1
|
Bednarczyk E. Chondrocytes In Vitro Systems Allowing Study of OA. Int J Mol Sci 2022; 23:ijms231810308. [PMID: 36142224 PMCID: PMC9499487 DOI: 10.3390/ijms231810308] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/17/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoarthritis (OA) is an extremely complex disease, as it combines both biological-chemical and mechanical aspects, and it also involves the entire joint consisting of various types of tissues, including cartilage and bone. This paper describes the methods of conducting cell cultures aimed at searching for the mechanical causes of OA development, therapeutic solutions, and methods of preventing the disease. It presents the systems for the cultivation of cartilage cells depending on the level of their structural complexity, and taking into account the most common solutions aimed at recreating the most important factors contributing to the development of OA, that is mechanical loads. In-vitro systems used in tissue engineering to investigate the phenomena associated with OA were specified depending on the complexity and purposefulness of conducting cell cultures.
Collapse
Affiliation(s)
- Ewa Bednarczyk
- Faculty of Mechanical and Industrial Engineering, Warsaw University of Technology, Narbutta 85, 02-524 Warsaw, Poland
| |
Collapse
|
2
|
Merkely G, Ackermann J, Gomoll AH. The Role of Hypertension in Cartilage Restoration: Increased Failure Rate After Autologous Chondrocyte Implantation but Not After Osteochondral Allograft Transplantation. Cartilage 2021; 13:1306S-1314S. [PMID: 31965812 PMCID: PMC8808780 DOI: 10.1177/1947603519900792] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Objectives. The purpose of this study was to examine whether patients with diagnosed hypertension have an increased risk of graft failure following cartilage repair with either autologous chondrocyte implantation (ACI) or osteochondral allograft transplantation (OCA). We hypothesized that hypertension is related to higher ACI and OCA graft failure. Design. Patients who underwent ACI or OCA transplantation between February 2009 and December 2016 were included in this study. Inclusion criteria were (1) at least 2 years' follow-up, (2) available information related to the living habits (smoking and medication status), and (3) available information related to the presence of hypertension, diabetes mellitus, or hyperlipidemia. To identify potential independent risk factors of graft failure, univariate screening was performed and factors with significance at a level of P < 0.1 were entered in multivariate logistic regression models. Results. A total of 368 patients (209 ACI and 159 OCA) were included into our study. In the ACI group, 61 patients' (29.1%) graft failed. Univariate screening identified older age, female gender, defect size, higher prevalence of hypertension, and smoking as a predictor of graft failure. Following, multivariate logistic regression revealed female gender (odds ratio [OR] 1.02, P = 0.048), defect size (OR 1.07, P = 0.035), and hypertension (OR 3.73, P = 0.023) as significant independent risk factors predicting graft failure after ACI. In the OCA group, 29 patients' (18.2%) graft failed and none of the included factors demonstrated to be a potential risk factor for graft failure. Conclusion. Hypertension, defect size, and female gender seem to predict ACI graft failure but not OCA failure.
Collapse
Affiliation(s)
- Gergo Merkely
- Cartilage Repair Center, Brigham and
Women’s Hospital, Harvard Medical School, Boston, MA, USA,Department of Traumatology, Semmelweis
University, Budapest, Hungary,Gergo Merkely, Cartilage Repair Center,
Brigham and Women’s Hospital, Harvard Medical School, 850 Boylston Steet # 112,
Chestnut Hill, Boston, MA 02467, USA.
| | - Jakob Ackermann
- Sports Medicine Center, Department of
Orthopaedic Surgery, Massachusetts General Hospital, Boston, MA, USA,Balgrist University Hospital, Zurich,
Switzerland
| | | |
Collapse
|
3
|
Jenei-Lanzl Z, Meurer A, Zaucke F. Interleukin-1β signaling in osteoarthritis - chondrocytes in focus. Cell Signal 2018; 53:212-223. [PMID: 30312659 DOI: 10.1016/j.cellsig.2018.10.005] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 10/07/2018] [Accepted: 10/08/2018] [Indexed: 12/20/2022]
Abstract
Osteoarthritis (OA) can be regarded as a chronic, painful and degenerative disease that affects all tissues of a joint and one of the major endpoints being loss of articular cartilage. In most cases, OA is associated with a variable degree of synovial inflammation. A variety of different cell types including chondrocytes, synovial fibroblasts, adipocytes, osteoblasts and osteoclasts as well as stem and immune cells are involved in catabolic and inflammatory processes but also in attempts to counteract the cartilage loss. At the molecular level, these changes are regulated by a complex network of proteolytic enzymes, chemokines and cytokines (for review: [1]). Here, interleukin-1 signaling (IL-1) plays a central role and its effects on the different cell types involved in OA are discussed in this review with a special focus on the chondrocyte.
Collapse
Affiliation(s)
- Zsuzsa Jenei-Lanzl
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim, Frankfurt/Main, Germany
| | - Andrea Meurer
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim, Frankfurt/Main, Germany
| | - Frank Zaucke
- Dr. Rolf M. Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim, Frankfurt/Main, Germany.
