-a-chip technology in bone and joint disease research, proposes a modular construction concept, and discusses the new opportunities and future challenges in the construction and application of bone and joint-on-chip platforms.
Collapse
Affiliation(s)
- Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Senrui Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Xiao
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhuolin Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
4
|
Pan C, Zhang Y, Yan J, Zhou Y, Wang S, Liu X, Zhang P, Yang H. Extreme environments and human health: From the immune microenvironments to immune cells. ENVIRONMENTAL RESEARCH 2023; 236:116800. [PMID: 37527745 DOI: 10.1016/j.envres.2023.116800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/20/2023] [Accepted: 07/29/2023] [Indexed: 08/03/2023]
Abstract
Exposure to extreme environments causes specific acute and chronic physiological responses in humans. The adaptation and the physiological processes under extreme environments predominantly affect multiple functional systems of the organism, in particular, the immune system. Dysfunction of the immune system affected by several extreme environments (including hyperbaric environment, hypoxia, blast shock, microgravity, hypergravity, radiation exposure, and magnetic environment) has been observed from clinical macroscopic symptoms to intracorporal immune microenvironments. Therefore, simulated extreme conditions are engineered for verifying the main influenced characteristics and factors in the immune microenvironments. This review summarizes the responses of immune microenvironments to these extreme environments during in vivo or in vitro exposure, and the approaches of engineering simulated extreme environments in recent decades. The related microenvironment engineering, signaling pathways, molecular mechanisms, clinical therapy, and prevention strategies are also discussed.
Collapse
Affiliation(s)
- Chengwei Pan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Yuzhi Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Jinxiao Yan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Yidan Zhou
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Sijie Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Xiru Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Pan Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; School of Food Science and Engineering, Shaanxi University of Science & Technology, 710021, China.
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
5
|
Chen Y, Luo M, Xie Y, Xing L, Han X, Tian Y. Periodontal ligament-associated protein-1 engages in teeth overeruption and periodontal fiber disorder following occlusal hypofunction. J Periodontal Res 2023; 58:131-142. [PMID: 36445954 DOI: 10.1111/jre.13075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/28/2022] [Accepted: 11/08/2022] [Indexed: 12/03/2022]
Abstract
BACKGROUND AND OBJECTIVE Teeth overeruption is a problem of clinical significance, but the underlying mechanism how changes in external occlusal force convert to the periodontium remodeling signals has been a largely under explored domain. And recently, periodontal ligament-associated protein-1 (PLAP-1)/asporin was found to play a pivotal role in maintaining periodontal homeostasis. The aim of this study was to explore the function of PLAP-1 in the periodontally hypofunctional tissue turnover. METHODS After extracting left maxillary molars in mice, the left and right mandibular molars were distributed into hypofunction group (HG) and control group (CG), respectively. Mice were sacrificed for radiographic, histological, and molecular biological analyses after 1, 4 and 12 weeks. In vitro, dynamic compression was applied using Flexcell FX-5000 Compression System to simulate intermittent occlusal force. The expression of PLAP1 in loaded and unloaded human periodontal ligament cells (hPDLCs) was compared, and its molecular biological effects were further explored by small interfering RNA (siRNA) targeting PLAP1. RESULTS In vivo, fiber disorder in periodontal ligament (PDL), bone apposition at furcation regions, and bone resorption in alveolar bone were illustrated in the HG compared with the CG. In addition, PLAP-1 positive area decreased significantly in PDL following occlusal unloading. In vitro, the loss of compressive loading relatively downregulated PLAP1 expression, which was essential to promote collagen I but inhibit osterix and osteocalcin expression in hPDLCs. CONCLUSIONS PLAP-1 presumably plays a pivotal role in occlusal force-regulated periodontal homeostasis by facilitating collagen fiber synthesis in hPDLCs and suppressing excessive osteoblast differentiation, further preventing teeth from overeruption. Further evidence in PLAP-1 conditional knockout mice is needed.
Collapse
Affiliation(s)
- Yilin Chen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mengqi Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yaxin Xie
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lu Xing
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xianglong Han
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ye Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Nix Z, Kota D, Ratnayake I, Wang C, Smith S, Wood S. Spectral characterization of cell surface motion for mechanistic investigations of cellular mechanobiology. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 176:3-15. [PMID: 36108781 DOI: 10.1016/j.pbiomolbio.2022.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 07/27/2022] [Accepted: 08/05/2022] [Indexed: 06/15/2023]
Abstract
Understanding the specific mechanisms responsible for anabolic and catabolic responses to static or dynamic force are largely poorly understood. Because of this, most research groups studying mechanotransduction due to dynamic forces employ an empirical approach in deciding what frequencies to apply during experiments. While this has been shown to elucidate valuable information regarding how cells respond under controlled provocation, it is often difficult or impossible to determine a true optimal frequency for force application, as many intracellular complexes are involved in receiving, propagating, and responding to a given stimulus. Here we present a novel adaptation of an analytical technique from the fields of civil and mechanical engineering that may open the door to direct measurement of mechanobiological cellular frequencies which could be used to target specific cell signaling pathways leveraging synergy between outside-in and inside-out mechanotransduction approaches. This information could be useful in identifying how specific proteins are involved in the homeostatic balance, or disruption thereof, of cells and tissue, furthering the understanding of the pathogenesis and progression of many diseases across a wide variety of cell types, which may one day lead to the development of novel mechanobiological therapies for clinical use.
Collapse
Affiliation(s)
- Zachary Nix
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA
| | - Divya Kota
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA
| | - Ishara Ratnayake
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA
| | - Congzhou Wang
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA
| | - Steve Smith
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA
| | - Scott Wood
- Department of Nanoscience & Biomedical Engineering, BioSystems Networks / Translational Research (BioSNTR), South Dakota School of Mines and Technology, USA.
| |
Collapse
|
7
|
Paggi CA, Hendriks J, Karperien M, Le Gac S. Emulating the chondrocyte microenvironment using multi-directional mechanical stimulation in a cartilage-on-chip. LAB ON A CHIP 2022; 22:1815-1828. [PMID: 35352723 DOI: 10.1039/d1lc01069g] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The multi-directional mechanical stimulation experienced by articular cartilage during motion is transferred to the chondrocytes through a thin layer of pericellular matrix around each cell; chondrocytes in turn respond by releasing matrix proteins and/or matrix-degrading enzymes. In the present study we investigated how different types of mechanical stimulation can affect a chondrocyte's phenotype and extracellular matrix (ECM) production. To this end, we employed a cartilage-on-chip system which allows exerting well-defined compressive and multi-directional mechanical stimulation on a 3D chondrocyte-laden agarose hydrogel using a thin deformable membrane and three individually addressed actuation chambers. First, the 3D chondrocyte culture in agarose responded to exposure to mechanical stimulation by an initial increase in IL-6 production and little-to-no change in IL-1β and TNF-α secretion after one day of on-chip culture. Exposure to mechanical stimulation enhanced COL2A1 (hyaline cartilage marker) and decreased COL1A1 (fibrotic cartilage) expression, this being more marked for the multi-directional stimulation. Remarkably, the production of glycosaminoglycans (GAGs), one of the main components of native cartilage ECM, was significantly increased after 15 days of on-chip culture and 14 days of mechanical stimulation. Specifically, a thin pericellular matrix shell (1-5 μm) surrounding the chondrocytes as well as an interstitial matrix, both reminiscent of the in vivo situation, were deposited. Matrix deposition was highest in chips exposed to multi-directional mechanical stimulation. Finally, exposure to mechanical cues enhanced the production of essential cartilage ECM markers, such as aggrecan, collagen II and collagen VI, a marker for the pericellular matrix. Altogether our results highlight the importance of mechanical cues, and using the right type of stimulation, to emulate in vitro, the chondrocyte microenvironment.
Collapse
Affiliation(s)
- Carlo Alberto Paggi
- Department of Developmental BioEngineering, TechMed Centre, and Organ-on-chip Centre, University of Twente, Enschede, The Netherlands.
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology & TechMed Centre, and Organ-on-chip Centre, University of Twente, Enschede, The Netherlands.
| | - Jan Hendriks
- Department of Developmental BioEngineering, TechMed Centre, and Organ-on-chip Centre, University of Twente, Enschede, The Netherlands.
| | - Marcel Karperien
- Department of Developmental BioEngineering, TechMed Centre, and Organ-on-chip Centre, University of Twente, Enschede, The Netherlands.
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology & TechMed Centre, and Organ-on-chip Centre, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
8
|
Joint-on-chip platforms: entering a new era of in vitro models for arthritis. Nat Rev Rheumatol 2022; 18:217-231. [DOI: 10.1038/s41584-021-00736-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2021] [Indexed: 12/12/2022]
|
9
|
Hopkins T, Wright KT, Kuiper NJ, Roberts S, Jermin P, Gallacher P, Kuiper JH. An In Vitro System to Study the Effect of Subchondral Bone Health on Articular Cartilage Repair in Humans. Cells 2021; 10:1903. [PMID: 34440671 PMCID: PMC8392168 DOI: 10.3390/cells10081903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/19/2022] Open
Abstract
Chondrocyte-based cartilage repair strategies, such as articular chondrocyte implantation, are widely used, but few studies addressed the communication between native subchondral bone cells and the transplanted chondrocytes. An indirect co-culture model was developed, representing a chondrocyte/scaffold-construct repair of a cartilage defect adjoining bone, where the bone could have varying degrees of degeneration. Human BM-MSCs were isolated from two areas of subchondral bone in each of five osteochondral tissue specimens from five patients undergoing knee arthroplasty. These two areas underlaid the macroscopically and histologically best and worst cartilage, representing early and late-stage OA, respectively. BM-MSCs were co-cultured with normal chondrocytes suspended in agarose, with the two cell types separated by a porous membrane. After 0, 7, 14 and 21 days, chondrocyte-agarose scaffolds were assessed by gene expression and biochemical analyses, and the abundance of selected proteins in conditioned media was assessed by ELISA. Co-culture with late-OA BM-MSCs resulted in a reduction in GAG deposition and a decreased expression of genes encoding matrix-specific proteins (COL2A1 and ACAN), compared to culturing with early OA BM-MSCs. The concentration of TGF-β1 was significantly higher in the early OA conditioned media. The results of this study have clinical implications for cartilage repair, suggesting that the health of the subchondral bone may influence the outcomes of chondrocyte-based repair strategies.