| |
Collapse
|
4
|
Praxenthaler H, Krämer E, Weisser M, Hecht N, Fischer J, Grossner T, Richter W. Extracellular matrix content and WNT/β-catenin levels of cartilage determine the chondrocyte response to compressive load. Biochim Biophys Acta Mol Basis Dis 2017; 1864:851-859. [PMID: 29277327 DOI: 10.1016/j.bbadis.2017.12.024] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/15/2017] [Accepted: 12/17/2017] [Indexed: 11/19/2022]
Abstract
During osteoarthritis (OA)-development extracellular matrix (ECM) molecules are lost from cartilage, thus changing gene-expression, matrix synthesis and biomechanical competence of the tissue. Mechanical loading is important for the maintenance of articular cartilage; however, the influence of an altered ECM content on the response of chondrocytes to loading is not well understood, but may provide important insights into underlying mechanisms as well as supplying new therapies for OA. Objective here was to explore whether a changing ECM-content of engineered cartilage affects major signaling pathways and how this alters the chondrocyte response to compressive loading. Activity of canonical WNT-, BMP-, TGF-β- and p38-signaling was determined during maturation of human engineered cartilage and followed after exposure to a single dynamic compression-episode. WNT/β-catenin- and pSmad1/5/9-levels declined with increasing ECM-content of cartilage. While loading significantly suppressed proteoglycan-synthesis and ACAN-expression at low ECM-content this catabolic response then shifted to an anabolic reaction at high ECM-content. A positive correlation was observed between GAG-content and load-induced alteration of proteoglycan-synthesis. Induction of high β-catenin levels by the WNT-agonist CHIR suppressed load-induced SOX9- and GAG-stimulation in mature constructs. In contrast, the WNT-antagonist IWP-2 was capable of attenuating load-induced GAG-suppression in immature constructs. In conclusion, either ECM accumulation-associated or pharmacologically induced silencing of WNT-levels allowed for a more anabolic reaction of chondrocytes to physiological loading. This is consistent with the role of proteoglycans in sequestering WNT-ligands in the ECM, thus reducing WNT-activity and also provides a novel explanation of why low WNT-activity in cartilage protects from OA-development in mechanically overstressed cartilage.
Collapse
Affiliation(s)
- Heiko Praxenthaler
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Elisabeth Krämer
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Melanie Weisser
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Nicole Hecht
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Jennifer Fischer
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Tobias Grossner
- Department of Orthopaedic and Trauma Surgery, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Wiltrud Richter
- Research Centre for Experimental Orthopaedics, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
5
|
Scholtes S, Krämer E, Weisser M, Roth W, Luginbühl R, Grossner T, Richter W. Global chondrocyte gene expression after a single anabolic loading period: Time evolution and re-inducibility of mechano-responses. J Cell Physiol 2017; 233:699-711. [PMID: 28369921 DOI: 10.1002/jcp.25933] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 03/24/2017] [Indexed: 11/09/2022]
Abstract
Aim of this study was a genome-wide identification of mechano-regulated genes and candidate pathways in human chondrocytes subjected to a single anabolic loading episode and characterization of time evolution and re-inducibility of the response. Osteochondral constructs consisting of a chondrocyte-seeded collagen-scaffold connected to β-tricalcium-phosphate were pre-cultured for 35 days and subjected to dynamic compression (25% strain, 1 Hz, 9 × 10 min over 3 hr) before microarray-profiling was performed. Proteoglycan synthesis was determined by 35 S-sulfate-incorporation over 24 hr. Cell viability and hardness of constructs were unaltered by dynamic compression while proteoglycan synthesis was significantly stimulated (1.45-fold, p = 0.016). Among 115 significantly regulated genes, 114 were up-regulated, 48 of them ≥ twofold. AP-1-relevant transcription factors FOSB and FOS strongly increased in line with elevated ERK1/2-phosphorylation and rising MAP3K4 expression. Expression of proteoglycan-synthesizing enzymes CHSY1 and GALNT4 was load-responsive as were factors associated with the MAPK-, TGF-β-, calcium-, retinoic-acid-, Wnt-, and Notch-signaling pathway which were significantly upregulated SOX9, and BMP6 levels rose significantly also after multiple loading episodes at daily intervals even at the 14th cycle with no indication for desensitation. Canonical pSmad2/3 and pSmad1/5/9-signaling showed no consistent regulation. This study associates novel genes with mechanoregulation in chondrocytes, raising SOX9 protein levels with anabolic loading and suggests that more pathways than so far anticipated apparently work together in a complex network of stimulators and feedback-regulators. Upregulation of mechanosensitive indicators extending differentially into the resting time provides crucial knowledge to maximize cartilage matrix deposition for the generation of high-level cartilage replacement tissue.
Collapse
Affiliation(s)
- Simone Scholtes
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | - Elisabeth Krämer
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | - Melanie Weisser
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | - Wolfgang Roth
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| | | | - Tobias Grossner
- Department of Orthopaedic and Trauma Surgery, Orthopaedic University Hospital Heidelberg, Heidelberg, Germany
| | - Wiltrud Richter
- Orthopaedic University Hospital Heidelberg, Research Centre for Experimental Orthopaedics, Heidelberg, Germany
| |
Collapse
|
6
|
Boswell BA, Korol A, West-Mays JA, Musil LS. Dual function of TGFβ in lens epithelial cell fate: implications for secondary cataract. Mol Biol Cell 2017; 28:907-921. [PMID: 28209733 PMCID: PMC5385940 DOI: 10.1091/mbc.e16-12-0865] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/06/2017] [Accepted: 02/07/2017] [Indexed: 12/29/2022] Open
Abstract
The most common vision-disrupting complication of cataract surgery is posterior capsule opacification (PCO; secondary cataract). PCO is caused by residual lens cells undergoing one of two very different cell fates: either transdifferentiating into myofibroblasts or maturing into lens fiber cells. Although TGFβ has been strongly implicated in lens cell fibrosis, the factors responsible for the latter process have not been identified. We show here for the first time that TGFβ can induce purified primary lens epithelial cells within the same culture to undergo differentiation into either lens fiber cells or myofibroblasts. Marker analysis confirmed that the two cell phenotypes were mutually exclusive. Blocking the p38 kinase pathway, either with direct inhibitors of the p38 MAP kinase or a small-molecule therapeutic that also inhibits the activation of p38, prevented TGFβ from inducing epithelial-myofibroblast transition and cell migration but did not prevent fiber cell differentiation. Rapamycin had the converse effect, linking MTOR signaling to induction of fiber cell differentiation by TGFβ. In addition to providing novel potential therapeutic strategies for PCO, our findings extend the so-called TGFβ paradox, in which TGFβ can induce two disparate cell fates, to a new epithelial disease state.
Collapse
Affiliation(s)
- Bruce A Boswell
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239
| | - Anna Korol
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Hamilton, ON L8N 3Z5, Canada
| | - Judith A West-Mays
- Department of Pathology and Molecular Medicine, McMaster University Health Science Centre, Hamilton, ON L8N 3Z5, Canada
| | - Linda S Musil
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, Portland, OR 97239
| |
Collapse
|
7
|
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]
|
8
|
Ageing is associated with reduction of mechanically-induced activation of Smad2/3P signaling in articular cartilage. Osteoarthritis Cartilage 2016; 24:146-57. [PMID: 26247611 DOI: 10.1016/j.joca.2015.07.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 07/06/2015] [Accepted: 07/27/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Mechanical signals control key cellular processes in articular cartilage. Previously we have shown that mechanical compression is an important ALK5/Smad2/3P activator in cartilage explants. However, age-related changes in the cartilage are known to affect tissue mechanosensitivity and also ALK5/Smad2/3P signaling. We have investigated whether ageing of cartilage is associated with an altered response to mechanical compression. DESIGN Articular cartilage explants of two different age groups (young-6-36 months old, aged-6 - 13 years old) were subjected to dynamic mechanical compression with 3 MPa (physiological) or 12 MPa (excessive) load. Subsequently, essential cartilage extracellular matrix (ECM) components and tissue growth factors gene expression was measured in young and aged cartilage by QPCR. Furthermore, the ability of young and aged cartilage, to activate the Smad2/3P signaling in response to compression was analyzed and compared. This was done by immunohistochemical (IH) Smad2P detection and Smad3-responsive gene expression analysis. RESULTS Aged cartilage showed a highly reduced capacity for mechanically-mediated activation of Smad2/3P signaling when compared to young cartilage. Compression of aged cartilage, induced collagen type II (Col2a1) and fibronectin (Fn1) expression to a far lesser extent than in young cartilage. Additionally, in aged cartilage no mechanically mediated up-regulation of bone morphogenetic protein 2 (Bmp2) and connective tissue growth factor (Ctgf) was observed. CONCLUSIONS We identified age-related changes in cellular responses to mechanical stimulation of articular cartilage. We propose that these changes might be associated with age-related alterations in cartilage functioning and can underlie mechanisms for development of age-related cartilage diseases like osteoarthritis (OA).
Collapse
|
9
|
Tan AR, VandenBerg CD, Attur M, Abramson SB, Knight MM, Bulinski JC, Ateshian GA, Cook JL, Hung CT. Cytokine preconditioning of engineered cartilage provides protection against interleukin-1 insult. Arthritis Res Ther 2015; 17:361. [PMID: 26667364 PMCID: PMC4704536 DOI: 10.1186/s13075-015-0876-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/26/2015] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND During osteoarthritis and following surgical procedures, the environment of the knee is rich in proinflammatory cytokines such as IL-1. Introduction of tissue-engineered cartilage constructs to a chemically harsh milieu may limit the functionality of the implanted tissue over long periods. A chemical preconditioning scheme (application of low doses of IL-1) was tested as a method to prepare developing engineered tissue to withstand exposure to a higher concentration of the cytokine, known to elicit proteolysis as well as rapid degeneration of cartilage. METHODS Using an established juvenile bovine model system, engineered cartilage was preconditioned with low doses of IL-1α (0.1 ng/mL, 0.5 ng/mL, and 1.0 ng/mL) for 7 days before exposure to an insult dose (10 ng/mL). The time frame over which this protection is afforded was investigated by altering the amount of time between preconditioning and insult as well as the time following insult. To explore a potential mechanism for this protection, one set of constructs was preconditioned with CoCl2, a chemical inducer of hypoxia, before exposure to the IL-1α insult. Finally, we examined the translation of this preconditioning method to extend to clinically relevant adult, passaged chondrocytes from a preclinical canine model. RESULTS Low doses of IL-1α (0.1 ng/mL and 0.5 ng/mL) protected against subsequent catabolic degradation by cytokine insult, preserving mechanical stiffness and biochemical composition. Regardless of amount of time between preconditioning scheme and insult, protection was afforded. In a similar manner, preconditioning with CoCl2 similarly allowed for mediation of catabolic damage by IL-1α. The effects of preconditioning on clinically relevant adult, passaged chondrocytes from a preclinical canine model followed the same trends with low-dose IL-1β offering variable protection against insult. CONCLUSIONS Chemical preconditioning schemes have the ability to protect engineered cartilage constructs from IL-1-induced catabolic degradation, offering potential modalities for therapeutic treatments.
Collapse
Affiliation(s)
- Andrea R Tan
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, USA.
| | - Curtis D VandenBerg
- Department of Orthopedic Surgery, St. Luke's-Roosevelt Hospital Center, 1000 10th Avenue, New York, NY, USA.
| | - Mukundan Attur
- New York University Hospital for Joint Disease, 301 E. 17th Street, New York, NY, USA.
| | - Steven B Abramson
- New York University Hospital for Joint Disease, 301 E. 17th Street, New York, NY, USA.
| | - Martin M Knight
- Institute of Bioengineering and School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, London, UK.
| | - J Chloe Bulinski
- Department of Biological Sciences, Columbia University, 1212 Amsterdam Avenue, New York, NY, USA.
| | - Gerard A Ateshian
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, USA.