Collapse
Affiliation(s)
- Timothy Hopkins
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| | - Karina T. Wright
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| | - Nicola J. Kuiper
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| | - Sally Roberts
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| | - Paul Jermin
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| | - Peter Gallacher
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
| | - Jan Herman Kuiper
- School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK; (K.T.W.); (N.J.K.); (S.R.); (P.J.); (P.G.); (J.H.K.)
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Shropshire SY10 7AG, UK
| |
Collapse
|
10
|
Caravaggi P, Assirelli E, Ensini A, Ortolani M, Mariani E, Leardini A, Neri S, Belvedere C. Biomechanical-Based Protocol for in vitro Study of Cartilage Response to Cyclic Loading: A Proof-of-Concept in Knee Osteoarthritis. Front Bioeng Biotechnol 2021; 9:634327. [PMID: 34012954 PMCID: PMC8126668 DOI: 10.3389/fbioe.2021.634327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/08/2021] [Indexed: 12/28/2022] Open
Abstract
Osteoarthritis (OA) is an evolving disease and a major cause of pain and impaired mobility. A deeper understanding of cartilage metabolism in response to loading is critical to achieve greater insight into OA mechanisms. While physiological joint loading helps maintain cartilage integrity, reduced or excessive loading have catabolic effects. The main scope of this study is to present an original methodology potentially capable to elucidate the effect of cyclic joint loading on cartilage metabolism, to identify mechanisms involved in preventing or slowing down OA progression, and to provide preliminary data on its application. In the proposed protocol, the combination of biomechanical data and medical imaging are integrated with molecular information about chondrocyte mechanotransduction and tissue homeostasis. The protocol appears to be flexible and suitable to analyze human OA knee cartilage explants, with different degrees of degeneration, undergoing ex vivo realistic cyclic joint loading estimated via gait analysis in patients simulating mild activities of daily living. The modulation of molecules involved in cartilage homeostasis, mechanotransduction, inflammation, pain and wound healing can be analyzed in chondrocytes and culture supernatants. A thorough analysis performed with the proposed methodology, combining in vivo functional biomechanical evaluations with ex vivo molecular assessments is expected to provide new insights on the beneficial effects of physiological loading and contribute to the design and optimization of non-pharmacological treatments limiting OA progression.
Collapse
Affiliation(s)
- Paolo Caravaggi
- Movement Analysis Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Assirelli
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Andrea Ensini
- I Orthopaedic and Traumatologic Clinic, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Maurizio Ortolani
- Movement Analysis Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Erminia Mariani
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy.,Department of Medical and Surgical Sciences, Alma Mater Studiorum-Università di Bologna, Bologna, Italy
| | - Alberto Leardini
- Movement Analysis Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Simona Neri
- Laboratory of Immunorheumatology and Tissue Regeneration, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Claudio Belvedere
- Movement Analysis Laboratory, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
11
|
Kunisaki SM, Jiang G, Biancotti JC, Ho KKY, Dye BR, Liu AP, Spence JR. Human induced pluripotent stem cell-derived lung organoids in an ex vivo model of the congenital diaphragmatic hernia fetal lung. Stem Cells Transl Med 2020; 10:98-114. [PMID: 32949227 PMCID: PMC7780804 DOI: 10.1002/sctm.20-0199] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/03/2020] [Accepted: 08/09/2020] [Indexed: 01/06/2023] Open
Abstract
Three‐dimensional lung organoids (LOs) derived from pluripotent stem cells have the potential to enhance our understanding of disease mechanisms and to enable novel therapeutic approaches in neonates with pulmonary disorders. We established a reproducible ex vivo model of lung development using transgene‐free human induced pluripotent stem cells generated from fetuses and infants with Bochdalek congenital diaphragmatic hernia (CDH), a polygenic disorder associated with fetal lung compression and pulmonary hypoplasia at birth. Molecular and cellular comparisons of CDH LOs revealed impaired generation of NKX2.1+ progenitors, type II alveolar epithelial cells, and PDGFRα+ myofibroblasts. We then subjected these LOs to disease relevant mechanical cues through ex vivo compression and observed significant changes in genes associated with pulmonary progenitors, alveolar epithelial cells, and mesenchymal fibroblasts. Collectively, these data suggest both primary cell‐intrinsic and secondary mechanical causes of CDH lung hypoplasia and support the use of this stem cell‐based approach for disease modeling in CDH.
Collapse
Affiliation(s)
- Shaun M Kunisaki
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Cell Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Guihua Jiang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Juan C Biancotti
- Department of Surgery, Johns Hopkins University, Baltimore, Maryland, USA.,Institute for Cell Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kenneth K Y Ho
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Briana R Dye
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason R Spence
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA.,Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
12
|
Lee D, Erickson A, Dudley AT, Ryu S. Mechanical stimulation of growth plate chondrocytes: Previous approaches and future directions. EXPERIMENTAL MECHANICS 2019; 59:1261-1274. [PMID: 31787777 PMCID: PMC6884322 DOI: 10.1007/s11340-018-0424-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Growth plate cartilage resides near the ends of long bones and is the primary driver of skeletal growth. During growth, both intrinsically and extrinsically generated mechanical stresses act on chondrocytes in the growth plate. Although the role of mechanical stresses in promoting tissue growth and homeostasis has been strongly demonstrated in articular cartilage of the major skeletal joints, effects of stresses on growth plate cartilage and bone growth are not as well established. Here, we review the literature on mechanobiology in growth plate cartilage at macroscopic and microscopic scales, with particular emphasis on comparison of results obtained using different methodological approaches, as well as from whole animal and in vitro experiments. To answer these questions, macroscopic mechanical stimulators have been developed and applied to study mechanobiology of growth plate cartilage and chondrocytes. However, the previous approaches have tested a limited number of stress conditions, and the mechanobiology of a single chondrocyte has not been well studied due to limitations of the macroscopic mechanical stimulators. We explore how microfluidics devices can overcome these limitations and improve current understanding of growth plate chondrocyte mechanobiology. In particular, microfluidic devices can generate multiple stress conditions in a single platform and enable real-time monitoring of metabolism and cellular behavior using optical microscopy. Systematic characterization of the chondrocytes using microfluidics will enhance our understanding of how to use mechanical stresses to control the bone growth and the properties of tissue-engineered growth plate cartilage.
Collapse
Affiliation(s)
- D. Lee
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
| | - A. Erickson
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
| | - A. T. Dudley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
- Corresponding Authors:; Tel: +1-402-559-2820. ; Tel: +1-402-472-4313
| | - S. Ryu
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588
- Nebraska Center for Materials and Nanoscience, University of Nebraska-Lincoln, Lincoln, NE 68588
- Corresponding Authors:; Tel: +1-402-559-2820. ; Tel: +1-402-472-4313
| |
Collapse
|
13
|
Lee D, Erickson A, Dudley AT, Ryu S. A Microfluidic Platform for Stimulating Chondrocytes with Dynamic Compression. J Vis Exp 2019. [PMID: 31566611 DOI: 10.3791/59676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Mechanical stimuli are known to modulate biological functions of cells and tissues. Recent studies have suggested that compressive stress alters growth plate cartilage architecture and results in growth modulation of long bones of children. To determine the role of compressive stress in bone growth, we created a microfluidic device actuated by pneumatic pressure, to dynamically (or statically) compress growth plate chondrocytes embedded in alginate hydrogel cylinders. In this article, we describe detailed methods for fabricating and characterizing this device. The advantages of our protocol are: 1) Five different magnitudes of compressive stress can be generated on five technical replicates in a single platform, 2) It is easy to visualize cell morphology via a conventional light microscope, 3) Cells can be rapidly isolated from the device after compression to facilitate downstream assays, and 4) The platform can be applied to study mechanobiology of any cell type that can grow in hydrogels.
Collapse
Affiliation(s)
- Donghee Lee
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center
| | - Alek Erickson
- Department of Physiology and Pharmacology, Karolinska Institutet
| | - Andrew T Dudley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center;
| | - Sangjin Ryu
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln; Nebraska Center for Materials and Nanoscience, University of Nebraska-Lincoln;
| |
Collapse
|
14
|
Kim BG, Sung JS, Jang Y, Cha YJ, Kang S, Han HH, Lee JH, Cho NH. Compression-induced expression of glycolysis genes in CAFs correlates with EMT and angiogenesis gene expression in breast cancer. Commun Biol 2019; 2:313. [PMID: 31428701 PMCID: PMC6694123 DOI: 10.1038/s42003-019-0553-9] [Citation(s) in RCA: 30] [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: 01/03/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
Tumor growth increases compressive stress within a tissue, which is associated with solid tumor progression. However, very little is known about how compressive stress contributes to tumor progression. Here, we show that compressive stress induces glycolysis in human breast cancer associated fibroblast (CAF) cells and thereby contributes to the expression of epithelial to mesenchymal (EMT)- and angiogenesis-related genes in breast cancer cells. Lactate production was increased in compressed CAF cells, in a manner dependent on the expression of metabolic genes ENO2, HK2, and PFKFB3. Conditioned medium from compressed CAFs promoted the proliferation of breast cancer cells and the expression of EMT and/or angiogenesis-related genes. In patient tissues with high compressive stress, the expression of compression-induced metabolic genes was significantly and positively correlated with EMT and/or angiogenesis-related gene expression and metastasis size. These findings illustrate a mechanotransduction pathway involving stromal glycolysis that may be relevant also for other solid tumours.