- Department of Mechanical Engineering, Columbia University, 500 W. 120th Street, New York, NY, USA.
| | - James L Cook
- Comparative Orthopaedic Laboratory, University of Missouri, 1100 Virginia Avenue, Columbia, MO, USA.
| | - Clark T Hung
- Department of Biomedical Engineering, Columbia University, 1210 Amsterdam Avenue, New York, NY, USA.
| |
Collapse
|
10
|
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
|
11
|
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
|
12
|
Ren K, Ma Y, Huang Y, Liang W, Liu F, Wang Q, Cui W, Liu Z, Yin G, Fan W. Periodic mechanical stress activates MEK1/2-ERK1/2 mitogenic signals in rat chondrocytes through Src and PLCγ1. Braz J Med Biol Res 2011; 44:1231-42. [DOI: 10.1590/s0100-879x2011007500150] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 10/25/2011] [Indexed: 12/18/2022] Open
Affiliation(s)
| | - Yimin Ma
- Nanjing Medical University, China
| | | | | | - Feng Liu
- Nanjing Medical University, China
| | | | | | | | | | | |
Collapse
|
13
|
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
|
14
|
Lee DA, Brand J, Salter D, Akanji OO, Chowdhury TT. Quantification of mRNA using real-time PCR and Western blot analysis of MAPK events in chondrocyte/agarose constructs. Methods Mol Biol 2011; 695:77-97. [PMID: 21042967 DOI: 10.1007/978-1-60761-984-0_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
In vitro models of chondrocyte mechanobiology have been used to compare the intracellular signalling pathways altered in normal and osteoarthritis-affected cartilage. However, differences in the model system and type of loading configuration have led to complicated pathways. This chapter is a follow-on of previous studies from our group utilising 3D agarose as a physiological model to study mechanotransduction pathways. Experimental methods are described to assess targets at the protein and gene expression level by Western blot analysis and real-time PCR, respectively. This chapter provides a quantitative gene expression approach to explore the intracellular pathways activated by both mechanical loading and inflammatory mediators and examine upstream phosphorylation events. Ultimately, development of methods used to analyse mechano-sensitive pathways will provide important information for the identification of appropriate pharmacological and physiotherapeutic agents for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- David A Lee
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | | | | | | | | |
Collapse
|
15
|
Green ME, Goforth PB, Satin LS, Love BJ. An integrated instrument for rapidly deforming living cells using rapid pressure pulses and simultaneously monitoring applied strain in near real time. THE REVIEW OF SCIENTIFIC INSTRUMENTS 2010; 81:125102. [PMID: 21198046 PMCID: PMC3017568 DOI: 10.1063/1.3520135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2010] [Accepted: 11/03/2010] [Indexed: 05/30/2023]
Abstract
Because many types of living cells are sensitive to applied strain, different in vitro models have been designed to elucidate the cellular and subcellular processes that respond to mechanical deformation at both the cell and tissue level. Our focus was to improve upon an already established strain system to make it capable of independently monitoring the deflection and applied pressure delivered to specific wells of a commercially available, deformable multiwell culture plate. To accomplish this, we devised a custom frame that was capable of mounting deformable 6 or 24 well plates, a pressurization system that could load wells within the plates, and a camera-based imaging system which was capable of capturing strain responses at a sufficiently high frame rate. The system used a user defined program constructed in Labview(®) to trigger plate pressurization while simultaneously allowing the deflection of the silicone elastomeric plate bottoms to be imaged in near real time. With this system, up to six wells could be pulsed simultaneously using compressed air or nitrogen. Digital image capture allowed near-real time monitoring of applied strain, strain rate, and the cell loading profiles. Although our ultimate goal is to determine how different strain rates applied to neurons modulates their intrinsic biochemical cascades, the same platform technology could be readily applied to other systems. Combining commercially available, deformable multiwell plates with a simple instrument having the monitoring capabilities described here should permit near real time calculations of stretch-induced membrane strain in multiple wells in real time for a wide variety of applications, including high throughput drug screening.
Collapse
Affiliation(s)
- M E Green
- Department of Materials Science and Engineering, The University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | |
Collapse
|
16
|
Nong L, Yin G, Ren K, Tang J, Fan W. Periodic mechanical stress enhances rat chondrocyte area expansion and migration through Src-PLCγ1-ERK1/2 signaling. Eur J Cell Biol 2010; 89:705-11. [DOI: 10.1016/j.ejcb.2010.04.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 04/20/2010] [Accepted: 04/20/2010] [Indexed: 10/19/2022] Open
|
17
|
Perera PM, Wypasek E, Madhavan S, Rath-Deschner B, Liu J, Nam J, Rath B, Huang Y, Deschner J, Piesco N, Wu C, Agarwal S. Mechanical signals control SOX-9, VEGF, and c-Myc expression and cell proliferation during inflammation via integrin-linked kinase, B-Raf, and ERK1/2-dependent signaling in articular chondrocytes. Arthritis Res Ther 2010; 12:R106. [PMID: 20509944 PMCID: PMC2911896 DOI: 10.1186/ar3039] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Revised: 03/24/2010] [Accepted: 05/28/2010] [Indexed: 12/18/2022] Open
Abstract
Introduction The importance of mechanical signals in normal and inflamed cartilage is well established. Chondrocytes respond to changes in the levels of proinflammatory cytokines and mechanical signals during inflammation. Cytokines like interleukin (IL)-1β suppress homeostatic mechanisms and inhibit cartilage repair and cell proliferation. However, matrix synthesis and chondrocyte (AC) proliferation are upregulated by the physiological levels of mechanical forces. In this study, we investigated intracellular mechanisms underlying reparative actions of mechanical signals during inflammation. Methods ACs isolated from articular cartilage were exposed to low/physiologic levels of dynamic strain in the presence of IL-1β. The cell extracts were probed for differential activation/inhibition of the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling cascade. The regulation of gene transcription was examined by real-time polymerase chain reaction. Results Mechanoactivation, but not IL-1β treatment, of ACs initiated integrin-linked kinase activation. Mechanical signals induced activation and subsequent C-Raf-mediated activation of MAP kinases (MEK1/2). However, IL-1β activated B-Raf kinase activity. Dynamic strain did not induce B-Raf activation but instead inhibited IL-1β-induced B-Raf activation. Both mechanical signals and IL-1β induced ERK1/2 phosphorylation but discrete gene expression. ERK1/2 activation by mechanical forces induced SRY-related protein-9 (SOX-9), vascular endothelial cell growth factor (VEGF), and c-Myc mRNA expression and AC proliferation. However, IL-1β did not induce SOX-9, VEGF, and c-Myc gene expression and inhibited AC cell proliferation. More importantly, SOX-9, VEGF, and Myc gene transcription and AC proliferation induced by mechanical signals were sustained in the presence of IL-1β. Conclusions The findings suggest that mechanical signals may sustain their effects in proinflammatory environments by regulating key molecules in the MAP kinase signaling cascade. Furthermore, the findings point to the potential of mechanosignaling in cartilage repair during inflammation.
Collapse
Affiliation(s)
- Priyangi M Perera
- Biomechanics and Tissue Engineering Laboratory, The Ohio State University, Postle Hall, 305 W 12th Avenue, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Ramage L, Nuki G, Salter DM. Signalling cascades in mechanotransduction: cell-matrix interactions and mechanical loading. Scand J Med Sci Sports 2009; 19:457-69. [PMID: 19538538 DOI: 10.1111/j.1600-0838.2009.00912.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mechanical loading of articular cartilage stimulates the metabolism of resident chondrocytes and induces the synthesis of molecules to maintain the integrity of the cartilage. Mechanical signals modulate biochemical activity and changes in cell behavior through mechanotransduction. Compression of cartilage results in complex changes within the tissue including matrix and cell deformation, hydrostatic and osmotic pressure, fluid flow, altered matrix water content, ion concentration and fixed charge density. These changes are detected by mechanoreceptors on the cell surface, which include mechanosensitive ion channels and integrins that on activation initiate intracellular signalling cascades leading to tissue remodelling. Excessive mechanical loading also influences chondrocyte metabolism but unlike physiological stimulation leads to a quantitative imbalance between anabolic and catabolic activity resulting in depletion of matrix components. In this article we focus on the role of mechanical signalling in the maintenance of articular cartilage, and discuss how alterations in normal signalling can lead to pathology.
Collapse
Affiliation(s)
- L Ramage
- Osteoarticular Research Group, Centre for Inflammation Research, The Queens Medical Research Institute, The University of Edinburgh, Edinburgh, UK.
| | | | | |
Collapse
|
19
|
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
|
20
|
Lima EG, Tan AR, Tai T, Bian L, Stoker AM, Ateshian GA, Cook JL, Hung CT. Differences in Interleukin-1 Response Between Engineered and Native Cartilage. Tissue Eng Part A 2008; 14:1721-30. [DOI: 10.1089/ten.tea.2007.0347] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Eric G. Lima
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Andrea R. Tan
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Timon Tai
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Liming Bian
- Department of Biomedical Engineering, Columbia University, New York, New York
| | - Aaron M. Stoker
- Comparative Orthopaedic Laboratory, University of Missouri, Columbia, Missouri
| | - Gerard A. Ateshian
- Department of Biomedical Engineering, Columbia University, New York, New York
- Department of Mechanical Engineering, Columbia University, New York, New York
| | - James L. Cook
- Comparative Orthopaedic Laboratory, University of Missouri, Columbia, Missouri
| | - Clark T. Hung
- Department of Biomedical Engineering, Columbia University, New York, New York
| |
Collapse
|
21
|
Lima EG, Tan AR, Tai T, Bian L, Ateshian GA, Cook JL, Hung CT. Physiologic deformational loading does not counteract the catabolic effects of interleukin-1 in long-term culture of chondrocyte-seeded agarose constructs. J Biomech 2008; 41:3253-9. [PMID: 18823628 DOI: 10.1016/j.jbiomech.2008.06.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2008] [Revised: 06/06/2008] [Accepted: 06/12/2008] [Indexed: 10/21/2022]
Abstract
An interplay of mechanical and chemical factors is integral to cartilage maintenance and/or degeneration. Interleukin-1 (IL-1) is a pro-inflammatory cytokine that is present at elevated concentrations in osteoarthritic joints and initiates the rapid degradation of cartilage when cultured in vitro. Several short-term studies have suggested that applied dynamic deformational loading may have a protective effect against the catabolic actions of IL-1. In the current study, we examine whether the long-term (42 days) application of dynamic deformational loading on chondrocyte-seeded agarose constructs can mitigate these catabolic effects. Three studies were carried out using two IL-1 isoforms (IL-1alpha and IL-1beta) in chemically defined medium supplemented with a broad range of cytokine concentrations and durations. Physiologic loading was unable to counteract the long-term catabolic effects of IL-1 under any of the conditions tested, and in some cases led to further degeneration over unloaded controls.