Collapse
Affiliation(s)
- Baek Gil Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Sol Sung
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Yeonsue Jang
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yoon Jin Cha
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Suki Kang
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
- Severance Biomedical Science Institute (SBSI), Yonsei University College of Medicine, Seoul, South Korea
| | - Hyun Ho Han
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Joo Hyun Lee
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Nam Hoon Cho
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
- Brain Korea 21 Plus Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
- Severance Biomedical Science Institute (SBSI), Yonsei University College of Medicine, Seoul, South Korea
- Global 5-5-10 System Biology, Yonsei University, Seoul, South Korea
| |
Collapse
|
15
|
Frank D, Cser A, Kolarovszki B, Farkas N, Miseta A, Nagy T. Mechanical stress alters protein O-GlcNAc in human periodontal ligament cells. J Cell Mol Med 2019; 23:6251-6259. [PMID: 31237748 PMCID: PMC6714205 DOI: 10.1111/jcmm.14509] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/10/2019] [Accepted: 06/04/2019] [Indexed: 12/27/2022] Open
Abstract
Protein O-linked N-acetylglucosamine (O-GlcNAc) is a post-translational modification of intracellular proteins that regulates several physiological and pathophysiological process, including response to various stressors. However, O-GlcNAc's response to mechanical stress has not been investigated yet. As human periodontal ligament (PDL) cells are stimulated by compression force during orthodontic tooth movement that results in structural remodelling, in this study we investigated whether mechanical stress induces any alteration in protein O-GlcNAc in PDL cells. In this study, PDL cells isolated from premolars extracted for orthodontic indications were exposed to 0, 1.5, 3, 7 and 14 g/cm2 compression forces for 12 hours. Cell viability was measured by flow cytometry, and protein O-GlcNAc was analysed by Western blot. Cellular structure and intracellular distribution of O-GlcNAc was studied by immunofluorescence microscopy. We found that between 1.5 and 3 g/cm2 mechanical compression, O-GlcNAc significantly elevated; however, at higher forces O-GlcNAc level was not increased. We also found that intracellular localization of O-GlcNAc proteins became more centralized under 2 g/cm2 compression force. Our results suggest that structural changes stimulated by compression forces have a significant effect on the regulation of O-GlcNAc; thus, it might play a role in the mechanical stress adaptation of PDL cells.
Collapse
Affiliation(s)
- Dorottya Frank
- Department of Dentistry, Oral and Maxillofacial Surgery, Medical School, University of Pécs, Pécs, Hungary
| | - Annamária Cser
- Department of Dentistry, Oral and Maxillofacial Surgery, Medical School, University of Pécs, Pécs, Hungary
| | - Béla Kolarovszki
- Department of Dentistry, Oral and Maxillofacial Surgery, Medical School, University of Pécs, Pécs, Hungary
| | - Nelli Farkas
- Institute of Bioanalysis, Medical School, University of Pécs, Pécs, Hungary
| | - Attila Miseta
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| | - Tamás Nagy
- Department of Laboratory Medicine, Medical School, University of Pécs, Pécs, Hungary
| |
Collapse
|
16
|
Perrier-Groult E, Pérès E, Pasdeloup M, Gazzolo L, Duc Dodon M, Mallein-Gerin F. Evaluation of the biocompatibility and stability of allogeneic tissue-engineered cartilage in humanized mice. PLoS One 2019; 14:e0217183. [PMID: 31107916 PMCID: PMC6527235 DOI: 10.1371/journal.pone.0217183] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/07/2019] [Indexed: 11/18/2022] Open
Abstract
Articular cartilage (AC) has poor capacities of regeneration and lesions often lead to osteoarthritis. Current AC reconstruction implies autologous chondrocyte implantation which requires tissue sampling and grafting. An alternative approach would be to use scaffolds containing off-the-shelf allogeneic human articular chondrocytes (HACs). To investigate tolerance of allogeneic HACs by the human immune system, we developed a humanized mouse model implanted with allogeneic cartilage constructs generated in vitro. A prerequisite of the study was to identify a scaffold that would not provoke inflammatory reaction in host. Therefore, we first compared the response of hu-mice to two biomaterials used in regenerative medicine, collagen sponge and agarose hydrogel. Four weeks after implantation in hu-mice, acellular collagen sponges, but not acellular agarose hydrogels, showed positive staining for CD3 (T lymphocytes) and CD68 (macrophages), suggesting that collagen scaffold elicits weak inflammatory reaction. These data led us to deepen our evaluation of the biocompatibility of allogeneic tissue-engineered cartilage by using agarose as scaffold. Agarose hydrogels were combined with allogeneic HACs to reconstruct cartilage in vitro. Particular attention was paid to HLA-A2 compatibility between HACs to be grafted and immune human cells of hu-mice: HLA-A2+ or HLA-A2- HACs agarose hydrogels were cultured in the presence of a chondrogenic cocktail and implanted in HLA-A2+ hu-mice. After four weeks implantation and regardless of the HLA-A2 phenotype, chondrocytes were well-differentiated and produced cartilage matrix in agarose. In addition, no sign of T-cell or macrophage infiltration was seen in the cartilaginous constructs and no significant increase in subpopulations of T lymphocytes and monocytes was detected in peripheral blood and spleen. We show for the first time that humanized mouse represents a useful model to investigate human immune responsiveness to tissue-engineered cartilage and our data together indicate that allogeneic cartilage constructs can be suitable for cartilage engineering.
Collapse
Affiliation(s)
- Emeline Perrier-Groult
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS-UMR5305, Lyon, France
- * E-mail:
| | - Eléonore Pérès
- Laboratory of Biology and Modeling of the Cell, Ecole Normale Supérieure (ENS) de Lyon, INSERM U1210, CNRS UMR5239, Lyon, France
| | - Marielle Pasdeloup
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS-UMR5305, Lyon, France
| | - Louis Gazzolo
- Laboratory of Biology and Modeling of the Cell, Ecole Normale Supérieure (ENS) de Lyon, INSERM U1210, CNRS UMR5239, Lyon, France
| | - Madeleine Duc Dodon
- Laboratory of Biology and Modeling of the Cell, Ecole Normale Supérieure (ENS) de Lyon, INSERM U1210, CNRS UMR5239, Lyon, France
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS-UMR5305, Lyon, France
| |
Collapse
|
17
|
Bregenzer ME, Horst EN, Mehta P, Novak CM, Raghavan S, Snyder CS, Mehta G. Integrated cancer tissue engineering models for precision medicine. PLoS One 2019; 14:e0216564. [PMID: 31075118 PMCID: PMC6510431 DOI: 10.1371/journal.pone.0216564] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tumors are not merely cancerous cells that undergo mindless proliferation. Rather, they are highly organized and interconnected organ systems. Tumor cells reside in complex microenvironments in which they are subjected to a variety of physical and chemical stimuli that influence cell behavior and ultimately the progression and maintenance of the tumor. As cancer bioengineers, it is our responsibility to create physiologic models that enable accurate understanding of the multi-dimensional structure, organization, and complex relationships in diverse tumor microenvironments. Such models can greatly expedite clinical discovery and translation by closely replicating the physiological conditions while maintaining high tunability and control of extrinsic factors. In this review, we discuss the current models that target key aspects of the tumor microenvironment and their role in cancer progression. In order to address sources of experimental variation and model limitations, we also make recommendations for methods to improve overall physiologic reproducibility, experimental repeatability, and rigor within the field. Improvements can be made through an enhanced emphasis on mathematical modeling, standardized in vitro model characterization, transparent reporting of methodologies, and designing experiments with physiological metrics. Taken together these considerations will enhance the relevance of in vitro tumor models, biological understanding, and accelerate treatment exploration ultimately leading to improved clinical outcomes. Moreover, the development of robust, user-friendly models that integrate important stimuli will allow for the in-depth study of tumors as they undergo progression from non-transformed primary cells to metastatic disease and facilitate translation to a wide variety of biological and clinical studies.
Collapse
Affiliation(s)
- Michael E. Bregenzer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Eric N. Horst
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Pooja Mehta
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Caymen M. Novak
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Shreya Raghavan
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Catherine S. Snyder
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Geeta Mehta
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
18
|
Dufour A, Buffier M, Vertu-Ciolino D, Disant F, Mallein-Gerin F, Perrier-Groult E. Combination of bioactive factors and IEIK13 self-assembling peptide hydrogel promotes cartilage matrix production by human nasal chondrocytes. J Biomed Mater Res A 2019; 107:893-903. [PMID: 30650239 DOI: 10.1002/jbm.a.36612] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/17/2018] [Accepted: 01/09/2019] [Indexed: 02/01/2023]
Abstract
Nasal reconstruction remains a challenge for every reconstructive surgeon. Alloplastic implants are proposed to repair nasal cartilaginous defects but they are often associated with high rates of extrusion and infection and poor biocompatibility. In this context, a porous polymeric scaffold filled with an autologous cartilage gel would be advantageous. In this study, we evaluated the capacity of IEIK13 self-assembling peptide (SAP) to serve as support to form such cartilage gel. Human nasal chondrocytes (HNC) were first amplified with FGF-2 and insulin, and then redifferentiated in IEIK13 with BMP-2, insulin, and T3 (BIT). Our results demonstrate that IEIK13 fosters HNC growth and survival. HNC phenotype was assessed by RT-PCR analysis and neo-synthesized extracellular matrix was characterized by western blotting and immunohistochemistry analysis. BIT-treated cells embedded in IEIK13 displayed round morphology and expressed cartilage-specific markers such as type II and type IX collagens and aggrecan. In addition, we did not detect significant production of type I and type X collagens and gene products of dedifferentiated and hypertrophic chondrocytes that are unwanted in hyaline cartilage. The whole of these results indicates that the SAP IEIK13 represents a suitable support for hydrogel-based tissue engineering of nasal cartilage. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 893-903, 2019.
Collapse
Affiliation(s)
- Alexandre Dufour
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS UMR 5305, Institute for Biology and Chemistry of Proteins, Lyon, France
| | | | - Delphine Vertu-Ciolino
- Department of otolaryngology-head and neck surgery, Édouard-Herriot hospital, Lyon, France
| | - François Disant
- Department of otolaryngology-head and neck surgery, Édouard-Herriot hospital, Lyon, France
| | - Frédéric Mallein-Gerin
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS UMR 5305, Institute for Biology and Chemistry of Proteins, Lyon, France
| | - Emeline Perrier-Groult
- Laboratory of Tissue Biology and Therapeutic Engineering (LBTI), CNRS UMR 5305, Institute for Biology and Chemistry of Proteins, Lyon, France
| |
Collapse
|
19
|
Ning B, Jin R, Wan L, Wang D. Cellular and molecular changes to chondrocytes in an in vitro model of developmental dysplasia of the hip‑an experimental model of DDH with swaddling position. Mol Med Rep 2018; 18:3873-3881. [PMID: 30106106 PMCID: PMC6131662 DOI: 10.3892/mmr.2018.9384] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Accepted: 07/19/2018] [Indexed: 11/18/2022] Open
Abstract
The aim of the present study was to assess the cellular and molecular changes to chondrocytes in a developmental dysplasia of the hip (DDH) model and to investigate the early metabolism of chondrocytes in DDH. Neonatal Wistar rats were used for the DDH model with swaddling position. Primary cultures of chondrocytes were prepared at serial interval stages (2, 4, 6 and 8 weeks) to investigate cellular proliferation. The expression of collagen II and aggrecan mRNA was detected to assess the anabolic ability of chondrocytes. The expression of matrix metallopeptidase (MMP)-13 and ADAM metallopeptidase with thrombospondin type 1 motif 5 (ADAMTS-5) mRNA was measured to investigate the degradation of collagen II and aggrecan, respectively. Morphological changes were observed in coronal dissection samples after the removal of fixation. Primary chondrocytes at serial intervals were assessed using a Cell Counting Kit-8 assay and the results revealed that DDH chondrocytes had more proliferative activity. The expression of collagen II mRNA was upregulated at 2 weeks and was more sensitive to mechanical loading compared with aggrecan. Similar changes occurred at 6 weeks. Furthermore, MMP-13 and ADAMTS-5 mRNA expression levels were upregulated at 2 weeks. It was also demonstrated that DDH chondrocytes exhibited high proliferative activity at the early stages and degeneration later.