Collapse
Affiliation(s)
- Eric G Lima
- Materials Characterization Laboratory, Cooper Union, 51 Astor Place, New York, NY 10003, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Bougault C, Paumier A, Aubert-Foucher E, Mallein-Gerin F. Molecular analysis of chondrocytes cultured in agarose in response to dynamic compression. BMC Biotechnol 2008; 8:71. [PMID: 18793425 PMCID: PMC2556324 DOI: 10.1186/1472-6750-8-71] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Accepted: 09/15/2008] [Indexed: 12/02/2022] Open
Abstract
Background Articular cartilage is exposed to high mechanical loads under normal physiological conditions and articular chondrocytes regulate the composition of cartilaginous matrix, in response to mechanical signals. However, the intracellular pathways involved in mechanotransduction are still being defined. Using the well-characterized chondrocyte/agarose model system and dynamic compression, we report protocols for preparing and characterizing constructs of murine chondrocytes and agarose, and analyzing the effect of compression on steady-state level of mRNA by RT-PCR, gene transcription by gene reporter assay, and phosphorylation state of signalling molecules by Western-blotting. The mouse model is of particular interest because of the availability of a large choice of bio-molecular tools suitable to study it, as well as genetically modified mice. Results Chondrocytes cultured in agarose for one week were surrounded by a newly synthesized pericellular matrix, as revealed by immunohistochemistry prior to compression experiments. This observation indicates that this model system is suitable to study the role of matrix molecules and trans-membrane receptors in cellular responsiveness to mechanical stress. The chondrocyte/agarose constructs were then submitted to dynamic compression with FX-4000C™ Flexercell® Compression Plus™ System (Flexcell). After clearing proteins off agarose, Western-blotting analysis showed transient activation of Mitogen-activated protein kinases (MAPK) in response to dynamic compression. After assessment by capillary electrophoresis of the quality of RNA extracted from agarose, steady-state levels of mRNA expression was measured by real time PCR. We observed an up-regulation of cFos and cJun mRNA levels as a response to compression, in accordance with the mechanosensitive character observed for these two genes in other studies using cartilage explants submitted to compression. To explore further the biological response of mouse chondrocytes to the dynamic compression at the transcriptional level, we also developed an approach for monitoring changes in gene transcription in agarose culture by using reporter promoter constructs. A decrease in promoter activity of the gene coding for type II procollagen, the most abundant protein in cartilage, was observed in response to dynamic loading. Conclusion The protocols developed here offer the possibility to perform an integrated analysis of the molecular mechanisms of mechanotransduction in chondrocytes, at the gene and protein level.
Collapse
Affiliation(s)
- Carole Bougault
- UMR 5086, CNRS, Université de Lyon, IFR 128, IBCP, Institut de Biologie et Chimie des Protéines, 7 passage du Vercors F-69367 Lyon FRANCE.
| | | | | | | |
Collapse
|
23
|
Dynamic compression counteracts IL-1beta induced inducible nitric oxide synthase and cyclo-oxygenase-2 expression in chondrocyte/agarose constructs. Arthritis Res Ther 2008; 10:R35. [PMID: 18348730 PMCID: PMC2453754 DOI: 10.1186/ar2389] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Revised: 02/28/2008] [Accepted: 03/18/2008] [Indexed: 01/16/2023] Open
Abstract
Background Nitric oxide and prostaglandin E2 (PGE2play pivotal roles in both the pathogenesis of osteoarthritis and catabolic processes in articular cartilage. These mediators are influenced by both IL-1β and mechanical loading, and involve alterations in the inducible nitric oxide synthase (iNOS) and cyclo-oxygenase (COX)-2 enzymes. To identify the specific interactions that are activated by both types of stimuli, we examined the effects of dynamic compression on levels of expression of iNOS and COX-2 and involvement of the p38 mitogen-activated protein kinase (MAPK) pathway. Methods Chondrocyte/agarose constructs were cultured under free-swelling conditions with or without IL-1β and/or SB203580 (inhibitor of p38 MAPK) for up to 48 hours. Using a fully characterized bioreactor system, constructs were subjected to dynamic compression for 6, 12 and 48 hours under similar treatments. The activation or inhibition of p38 MAPK by IL-1β and/or SB203580 was analyzed by western blotting. iNOS, COX-2, aggrecan and collagen type II signals were assessed utilizing real-time quantitative PCR coupled with molecular beacons. Release of nitrite and PGE2 was quantified using biochemical assays. Two-way analysis of variance and the post hoc Bonferroni-corrected t-test were used to examine data. Results IL-1β activated the phosphorylation of p38 MAPK and this effect was abolished by SB203580. IL-1β induced a transient increase in iNOS expression and stimulated the production of nitrite release. Stimulation by either dynamic compression or SB203580 in isolation reduced the IL-1β induced iNOS expression and nitrite production. However, co-stimulation with both dynamic compression and SB203580 inhibited the expression levels of iNOS and production of nitrite induced by the cytokine. IL-1β induced a transient increase in COX-2 expression and stimulated the cumulative production of PGE2 release. These effects were inhibited by dynamic compression or SB203580. Co-stimulation with both dynamic compression and SB203580 restored cytokine-induced inhibition of aggrecan expression. This is in contrast to collagen type II, in which we observed no response with the cytokine and/or SB203580. Conclusion These data suggest that dynamic compression directly influences the expression levels of iNOS and COX-2. These molecules are current targets for pharmacological intervention, raising the possibility for integrated pharmacological and biophysical therapies for the treatment of cartilage joint disorders.