Collapse
Affiliation(s)
- Bo Ning
- Department of Pediatric Orthopedics, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| | - Rui Jin
- Department of Pediatric Orthopedics, Children's Hospital of Anhui Medical University, Hefei, Anhui 230051, P.R. China
| | - Lin Wan
- Department of Cardiothoracic Surgery, Shanghai Children's Hospital, Shanghai 230041, P.R. China
| | - Dahui Wang
- Department of Pediatric Orthopedics, Children's Hospital of Fudan University, Shanghai 201102, P.R. China
| |
Collapse
|
20
|
Yamagishi K, Tsukamoto I, Nakamura F, Hashimoto K, Ohtani K, Akagi M. Activation of the renin-angiotensin system in mice aggravates mechanical loading-induced knee osteoarthritis. Eur J Histochem 2018; 62. [PMID: 30043596 PMCID: PMC6060485 DOI: 10.4081/ejh.2018.2930] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 06/17/2018] [Indexed: 12/31/2022] Open
Abstract
Epidemiological studies have shown an association between hypertension and knee osteoarthritis (OA). The purpose of this study was to investigate whether activation of the renin–angiotensin system (RAS) can aggravate mechanical loading-induced knee OA in mice. Eight-week-old male Tsukuba hypertensive mice (THM) and C57BL/6 mice were divided into four groups: i) running THM group, ii) running C57BL/6 mice group, iii) non-running THM group, and iv) non-running C57BL/6 mice group. Mice in the running group were forced to run (25 m/min, 30 min/day, 5 days/week) on a treadmill. All mice in the four groups (n=10 in each group) were euthanized after 0, 2, 4, 6, or 8 weeks of running or natural breeding. Cartilage degeneration in the left knees was histologically evaluated using the modified Mankin score. Expression of Col X, MMP-13, angiotensin type 1 receptor (AT1R), and AT2R was examined immunohistochemically. To study the effects of stimulation of the AT1R in chondrocytes by mechanical loading and/or Angiotensin II (AngII) on transduction of intracellular signals, phosphorylation levels of JNK and Src were measured in bovine articular chondrocytes cultured in three-dimensional agarose scaffolds. After 4 weeks, the mean Mankin score for the lateral femoral condylar cartilage was significantly higher in the THM running group than in the C57BL/6 running group and non-running groups. AT1R and AT2R expression was not detected at 0 weeks in any group but was noted after 4 weeks in the THM running group. AT1R expression was also noted at 8 weeks in the C57BL/6 running group. The expression levels of AT1R, COL X, and MMP-13 in chondrocytes were significantly higher in the THM running group than in the control groups. Positive significant correlations were noted between the Mankin score and the rate of AT1R-immunopositive cells, between the rates of AT1R- and Col X-positive cells, and between the rates of AT1Rand AT2R-positive cells. The phosphorylation level of JNK was increased by cyclic compression loading or addition of AngII to the cultured chondrocytes and was reversed by pretreatment with an AT1R blocker. A synergistic effect on JNK phosphorylation was observed between compression loading and AngII addition. Transgene activation of renin and angiotensinogen aggravated mechanical load-induced knee OA in mice. These findings suggest that AT1R expression in chondrocytes is associated with early knee OA and plays a role in the progression of cartilage degeneration. The RAS may be a common molecular mechanism involved in the pathogenesis of hypertension and knee OA.
Collapse
|
21
|
Lee D, Erickson A, You T, Dudley AT, Ryu S. Pneumatic microfluidic cell compression device for high-throughput study of chondrocyte mechanobiology. LAB ON A CHIP 2018; 18:2077-2086. [PMID: 29897088 PMCID: PMC6467204 DOI: 10.1039/c8lc00320c] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Hyaline cartilage is a specialized type of connective tissue that lines many moveable joints (articular cartilage) and contributes to bone growth (growth plate cartilage). Hyaline cartilage is composed of a single cell type, the chondrocyte, which produces a unique hydrated matrix to resist compressive stress. Although compressive stress has profound effects on transcriptional networks and matrix biosynthesis in chondrocytes, mechanistic relationships between strain, signal transduction, cell metabolism, and matrix production remain superficial. Here, we describe development and validation of a polydimethylsiloxane (PDMS)-based pneumatic microfluidic cell compression device which generates multiple compression conditions in a single platform. The device contained an array of PDMS balloons of different sizes which were actuated by pressurized air, and the balloons compressed chondrocytes cells in alginate hydrogel constructs. Our characterization and testing of the device showed that the developed platform could compress chondrocytes with various magnitudes simultaneously with negligible effect on cell viability. Also, the device is compatible with live cell imaging to probe early effects of compressive stress, and it can be rapidly dismantled to facilitate molecular studies of compressive stress on transcriptional networks. Therefore, the proposed device will enhance the productivity of chondrocyte mechanobiology studies, and it can be applied to study mechanobiology of other cell types.
Collapse
Affiliation(s)
- Donghee Lee
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA.
| | | | | | | | | |
Collapse
|
22
|
Nakamura F, Tsukamoto I, Inoue S, Hashimoto K, Akagi M. Cyclic compressive loading activates angiotensin II type 1 receptor in articular chondrocytes and stimulates hypertrophic differentiation through a G-protein-dependent pathway. FEBS Open Bio 2018; 8:962-973. [PMID: 29928576 PMCID: PMC5986009 DOI: 10.1002/2211-5463.12438] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 03/21/2018] [Accepted: 04/25/2018] [Indexed: 01/26/2023] Open
Abstract
Angiotensin II type 1 receptor (AT1R) appears to have a mechanosensing function in a number of cell types. The purpose of this study was to examine whether AT1R expressed in articular chondrocytes is involved in osteoarthritis (OA) progression in vivo and whether cyclic compressive loading activates the AT1R and stimulates hypertrophic differentiation of chondrocytes in vitro. The relationships between the modified Mankin score for cartilage degeneration and the expression of AT1R and type X collagen (Col X) were studied in mouse knees with OA induced using the destabilization-of-medial-meniscus model. Cyclic compressive loads were applied to cultured bovine articular chondrocytes in three-dimensional agarose scaffolds. Expression of Col X and runt-related transcription factor 2 (Runx2) was analyzed using RT-PCR and western blotting. We dissected the downstream pathway for intracellular signal transductions of AT1R including G-protein-dependent and G-protein-independent pathways. Positive significant correlations between the Mankin score and the rate of AT1R-immunopositive cells and between the rates of AT1R and Col X expression were noted. The expression of Col X and Runx2 was increased by compressive loading but suppressed by addition of olmesartan, an Ang II receptor blocker, to the agarose scaffolds. Compressive loading upregulated the phosphorylation of c-Jun N-terminal kinase (JNK), Src, and STAT1, but olmesartan significantly suppressed only JNK phosphorylation. We conclude that AT1R expressed by articular chondrocytes may be involved in OA progression in vivo. Mechanical stress can activate AT1R and stimulate hypertrophic differentiation of chondrocytes through the G-protein-dependent pathway. AT1R has a mechanosensing function in chondrocytes and may be a new therapeutic target in OA.
Collapse
Affiliation(s)
- Fumihisa Nakamura
- Department of Orthopaedic Surgery Kindai University Hospital Osaka-Sayama Japan
| | - Ichiro Tsukamoto
- Department of Orthopaedic Surgery Kindai University Hospital Osaka-Sayama Japan
| | - Shinji Inoue
- Department of Orthopaedic Surgery Kindai University Hospital Osaka-Sayama Japan
| | - Kazuhiko Hashimoto
- Department of Orthopaedic Surgery Kindai University Hospital Osaka-Sayama Japan
| | - Masao Akagi
- Department of Orthopaedic Surgery Kindai University Hospital Osaka-Sayama Japan
| |
Collapse
|
23
|
Zarrintaj P, Manouchehri S, Ahmadi Z, Saeb MR, Urbanska AM, Kaplan DL, Mozafari M. Agarose-based biomaterials for tissue engineering. Carbohydr Polym 2018; 187:66-84. [PMID: 29486846 DOI: 10.1016/j.carbpol.2018.01.060] [Citation(s) in RCA: 323] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/28/2017] [Accepted: 01/18/2018] [Indexed: 01/08/2023]
Abstract
Agarose is a natural polysaccharide polymer having unique characteristics that give reason to consider it for tissue engineering applications. Special characteristics of agarose such as its excellent biocompatibility, thermo-reversible gelation behavior and physiochemical features support its use as a biomaterial for cell growth and/or controlled/localized drug delivery. The resemblance of this natural carbohydrate polymer to the extracellular matrix results in attractive features that bring about a strong interest in its usage in the field. The scope of this review is to summarize the extensive researches addressing agarose-based biomaterials in order to provide an in-depth understanding of its tissue engineering-related applications.