Collapse
|
24
|
Gilbert SJ, Blain EJ, Duance VC, Mason DJ. Sphingomyelinase decreases type II collagen expression in bovine articular cartilage chondrocytes via the ERK signaling pathway. ACTA ACUST UNITED AC 2008; 58:209-20. [PMID: 18163502 DOI: 10.1002/art.23172] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE Ceramide, a mediator of proinflammatory cytokine signaling, induces cartilage degradation and reduces type II collagen synthesis in articular cartilage. The accumulation of ceramide is associated with arthritis in Farber's disease. The aim of this study was to investigate the mechanism of ceramide-induced down-regulation of type II collagen. METHODS Bovine articular chondrocytes were stimulated with sphingomyelinase (SMase) to increase levels of endogenous ceramide. Components of the ERK pathway were inhibited by Raf-1 kinase inhibitor and the MEK inhibitor, PD98059. Cell extracts were analyzed by Western blotting for ERK-1/2, SOX9, c-Fos, and type II collagen, and the level of c-fos messenger RNA (mRNA) was analyzed by quantitative polymerase chain reaction. Localization of ERK-1/2, SOX9, and c-Fos was assessed by immunocytochemistry and confocal microscopy. RESULTS SMase treatment of chondrocytes caused sustained phosphorylation of ERK-1/2 throughout the cytoplasm and nucleus that was reduced by inhibitors of Raf-1 kinase and MEK-1/2. SMase treatment of chondrocytes also induced translocation of c-Fos to the nucleus and phospho-SOX9 to the cytoplasm and increased expression of c-fos mRNA. Type II collagen expression, which was down-regulated by SMase treatment, was restored by the MEK-1/2 inhibitor, PD98059. CONCLUSION SMase down-regulates type II collagen in articular chondrocytes via activation of the ERK signaling cascade, redistribution of SOX9, and recruitment of c-Fos. This new mechanism for cartilage degradation provides potential targets for future treatment of arthritic disease.
Collapse
|
25
|
Fitzgerald JB, Jin M, Chai DH, Siparsky P, Fanning P, Grodzinsky AJ. Shear- and Compression-induced Chondrocyte Transcription Requires MAPK Activation in Cartilage Explants. J Biol Chem 2008; 283:6735-43. [DOI: 10.1074/jbc.m708670200] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
26
|
De Croos JNA, Jang B, Dhaliwal SS, Grynpas MD, Pilliar RM, Kandel RA. Membrane type-1 matrix metalloproteinase is induced following cyclic compression of in vitro grown bovine chondrocytes. Osteoarthritis Cartilage 2007; 15:1301-10. [PMID: 17548215 DOI: 10.1016/j.joca.2007.04.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Accepted: 04/15/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To determine if membrane type-1 matrix metalloproteinase (MT1-MMP) will respond to cyclic compression of chondrocytes grown in vitro and the regulatory mechanisms underlying this response. METHODS Cyclic compression (30min, 1kPa, 1Hz) was applied to bovine chondrocytes (6-9-month-old animals) grown on top of a biodegradable substrate within 3 days of initiating culture. Luciferase assays using bovine articular chondrocytes were undertaken to demonstrate the mechanosensitivity of MT1-MMP. Semi-quantitative reverse-transcription polymerase chain reaction (RT-PCR) and western blot analysis were used to establish the time course of gene and protein upregulation in response to cyclic compression. The regulation of MT1-MMP was assessed by electrophoretic mobility shift assays, RT-PCR and western blot analysis. As well, an MT1-MMP decoy oligonucleotide and an extracellular signal-regulated kinase 1/2 (ERK1/2) pharmacological inhibitor were utilized to further characterize MT1-MMP regulation. RESULTS After cyclic compression, MT1-MMP showed a rapid and transient increase in gene expression. Elevated protein levels were detected within 2h of stimulation which returned to baseline by 6h. During cyclic compression, phosphorylation of the mitogen activated protein kinase ERK1/2 increased significantly. This was followed by increased gene and protein expression of the transcription factor; early growth factor-1 (Egr-1) and Egr-1 binding to the MT1-MMP promoter. Blocking Egr-1 DNA binding with a decoy MT1-MMP oligonucleotide, downregulated MT1-MMP gene expression. The ERK1/2 inhibitor U0126 also reduced Egr-1 DNA binding activity to MT1-MMP promoter sequences and subsequent transcription of MT1-MMP. CONCLUSIONS These data suggest that cyclic compression of chondrocytes in vitro upregulates MT1-MMP via ERK1/2 dependent activation of Egr-1 binding. Delineation of the regulatory pathways activated by mechanical stimulation will further our understating of the mechanisms influencing tissue remodeling.
Collapse
Affiliation(s)
- J N A De Croos
- CIHR BioEngineering of Skeletal Tissues Team, Mount Sinai Hospital, Canada
| | | | | | | | | | | |
Collapse
|
27
|
Bueno EM, Laevsky G, Barabino GA. Enhancing cell seeding of scaffolds in tissue engineering through manipulation of hydrodynamic parameters. J Biotechnol 2007; 129:516-31. [PMID: 17324484 DOI: 10.1016/j.jbiotec.2007.01.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2006] [Revised: 12/01/2006] [Accepted: 01/03/2007] [Indexed: 11/30/2022]
Abstract
The seeding of cells onto biocompatible scaffolds is a determinant step in the attainment of functional properties of engineered tissues. Efficient, fast and spatially uniform cell seeding can improve the clinical potential of engineered tissue templates. One way to approach these cell seeding requirements is through bioreactor design. In the present study, bovine chondrocytes were seeded (2.5, 5.0 or 10.0 million cells per scaffold) onto polyglycolic acid scaffolds within the hydrodynamic environments of wavy-walled and spinner flask bioreactors. Previous characterizations of the hydrodynamic environment in the vicinity of constructs cultivated in these bioreactors suggested decreased flow-induced shear stress as well as increased recirculation and magnitude of the axial fluid velocities in the wavy-walled bioreactor. Here we report more efficient and spatially uniform cell seeding in the wavy-walled bioreactor, and at intermediate initial cell densities (5 million cells per scaffold). This study constitutes an important step towards the achievement of functional tissue-engineered implants by (i) increasing our understanding of the influence of hydrodynamic parameters on the efficiency and spatial distribution of cell attachment to scaffolds and the production of extracellular matrix and (ii) introducing a comprehensive approach to the investigation of the effects of bioprocessing conditions on tissue morphology and composition.