Collapse
Affiliation(s)
- Payam Zarrintaj
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Saeed Manouchehri
- School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Zahed Ahmadi
- Department of Chemistry, Amirkabir University of Technology, Tehran, Iran
| | - Mohammad Reza Saeb
- Department of Resin and Additives, Institute for Color Science and Technology, P.O. Box: 16765-654, Tehran, Iran.
| | | | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Masoud Mozafari
- Bioengineering Research Group, Nanotechnology and Advanced Materials Department, Materials and Energy Research Center (MERC), Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran; Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
24
|
Anderson DE, Johnstone B. Dynamic Mechanical Compression of Chondrocytes for Tissue Engineering: A Critical Review. Front Bioeng Biotechnol 2017; 5:76. [PMID: 29322043 PMCID: PMC5732133 DOI: 10.3389/fbioe.2017.00076] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/23/2017] [Indexed: 01/19/2023] Open
Abstract
Articular cartilage functions to transmit and translate loads. In a classical structure-function relationship, the tissue resides in a dynamic mechanical environment that drives the formation of a highly organized tissue architecture suited to its biomechanical role. The dynamic mechanical environment includes multiaxial compressive and shear strains as well as hydrostatic and osmotic pressures. As the mechanical environment is known to modulate cell fate and influence tissue development toward a defined architecture in situ, dynamic mechanical loading has been hypothesized to induce the structure-function relationship during attempts at in vitro regeneration of articular cartilage. Researchers have designed increasingly sophisticated bioreactors with dynamic mechanical regimes, but the response of chondrocytes to dynamic compression and shear loading remains poorly characterized due to wide variation in study design, system variables, and outcome measurements. We assessed the literature pertaining to the use of dynamic compressive bioreactors for in vitro generation of cartilaginous tissue from primary and expanded chondrocytes. We used specific search terms to identify relevant publications from the PubMed database and manually sorted the data. It was very challenging to find consensus between studies because of species, age, cell source, and culture differences, coupled with the many loading regimes and the types of analyses used. Early studies that evaluated the response of primary bovine chondrocytes within hydrogels, and that employed dynamic single-axis compression with physiologic loading parameters, reported consistently favorable responses at the tissue level, with upregulation of biochemical synthesis and biomechanical properties. However, they rarely assessed the cellular response with gene expression or mechanotransduction pathway analyses. Later studies that employed increasingly sophisticated biomaterial-based systems, cells derived from different species, and complex loading regimes, did not necessarily corroborate prior positive results. These studies report positive results with respect to very specific conditions for cellular responses to dynamic load but fail to consistently achieve significant positive changes in relevant tissue engineering parameters, particularly collagen content and stiffness. There is a need for standardized methods and analyses of dynamic mechanical loading systems to guide the field of tissue engineering toward building cartilaginous implants that meet the goal of regenerating articular cartilage.
Collapse
Affiliation(s)
- Devon E Anderson
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, OR, United States
| | - Brian Johnstone
- Department of Orthopaedics and Rehabilitation, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
25
|
Erickson AG, Laughlin TD, Romereim SM, Sargus-Patino CN, Pannier AK, Dudley AT. A Tunable, Three-Dimensional In Vitro Culture Model of Growth Plate Cartilage Using Alginate Hydrogel Scaffolds. Tissue Eng Part A 2017; 24:94-105. [PMID: 28525313 DOI: 10.1089/ten.tea.2017.0091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Defining the final size and geometry of engineered tissues through precise control of the scalar and vector components of tissue growth is a necessary benchmark for regenerative medicine, but it has proved to be a significant challenge for tissue engineers. The growth plate cartilage that promotes elongation of the long bones is a good model system for studying morphogenetic mechanisms because cartilage is composed of a single cell type, the chondrocyte; chondrocytes are readily maintained in culture; and growth trajectory is predominately in a single vector. In this cartilage, growth is generated via a differentiation program that is spatially and temporally regulated by an interconnected network composed of long- and short-range signaling mechanisms that together result in the formation of functionally distinct cellular zones. To facilitate investigation of the mechanisms underlying anisotropic growth, we developed an in vitro model of the growth plate cartilage by using neonatal mouse growth plate chondrocytes encapsulated in alginate hydrogel beads. In bead cultures, encapsulated chondrocytes showed high viability, cartilage matrix deposition, low levels of chondrocyte hypertrophy, and a progressive increase in cell proliferation over 7 days in culture. Exogenous factors were used to test functionality of the parathyroid-related protein-Indian hedgehog (PTHrP-IHH) signaling interaction, which is a crucial feedback loop for regulation of growth. Consistent with in vivo observations, exogenous PTHrP stimulated cell proliferation and inhibited hypertrophy, whereas IHH signaling stimulated chondrocyte hypertrophy. Importantly, the treatment of alginate bead cultures with IHH or thyroxine resulted in formation of a discrete domain of hypertrophic cells that mimics tissue architecture of native growth plate cartilage. Together, these studies are the first demonstration of a tunable in vitro system to model the signaling network interactions that are required to induce zonal architecture in growth plate chondrocytes, which could also potentially be used to grow cartilage cultures of specific geometries to meet personalized patient needs.
Collapse
Affiliation(s)
- Alek G Erickson
- 1 Department of Genetics, Cell Biology, and Anatomy, University Nebraska Medical Center , Omaha, Nebraska
| | - Taylor D Laughlin
- 2 Department of Biological Systems Engineering, University Nebraska Lincoln , Lincoln, Nebraska
| | - Sarah M Romereim
- 1 Department of Genetics, Cell Biology, and Anatomy, University Nebraska Medical Center , Omaha, Nebraska.,3 Department of Animal Science, University Nebraska Lincoln , Lincoln, Nebraska
| | | | - Angela K Pannier
- 2 Department of Biological Systems Engineering, University Nebraska Lincoln , Lincoln, Nebraska
| | - Andrew T Dudley
- 1 Department of Genetics, Cell Biology, and Anatomy, University Nebraska Medical Center , Omaha, Nebraska
| |
Collapse
|
26
|
Mechanical compression induces VEGFA overexpression in breast cancer via DNMT3A-dependent miR-9 downregulation. Cell Death Dis 2017; 8:e2646. [PMID: 28252641 PMCID: PMC5386566 DOI: 10.1038/cddis.2017.73] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 01/08/2017] [Accepted: 02/02/2017] [Indexed: 02/07/2023]
Abstract
Tumor growth generates mechanical compression, which may trigger mechanotransduction in cancer and stromal cells and promote tumor progression. However, very little is known about how compression stimulates signal transduction and contributes to tumor progression. In the present study, we demonstrated that compression enhances a tumor progression phenotype using an in vitro compression model, and validated the results from the in vitro model with high- and low-compressed breast cancer tissues. Mechanical compression induced miR-9 downregulation by DNMT3A-dependent promoter methylation in the MDA-MB-231 and BT-474 breast cancer cell lines and in cancer-associated fibroblasts. The overexpression of miR-9 target genes (LAMC2, ITGA6, and EIF4E) was induced by miR-9 downregulation, which eventually enhanced vascular endothelial growth factors production. Demethylation and decompression could reverse compression-induced miR-9 downregulation and following overexpression of miR-9 target genes and VEGFA.
Collapse
|
27
|
Carlson AK, McCutchen CN, June RK. Mechanobiological implications of articular cartilage crystals. Curr Opin Rheumatol 2017; 29:157-162. [DOI: 10.1097/bor.0000000000000368] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
28
|
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: 5.4] [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
|
29
|
Chen C, Wei X, Lv Z, Sun X, Wang S, Zhang Y, Jiao Q, Wang X, Li Y, Wei L. Cyclic Equibiaxial Tensile Strain Alters Gene Expression of Chondrocytes via Histone Deacetylase 4 Shuttling. PLoS One 2016; 11:e0154951. [PMID: 27149270 PMCID: PMC4858146 DOI: 10.1371/journal.pone.0154951] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 04/21/2016] [Indexed: 12/23/2022] Open
Abstract
Objectives This paper aims to investigate whether equibiaxial tensile strain alters chondrocyte gene expression via controlling subcellular localization of histone deacetylase 4 (HDAC4). Materials and Methods Murine chondrocytes transfected with GFP-HDAC4 were subjected to 3 h cyclic equibiaxial tensile strain (CTS, 6% strain at 0.25 Hz) by a Flexcell® FX-5000™ Tension System. Fluorescence microscope and western blot were used to observe subcellular location of HDAC4. The gene expression was analyzed by real-time RT-PCR. The concentration of Glycosaminoglycans in culture medium was quantified by bimethylmethylene blue dye; Collagen II protein was evaluated by western blot. Cells phenotype was identified by immunohistochemistry. Cell viability was evaluated by live-dead cell detect kit. Okadaic acid, an inhibitor of HDAC4 nuclear relocation, was used to further validate whether HDAC4 nuclear relocation plays a role in gene expression in response to tension stimulation. Results 87.5% of HDAC4 was located in the cytoplasm in chondrocytes under no loading condition, but it was relocated to the nucleus after CTS. RT-PCR analysis showed that levels of mRNA for aggrecan, collagen II, LK1 and SOX9 were all increased in chondrocytes subjected to CTS as compared to no loading control chondrocytes; in contrast, the levels of type X collagen, MMP-13, IHH and Runx2 gene expression were decreased in the chondrocytes subjected to CTS as compared to control chondrocytes. Meanwhile, CTS contributed to elevation of glycosaminoglycans and collagen II protein, but did not change collagen I production. When Okadaic acid blocked HDAC4 relocation from the cytoplasm to nucleus, the changes of the chondrocytes induced by CTS were abrogated. There was no chondrocyte dead detected in this study in response to CTS. Conclusions CTS is able to induce HDAC4 relocation from cytoplasm to nucleus. Thus, CTS alters chondrocytes gene expression in association with the relocation of HDAC4 induced by CTS.
Collapse
Affiliation(s)
- Chongwei Chen
- Department of Orthopaedics, the Second Hospital of Shanxi Medical University; Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, China
| | - Xiaochun Wei
- Department of Orthopaedics, the Second Hospital of Shanxi Medical University; Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, China
| | - Zhi Lv
- Department of Orthopaedics, the Second Hospital of Shanxi Medical University; Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, China
| | - Xiaojuan Sun
- Department of Orthopaedics, the Second Hospital of Shanxi Medical University; Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, China
| | - Shaowei Wang
- Department of Orthopaedics, the Second Hospital of Shanxi Medical University; Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, China
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hosptal, Providence, Rhode Island, United States of America
| | - Yang Zhang
- Department of Orthopaedics, the Second Hospital of Shanxi Medical University; Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, China
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hosptal, Providence, Rhode Island, United States of America
| | - Qiang Jiao
- Department of Orthopaedics, the Second Hospital of Shanxi Medical University; Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, China
| | - Xiaohu Wang
- Department of Orthopaedics, the Second Hospital of Shanxi Medical University; Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, China
| | - Yongping Li
- Department of Orthopaedics, the Second Hospital of Shanxi Medical University; Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, China
| | - Lei Wei
- Department of Orthopaedics, the Second Hospital of Shanxi Medical University; Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan, Shanxi, China
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hosptal, Providence, Rhode Island, United States of America
- * E-mail:
| |
Collapse
|
30
|
Chen C, Wei X, Wang S, Jiao Q, Zhang Y, Du G, Wang X, Wei F, Zhang J, Wei L. Compression regulates gene expression of chondrocytes through HDAC4 nuclear relocation via PP2A-dependent HDAC4 dephosphorylation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1633-42. [PMID: 27106144 DOI: 10.1016/j.bbamcr.2016.04.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 04/13/2016] [Accepted: 04/17/2016] [Indexed: 11/15/2022]
Abstract
Biomechanics plays a critical role in the modulation of chondrocyte function. The mechanisms by which mechanical loading is transduced into intracellular signals that regulate chondrocyte gene expression remain largely unknown. Histone deacetylase 4 (HDAC4) is specifically expressed in chondrocytes. Mice lacking HDAC4 display chondrocyte hypertrophy, ectopic and premature ossification, and die early during the perinatal period. HDAC4 has a remarkable ability to translocate between the cell's cytoplasm and nucleus. It has been established that subcellular relocation of HDAC4 plays a critical role in chondrocyte differentiation and proliferation. However, it remains unclear whether subcellular relocation of HDAC4 in chondrocytes can be induced by mechanical loading. In this study, we first report that compressive loading induces HDAC4 relocation from the cytoplasm to the nucleus of chondrocytes via stimulation of Ser/Thr-phosphoprotein phosphatases 2A (PP2A) activity, which results in dephosphorylation of HDAC4. Dephosphorylated HDAC4 relocates to the nucleus to achieve transcriptional repression of Runx2 and regulates chondrocyte gene expression in response to compression. Our results elucidate the mechanism by which mechanical compression regulates chondrocyte gene expression through HDAC4 relocation from the cell's cytoplasm to the nucleus via PP2A-dependent HDAC4 dephosphorylation.
Collapse
Affiliation(s)
- Chongwei Chen
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Xiaochun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Shaowei Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China; Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | - Qiang Jiao
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Yang Zhang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China; Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | - Guoqing Du
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA
| | - Xiaohu Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China
| | - Fangyuan Wei
- Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Jianzhong Zhang
- Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Lei Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Taiyuan 030001, China; Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI 02903, USA.
| |
Collapse
|
31
|
Correro-Shahgaldian MR, Introvigne J, Ghayor C, Weber FE, Gallo LM, Colombo V. Properties and Mechanobiological Behavior of Bovine Nasal Septum Cartilage. Ann Biomed Eng 2015; 44:1821-31. [PMID: 26502171 DOI: 10.1007/s10439-015-1481-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 10/03/2015] [Indexed: 12/23/2022]
Abstract
Bovine nasal septum (BNS) is a source of non-load bearing hyaline cartilage. Little information is available on its mechanical and biological properties. The aim of this work was to assess the characteristics of BNS cartilage and investigate its behavior in in vitro mechanobiological experiments. Mechanical tests, biochemical assays, and microscopic assessment were performed for tissue characterization. Compressions tests showed that the tissue is viscoelastic, although values of elastic moduli differ from the ones of other cartilaginous tissues. Water content was 78 ± 1.4%; glycosaminoglycans and collagen contents-measured by spectrophotometric assay and hydroxyproline assay-were 39 ± 5% and 25 ± 2.5% of dry weight, respectively. Goldner's Trichrome staining and transmission electron microscopy proved isotropic cells distribution and results of earlier cell division. Furthermore, gene expression was measured after uniaxial compression, showing variations depending on compression time as well as trends depending on equilibration time. In conclusion, BNS has been characterized at several levels, revealing that bovine nasal tissue is regionally homogeneous. Results suggest that, under certain conditions, BNS could be used to perform in vitro cartilage loading experiments.
Collapse
Affiliation(s)
- Maria Rita Correro-Shahgaldian
- Clinic for Masticatory Disorders, Removable Prosthodontics and Special Care, Center for Dental Medicine, University of Zurich, Plattenstrasse 11, 8032, Zurich, Switzerland.,Oral Biotechnology & Bioengineering, Department of Cranio-Maxillofacial Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Jasmin Introvigne
- Clinic for Masticatory Disorders, Removable Prosthodontics and Special Care, Center for Dental Medicine, University of Zurich, Plattenstrasse 11, 8032, Zurich, Switzerland
| | - Chafik Ghayor
- Oral Biotechnology & Bioengineering, Department of Cranio-Maxillofacial Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Franz E Weber
- Oral Biotechnology & Bioengineering, Department of Cranio-Maxillofacial Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Luigi M Gallo
- Clinic for Masticatory Disorders, Removable Prosthodontics and Special Care, Center for Dental Medicine, University of Zurich, Plattenstrasse 11, 8032, Zurich, Switzerland
| | - Vera Colombo
- Clinic for Masticatory Disorders, Removable Prosthodontics and Special Care, Center for Dental Medicine, University of Zurich, Plattenstrasse 11, 8032, Zurich, Switzerland.
| |
Collapse
|
32
|
The AP-1 transcription factor homolog Pf-AP-1 activates transcription of multiple biomineral proteins and potentially participates in Pinctada fucata biomineralization. Sci Rep 2015; 5:14408. [PMID: 26404494 PMCID: PMC4585884 DOI: 10.1038/srep14408] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 08/26/2015] [Indexed: 12/25/2022] Open
Abstract
Activator protein-1 (AP-1) is an important bZIP transcription factor that regulates a series of physiological processes by specifically activating transcription of several genes, and one of its well-chartered functions in mammals is participating in bone mineralization. We isolated and cloned the complete cDNA of a Jun/AP-1 homolog from Pinctada fucata and called it Pf-AP-1. Pf-AP-1 had a highly conserved bZIP region and phosphorylation sites compared with those from mammals. A tissue distribution analysis showed that Pf-AP-1 was ubiquitously expressed in P. fucata and the mRNA level of Pf-AP-1 is extremely high in mantle. Pf-AP-1 expression was positively associated with multiple biomineral proteins in the mantle. The luciferase reporter assay in a mammalian cell line showed that Pf-AP-1 significantly up-regulates the transcriptional activity of the promoters of KRMP, Pearlin, and Prisilkin39. Inhibiting the activity of Pf-AP-1 depressed the expression of multiple matrix proteins. Pf-AP-1 showed a unique expression pattern during shell regeneration and pearl sac development, which was similar to the pattern observed for biomineral proteins. These results suggest that the Pf-AP-1 AP-1 homolog is an important transcription factor that regulates transcription of several biomineral proteins simultaneously and plays a role in P. fucata biomineralization, particularly during pearl and shell formation.
Collapse
|
33
|
Cartilage tissue engineering: molecular control of chondrocyte differentiation for proper cartilage matrix reconstruction. Biochim Biophys Acta Gen Subj 2014; 1840:2414-40. [PMID: 24608030 DOI: 10.1016/j.bbagen.2014.02.030] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 02/06/2014] [Accepted: 02/26/2014] [Indexed: 12/18/2022]
Abstract
BACKGROUND Articular cartilage defects are a veritable therapeutic problem because therapeutic options are very scarce. Due to the poor self-regeneration capacity of cartilage, minor cartilage defects often lead to osteoarthritis. Several surgical strategies have been developed to repair damaged cartilage. Autologous chondrocyte implantation (ACI) gives encouraging results, but this cell-based therapy involves a step of chondrocyte expansion in a monolayer, which results in the loss in the differentiated phenotype. Thus, despite improvement in the quality of life for patients, reconstructed cartilage is in fact fibrocartilage. Successful ACI, according to the particular physiology of chondrocytes in vitro, requires active and phenotypically stabilized chondrocytes. SCOPE OF REVIEW This review describes the unique physiology of cartilage, with the factors involved in its formation, stabilization and degradation. Then, we focus on some of the most recent advances in cell therapy and tissue engineering that open up interesting perspectives for maintaining or obtaining the chondrogenic character of cells in order to treat cartilage lesions. MAJOR CONCLUSIONS Current research involves the use of chondrocytes or progenitor stem cells, associated with "smart" biomaterials and growth factors. Other influential factors, such as cell sources, oxygen pressure and mechanical strain are considered, as are recent developments in gene therapy to control the chondrocyte differentiation/dedifferentiation process. GENERAL SIGNIFICANCE This review provides new information on the mechanisms regulating the state of differentiation of chondrocytes and the chondrogenesis of mesenchymal stem cells that will lead to the development of new restorative cell therapy approaches in humans. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
|
34
|
Sassi N, Gadgadi N, Laadhar L, Allouche M, Mourali S, Zandieh-Doulabi B, Hamdoun M, Nulend JK, Makni S, Sellami S. Notch signaling is involved in human articular chondrocytes de-differentiation during osteoarthritis. J Recept Signal Transduct Res 2013; 34:48-57. [PMID: 24251351 DOI: 10.3109/10799893.2013.856920] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
CONTEXT During osteoarthritis (OA), chondrocytes undergo de-differentiation, resulting in the acquisition of a fibroblast-like morphology, decreased expression of collagen type II (colII) and aggrecan, and increased expression of collagen type I (colI), metalloproteinase 13 (MMP13) and nitric oxide synthase (eNOS). Notch signaling plays a crucial role during embryogenesis. Several studies showed that Notch is expressed in adulthood. OBJECTIVE The aim of our study was to confirm the involvement of Notch signaling in human OA at in vitro and ex vivo levels. MATERIALS AND METHODS Normal human articular chondrocytes were cultured during four passages either treated or not with a Notch inhibitor: DAPT. Human OA cartilage was cultured with DAPT for five days. Chondrocytes secreted markers and some Notch pathway components were analyzed using Western blotting and qPCR. RESULTS Passaging chondrocytes induced a decrease in the cartilage markers: colII and aggrecan. DAPT-treated chondrocytes and OA cartilage showed a significant increase in healthy cartilage markers. De-differentiation markers, colI, MMP13 and eNOS, were significantly reduced in DAPT-treated chondrocytes and OA cartilage. Notch1 expression was proportional to colI, MMP13 and eNOS expression and inversely proportional to colII and aggrecan expression in nontreated cultured chondrocytes. Notch ligand: Jagged1 increased in chondrocytes culture. DAPT treatment resulted in reduced Jagged1 expression. Notch target gene HES1 increased during chondrocyte culture and was reduced when treated with DAPT. CONCLUSION Targeting Notch signaling during OA might lead to the restitution of the typical chondrocyte phenotype and even to chondrocyte redifferentiation during the pathology.
Collapse
Affiliation(s)
- Nadia Sassi
- Department of Rheumatology, Immuno-Rheumatology Research Laboratory, La Rabta Hospital, University of Tunis-El Manar , Tunis , Tunisia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zignego DL, Jutila AA, Gelbke MK, Gannon DM, June RK. The mechanical microenvironment of high concentration agarose for applying deformation to primary chondrocytes. J Biomech 2013; 47:2143-8. [PMID: 24275437 DOI: 10.1016/j.jbiomech.2013.10.051] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 10/25/2013] [Accepted: 10/31/2013] [Indexed: 11/16/2022]
Abstract
Cartilage and chondrocytes experience loading that causes alterations in chondrocyte biological activity. In vivo chondrocytes are surrounded by a pericellular matrix with a stiffness of ~25-200kPa. Understanding the mechanical loading environment of the chondrocyte is of substantial interest for understanding chondrocyte mechanotransduction. The first objective of this study was to analyze the spatial variability of applied mechanical deformations in physiologically stiff agarose on cellular and sub-cellular length scales. Fluorescent microspheres were embedded in physiologically stiff agarose hydrogels. Microsphere positions were measured via confocal microscopy and used to calculate displacement and strain fields as a function of spatial position. The second objective was to assess the feasibility of encapsulating primary human chondrocytes in physiologically stiff agarose. The third objective was to determine if primary human chondrocytes could deform in high-stiffness agarose gels. Primary human chondrocyte viability was assessed using live-dead imaging following 24 and 72h in tissue culture. Chondrocyte shape was measured before and after application of 10% compression. These data indicate that (1) displacement and strain precision are ~1% and 6.5% respectively, (2) high-stiffness agarose gels can maintain primary human chondrocyte viability of >95%, and (3) compression of chondrocytes in 4.5% agarose can induce shape changes indicative of cellular compression. Overall, these results demonstrate the feasibility of using high-concentration agarose for applying in vitro compression to chondrocytes as a model for understanding how chondrocytes respond to in vivo loading.
Collapse
Affiliation(s)
- Donald L Zignego
- Department of Mechanical and Industrial Engineering, Montana State University, Bozeman, MT 59718-3800, USA
| | - Aaron A Jutila
- Department of Mechanical and Industrial Engineering, Montana State University, Bozeman, MT 59718-3800, USA
| | - Martin K Gelbke
- Bridger Orthopedic and Sports Medicine, Bozeman, MT 59715, USA
| | - Daniel M Gannon
- Bridger Orthopedic and Sports Medicine, Bozeman, MT 59715, USA
| | - Ronald K June
- Department of Mechanical and Industrial Engineering, Montana State University, Bozeman, MT 59718-3800, USA; Department of Cell Biology and Neuroscience, Montana State University, Bozeman, MT 59718-3800, USA.
| |
Collapse
|
36
|
Xu H, Zhang X, Wang H, Zhang Y, Shi Y, Zhang X. Continuous cyclic mechanical tension increases ank expression in endplate chondrocytes through the TGF-β1 and p38 pathway. Eur J Histochem 2013; 57:e28. [PMID: 24085277 PMCID: PMC3794359 DOI: 10.4081/ejh.2013.e28] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2013] [Revised: 08/22/2013] [Accepted: 08/23/2013] [Indexed: 01/06/2023] Open
Abstract
The normal ANK protein has a strong influence on anti-calcification. It is known that TGF-β1 is also able to induce extracellular inorganic pyrophosphate (ePPi) elaboration via the TGF-β1-induced ank gene expression and the mitogen-activated protein kinase (MAPK) signaling acts as a downstream effector of TGF-β1. We hypothesized that the expression of the ank gene is regulated by mechanics through TGF-β1-p38 pathway. In this study, we investigated the mechanism of short-time mechanical tension-induced ank gene expression. We found that the continuous cyclic mechanical tension (CCMT) increased the ank gene expression in the endplate chondrocytes, and there was an increase in the TGF-β1 expression after CCMT stimulation. The ank gene expression significantly increased when treated by TGF-β1 in a dose-dependent manner and decreased when treated by SB431542 (ALK inhibitor) in a dose-dependent manner. Our study results indicate that CCMT-induced ank gene expressions may be regulated by TGF-β1 and p38 MAPK pathway.
Collapse
|
37
|
Sassi N, Laadhar L, Allouche M, Zandieh-Doulabi B, Hamdoun M, Klein-Nulend J, Makni S, Sellami S. Wnt signaling is involved in human articular chondrocyte de-differentiation in vitro. Biotech Histochem 2013; 89:29-40. [PMID: 23901947 DOI: 10.3109/10520295.2013.811285] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Osteoarthritis is the most prevalent form of arthritis in the world. Certain signaling pathways, such as the wnt pathway, are involved in cartilage pathology. Osteoarthritic chondrocytes undergo morphological and biochemical changes that lead to chondrocyte de-differentiation. We investigated whether the Wnt pathway is involved in de-differentiation of human articular chondrocytes in vitro. Human articular chondrocytes were cultured for four passages in the presence or absence of IL-1 in monolayer or micromass culture. Changes in cell morphology were monitored by light microscopy. Protein and gene expression of chondrocyte markers and Wnt pathway components were determined by Western blotting and qPCR after culture. After culturing for four passages, chondrocytes exhibited a fibroblast-like morphology. Collagen type II and aggrecan protein and gene expression decreased, while collagen type I, matrix metalloproteinase 13, and nitric oxide synthase expressions increased. Wnt molecule expression profiles changed; Wnt5a protein expression, the Wnt target gene, c-jun, and in Wnt pathway regulator, sFRP4 increased. Treatment with IL-1 caused chondrocyte morphology to become more filament-like. This change in morphology was accompanied by extinction of col II expression and increased col I, MMP13 and eNOS expression. Changes in expression of the Wnt pathway components also were observed. Wnt7a decreased significantly, while Wnt5a, LRP5, β-catenin and c-jun expressions increased. Culture of human articular chondrocytes with or without IL-1 not only induced chondrocyte de-differentiation, but also changed the expression profiles of Wnt components, which suggests that the Wnt pathway is involved in chondrocyte de-differentiation in vitro.
Collapse
Affiliation(s)
- N Sassi
- Immuno-Rheumatology Research Laboratory, Department of Rheumatology, La Rabta Hospital, University of Tunis-El Manar
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Gadjanski I, Yodmuang S, Spiller K, Bhumiratana S, Vunjak-Novakovic G. Supplementation of exogenous adenosine 5'-triphosphate enhances mechanical properties of 3D cell-agarose constructs for cartilage tissue engineering. Tissue Eng Part A 2013; 19:2188-200. [PMID: 23651296 DOI: 10.1089/ten.tea.2012.0352] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Formation of tissue-engineered cartilage is greatly enhanced by mechanical stimulation. However, direct mechanical stimulation is not always a suitable method, and the utilization of mechanisms underlying mechanotransduction might allow for a highly effective and less aggressive alternate means of stimulation. In particular, the purinergic, adenosine 5'-triphosphate (ATP)-mediated signaling pathway is strongly implicated in mechanotransduction within the articular cartilage. We investigated the effects of transient and continuous exogenous ATP supplementation on mechanical properties of cartilaginous constructs engineered using bovine chondrocytes and human mesenchymal stem cells (hMSCs) encapsulated in an agarose hydrogel. For both cell types, we have observed significant increases in equilibrium and dynamic compressive moduli after transient ATP treatment applied in the fourth week of cultivation. Continuous ATP treatment over 4 weeks of culture only slightly improved the mechanical properties of the constructs, without major changes in the total glycosaminoglycan (GAG) and collagen content. Structure-function analyses showed that transiently ATP-treated constructs, and in particular those based on hMSCs, had the highest level of correlation between compositional and mechanical properties. Transiently treated groups showed intense staining of the territorial matrix for GAGs and collagen type II. These results indicate that transient ATP treatment can improve functional mechanical properties of cartilaginous constructs based on chondrogenic cells and agarose hydrogels, possibly by improving the structural organization of the bulk phase and territorial extracellular matrix (ECM), that is, by increasing correlation slopes between the content of the ECM components (GAG, collagen) and mechanical properties of the construct.
Collapse
Affiliation(s)
- Ivana Gadjanski
- Department of Biomedical Engineering, Columbia University, 622 West 168th Street, New York, NY 10032, USA
| | | | | | | | | |
Collapse
|
39
|
Perrier-Groult E, Pasdeloup M, Malbouyres M, Galéra P, Mallein-Gerin F. Control of collagen production in mouse chondrocytes by using a combination of bone morphogenetic protein-2 and small interfering RNA targeting Col1a1 for hydrogel-based tissue-engineered cartilage. Tissue Eng Part C Methods 2013; 19:652-64. [PMID: 23311625 DOI: 10.1089/ten.tec.2012.0396] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Because articular cartilage does not self-repair, tissue-engineering strategies should be considered to regenerate this tissue. Autologous chondrocyte implantation is already used for treatment of focal damage of articular cartilage. Unfortunately, this technique includes a step of cell amplification, which results in dedifferentiation of chondrocytes, with expression of type I collagen, a protein characteristic of fibrotic tissues. Therefore, the risk of producing a fibrocartilage exists. The aim of this study was to propose a new strategy for authorizing the recovery of the differentiated status of the chondrocytes after their amplification on plastic. Because the bone morphogenetic protein (BMP)-2 and the transforming growth factor (TGF)-β1 are cytokines both proposed as stimulants for cartilage repair, we undertook a detailed comparative analysis of their biological effects on chondrocytes. As a cellular model, we used mouse chondrocytes after their expansion on plastic and we tested the capability of BMP-2 or TGF-β1 to drive their redifferentiation, with special attention given to the nature of the proteins synthesized by the cells. To prevent any fibrotic character of the newly synthesized extracellular matrix, we silenced type I collagen by transfecting small interfering RNA (siRNA) into the chondrocytes, before their exposure to BMP-2 or TGF-β1. Our results showed that addition of siRNA targeting the mRNA encoded by the Col1a1 gene (Col1a1 siRNA) and BMP-2 represents the most efficient combination to control the production of cartilage-characteristic collagen proteins. To go one step further toward scaffold-based cartilage engineering, Col1a1 siRNA-transfected chondrocytes were encapsulated in agarose hydrogel and cultured in vitro for 1 week. The analysis of the chondrocyte-agarose constructs by using real-time polymerase chain reaction, Western-blotting, immunohistochemistry, and electron microscopy techniques demonstrated that the BMP-2/Col1a1 siRNA combination is effective in reinitializing correct production and assembly of the cartilage-characteristic matrix in agarose hydrogel, without production of type I collagen. Because agarose is known to favor long-term expression of the chondrocyte phenotype and agarose-based hydrogels are approved for clinical trials, this strategy appears very promising to repair hyaline cartilage.
Collapse
|
40
|
Rodrigues MT, Lee SJ, Gomes ME, Reis RL, Atala A, Yoo JJ. Bilayered constructs aimed at osteochondral strategies: the influence of medium supplements in the osteogenic and chondrogenic differentiation of amniotic fluid-derived stem cells. Acta Biomater 2012; 8:2795-806. [PMID: 22510402 DOI: 10.1016/j.actbio.2012.04.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Revised: 04/04/2012] [Accepted: 04/09/2012] [Indexed: 12/18/2022]
Abstract
The development of osteochondral tissue engineered interfaces would be a novel treatment for traumatic injuries and aging associated diseases that affect joints. This study reports the development of a bilayered scaffold, which consists of both bone and cartilage regions. On the other hand, amniotic fluid-derived stem cells (AFSCs) could be differentiated into either osteogenic or chondrogenic cells, respectively. In this study we have developed a bilayered scaffolding system, which includes a starch/polycaprolactone (SPCL) scaffold for osteogenesis and an agarose hydrogel for chondrogenesis. AFSC-seeded scaffolds were cultured for 1 or 2 weeks in an osteochondral-defined culture medium containing both osteogenic and chondrogenic differentiation factors. Additionally, the effect of the presence or absence of insulin-like growth factor-1 (IGF-1) in the culture medium was assessed. Cell viability and phenotypic expression were assessed within the constructs in order to determine the influence of the osteochondral differentiation medium. The results indicated that, after osteogenic differentiation, AFSCs that had been seeded onto SPCL scaffolds did not require osteochondral medium to maintain their phenotype, and they produced a protein-rich, mineralized extracellular matrix (ECM) for up to 2 weeks. However, AFSCs differentiated into chondrocyte-like cells appeared to require osteochondral medium, but not IGF-1, to synthesize ECM proteins and maintain the chondrogenic phenotype. Thus, although IGF-1 was not essential for creating osteochondral constructs with AFSCs in this study, the osteochondral supplements used appear to be important to generate cartilage in long-term tissue engineering approaches for osteochondral interfaces. In addition, constructs generated from agarose-SPCL bilayered scaffolds containing pre-differentiated AFSCs may be useful for potential applications in regeneration strategies for damaged or diseased joints.
Collapse
Affiliation(s)
- Márcia T Rodrigues
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | | | | | | | | | | |
Collapse
|
41
|
Dynamic compression of chondrocyte-agarose constructs reveals new candidate mechanosensitive genes. PLoS One 2012; 7:e36964. [PMID: 22615857 PMCID: PMC3355169 DOI: 10.1371/journal.pone.0036964] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Accepted: 04/16/2012] [Indexed: 11/19/2022] Open
Abstract
Articular cartilage is physiologically exposed to repeated loads. The mechanical properties of cartilage are due to its extracellular matrix, and homeostasis is maintained by the sole cell type found in cartilage, the chondrocyte. Although mechanical forces clearly control the functions of articular chondrocytes, the biochemical pathways that mediate cellular responses to mechanical stress have not been fully characterised. The aim of our study was to examine early molecular events triggered by dynamic compression in chondrocytes. We used an experimental system consisting of primary mouse chondrocytes embedded within an agarose hydrogel; embedded cells were pre-cultured for one week and subjected to short-term compression experiments. Using Western blots, we demonstrated that chondrocytes maintain a differentiated phenotype in this model system and reproduce typical chondrocyte-cartilage matrix interactions. We investigated the impact of dynamic compression on the phosphorylation state of signalling molecules and genome-wide gene expression. After 15 min of dynamic compression, we observed transient activation of ERK1/2 and p38 (members of the mitogen-activated protein kinase (MAPK) pathways) and Smad2/3 (members of the canonical transforming growth factor (TGF)-β pathways). A microarray analysis performed on chondrocytes compressed for 30 min revealed that only 20 transcripts were modulated more than 2-fold. A less conservative list of 325 modulated genes included genes related to the MAPK and TGF-β pathways and/or known to be mechanosensitive in other biological contexts. Of these candidate mechanosensitive genes, 85% were down-regulated. Down-regulation may therefore represent a general control mechanism for a rapid response to dynamic compression. Furthermore, modulation of transcripts corresponding to different aspects of cellular physiology was observed, such as non-coding RNAs or primary cilium. This study provides new insight into how chondrocytes respond to mechanical forces.
Collapse
|
42
|
Noriega S, Budhiraja G, Subramanian A. Remodeling of chromatin under low intensity diffuse ultrasound. Int J Biochem Cell Biol 2012; 44:1331-6. [PMID: 22575092 DOI: 10.1016/j.biocel.2012.04.027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Revised: 04/23/2012] [Accepted: 04/30/2012] [Indexed: 11/15/2022]
Abstract
A variety of mechanotransduction pathways mediate the response of fibroblasts or chondrocytes to ultrasound stimulation. In addition, regulatory pathways that co-ordinate stimulus-specific cellular responses are likely to exist. In this study, analysis was confined to the hypothesis that ultrasound stimulation (US) influences the chromatin structure, and that these changes may reflect a regulatory pathway that connects nuclear architecture, chromatin structure and gene expression. Murine fibroblasts seeded on tissue culture plates were stimulated with US (5.0 MHz (14 kPa), 51-s per application) and the thermal denaturation profiles of nuclei isolated from fibroblasts were assessed by dynamic scanning calorimetry (DSC). When compared to the thermal profiles obtained from the nuclei of non-stimulated cells, the nuclei obtained from stimulated cells showed a change in peak profiles and peak areas, which is indicative of chromatin remodeling. Independently, US was also observed to impact the histone (H1):chromatin association as measured indirectly by DAPI staining. Based on our work, it appears plausible that US can produce a remodeling of chromatin, thus triggering signal cascade and other intracellular mechanisms.
Collapse
Affiliation(s)
- Sandra Noriega
- Department of Chemical and Biomolecular Engineering, University of Nebraska, Lincoln, Lincoln, NE 68516, USA
| | | | | |
Collapse
|
43
|
Biomechanical influence of cartilage homeostasis in health and disease. ARTHRITIS 2011; 2011:979032. [PMID: 22046527 PMCID: PMC3196252 DOI: 10.1155/2011/979032] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/26/2011] [Indexed: 11/30/2022]
Abstract
There is an urgent demand for long term solutions to improve osteoarthritis treatments in the ageing population. There are drugs that control the pain but none that stop the progression of the disease in a safe and efficient way. Increased intervention efforts, augmented by early diagnosis and integrated biophysical therapies are therefore needed. Unfortunately, progress has been hampered due to the wide variety of experimental models which examine the effect of mechanical stimuli and inflammatory mediators on signal transduction pathways. Our understanding of the early mechanopathophysiology is poor, particularly the way in which mechanical stimuli influences cell function and regulates matrix synthesis. This makes it difficult to identify reliable targets and design new therapies. In addition, the effect of mechanical loading on matrix turnover is dependent on the nature of the mechanical stimulus. Accumulating evidence suggests that moderate mechanical loading helps to maintain cartilage integrity with a low turnover of matrix constituents. In contrast, nonphysiological mechanical signals are associated with increased cartilage damage and degenerative changes. This review will discuss the pathways regulated by compressive loading regimes and inflammatory signals in animal and in vitro 3D models. Identification of the chondroprotective pathways will reveal novel targets for osteoarthritis treatments.
Collapse
|
44
|
Claus S, Aubert-Foucher E, Perrier-Groult E, Bougault C, Ronzière MC, Freyria AM, Legendre F, Ollitrault D, Boumediene K, Demoor M, Galera P, Tian T, Flajollet S, Duterque-Coquillaud M, Damour O, Chajra H, Mallein-Gerin F. Décryptage des signalisations moléculaires contrôlant la différenciation des chondrocytes : retombées pour l’ingénierie tissulaire du cartilage : le projet ANR-TecSan PROMOCART. Ing Rech Biomed 2011. [DOI: 10.1016/j.irbm.2011.01.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
45
|
Bougault C, Paumier A, Aubert-Foucher E, Mallein-Gerin F. Investigating conversion of mechanical force into biochemical signaling in three-dimensional chondrocyte cultures. Nat Protoc 2009; 4:928-38. [PMID: 19478808 DOI: 10.1038/nprot.2009.63] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The culture of chondrocytes embedded within agarose hydrogels maintains chondrocytic phenotype over extended periods and allows analysis of the chondrocyte response to mechanical forces. The mechanisms involved in the transduction of a mechanical stimulus to a physiological process are not completely deciphered. We present protocols to prepare and characterize constructs of murine chondrocytes and agarose (1 week pre-culture period), to analyze the effect of compression on mRNA level by RT-PCR (2-3 d), gene transcription by gene reporter assay (3 d) and phosphorylation state of signaling molecules by western blotting (3-4 d). The protocols can be carried out with a limited number of mouse embryos or newborns and this point is particularly important regarding genetically modified mice.
Collapse
Affiliation(s)
- Carole Bougault
- UMR5086, CNRS, IFR128, IBCP (Institut de Biologie et Chimie des Protéines), Université de Lyon, Lyon, France
| | | | | | | |
Collapse
|
46
|
|
47
|
Abstract
Articular cartilage repair and regeneration continue to be largely intractable because of the poor regenerative properties of this tissue. The field of articular cartilage tissue engineering, which aims to repair, regenerate, and/or improve injured or diseased articular cartilage functionality, has evoked intense interest and holds great potential for improving articular cartilage therapy. This review provides an overall description of the current state of and progress in articular cartilage repair and regeneration. Traditional therapies and related problems are introduced. More importantly, a variety of promising cell sources, biocompatible tissue engineered scaffolds, scaffoldless techniques, growth factors, and mechanical stimuli used in current articular cartilage tissue engineering are reviewed. Finally, the technical and regulatory challenges of articular cartilage tissue engineering and possible future directions are also discussed.
Collapse
Affiliation(s)
- Lijie Zhang
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Jerry Hu
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| | - Kyriacos A. Athanasiou
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA 95616, USA
| |
Collapse
|