Collapse
Affiliation(s)
- Ericka M Bueno
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 342 Snell Engineering, Boston, MA 02115, USA.
| | | | | |
Collapse
|
28
|
Aufderheide AC, Athanasiou KA. A Direct Compression Stimulator for Articular Cartilage and Meniscal Explants. Ann Biomed Eng 2006; 34:1463-74. [PMID: 16897420 DOI: 10.1007/s10439-006-9157-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2006] [Accepted: 06/22/2006] [Indexed: 11/29/2022]
Abstract
This paper describes the development and use of a direct compression stimulator for culturing explants from the meniscus of the knee and articular cartilage. Following design and fabrication of the instrument along with its data acquisition system, the function of the machine was verified by both mechanical means and tissue effect. The loading chamber can hold up to 45 5 mm diameter samples. While designed to stimulate samples up to 4 mm thick, axial displacements as little as 0.127 microm are within the theoretical capacity of the stimulator. In gene expression studies, collagen II and aggrecan expression were examined in explants from articular cartilage as well as medial and lateral menisci subjected to dynamic stimulation and static compression. These results were then compared to free swelling samples. It was found that static compression to cut thickness down-regulated aggrecan and collagen II expression in articular cartilage explants compared to free swelling controls by 94% and 90%, respectively. The application of a dynamic, intermittent, 2% oscillation around the cut thickness returned expression levels to those of free swelling controls at 4 h but not at 76 h. In medial meniscus samples, dynamic compression up-regulated aggrecan expression by 108%, but not collagen II expression, at 4 and 76 h compared to static controls. No difference in gene expression was observed for lateral meniscal explants. Thus, effects of direct compression seen in articular cartilage may not necessarily translate to the knee meniscus. The design of this stimulator will allow a variety of tissues and loading regimens to be examined. It is hoped that regimens can be found that not only return samples to the production levels of free swelling controls, but also surpass them in terms of gene expression, protein synthesis, and functional properties.
Collapse
Affiliation(s)
- Adam C Aufderheide
- Department of Bioengineering, Rice University, Houston, Texas 77251, USA
| | | |
Collapse
|
29
|
Fitzgerald JB, Jin M, Grodzinsky AJ. Shear and Compression Differentially Regulate Clusters of Functionally Related Temporal Transcription Patterns in Cartilage Tissue. J Biol Chem 2006; 281:24095-103. [PMID: 16782710 DOI: 10.1074/jbc.m510858200] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chondrocytes are subjected to a variety of biophysical forces and flows during physiological joint loading, including mechanical deformation, fluid flow, hydrostatic pressure, and streaming potentials; however, the role of these physical stimuli in regulating chondrocyte behavior is still being elucidated. To isolate the effects of these forces, we subjected intact cartilage explants to 1-24 h of continuous dynamic compression or dynamic shear loading at 0.1 Hz. We then measured the transcription levels of 25 genes known to be involved in cartilage homeostasis using real-time PCR and compared the gene expression profiles obtained from dynamic compression, dynamic shear, and our recent results on static compression amplitude and duration. Using clustering analysis, we determined that transcripts for proteins with similar function had correlated responses to loading. However, the temporal expression patterns were strongly dependent on the type of loading applied. Most matrix proteins were up-regulated by 24 h of dynamic compression or dynamic shear, but down-regulated by 24 h of 50% static compression, suggesting that cyclic matrix deformation is a key stimulator of matrix protein expression. Most matrix proteases were up-regulated by 24 h under all loading types. Transcription factors c-Fos and c-Jun maximally responded within 1 h to all loading types. Pre-incubating cartilage explants with either a chelator of intracellular calcium or an inhibitor of the cyclic AMP pathway demonstrated the involvement of both pathways in transcription induced by dynamic loading.
Collapse
Affiliation(s)
- Jonathan B Fitzgerald
- Biological Engineering Division, Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
30
|
Ferretti M, Madhavan S, Deschner J, Rath-Deschner B, Wypasek E, Agarwal S. Dynamic biophysical strain modulates proinflammatory gene induction in meniscal fibrochondrocytes. Am J Physiol Cell Physiol 2006; 290:C1610-5. [PMID: 16452158 PMCID: PMC4950929 DOI: 10.1152/ajpcell.00529.2005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fibrochondrocytes of meniscus adapt to changes in their biomechanical environment by mechanisms that are yet to be elucidated. In this study, the mechanoresponsiveness of fibrochondrocytes under normal and inflammatory conditions was investigated. Fibrochondrocytes from rat meniscus were exposed to dynamic tensile forces (DTF) at various magnitudes and frequencies. The mechanoresponsiveness was assessed by examining the expression of inducible nitric oxide synthase (iNOS), tumor necrosis factor-alpha (TNF-alpha), and matrix metalloproteinase-13 mRNA expression. The mRNA and protein analyses revealed that DTF at magnitudes of 5% to 20% did not induce proinflammatory gene expression. IL-1beta induced a rapid increase in the iNOS mRNA. DTF strongly repressed IL-1beta-dependent iNOS induction in a magnitude-dependent manner. Exposure to 15% DTF resulted in >90% suppression of IL-1beta-induced mRNA within 4 h and this suppression was sustained for the ensuing 20 h. The mechanosensitivity of fibrochondrocytes was also frequency dependent and maximal suppression of iNOS mRNA expression was observed at rapid frequencies of DTF compared with lower frequencies. Like iNOS, DTF also inhibited IL-1beta-induced expression of proinflammatory mediators involved in joint inflammation. The examination of temporal effects of DTF revealed that 4- or 8-h exposure of DTF was sufficient for its sustained anti-inflammatory effects during the next 20 or 16 h, respectively. Our findings indicate that mechanical signals act as potent anti-inflammatory signals, where their magnitude and frequency are critical determinants of their actions. Furthermore, mechanical signals continue attenuating proinflammatory gene transcription for prolonged periods of time after their removal.
Collapse
Affiliation(s)
- Mario Ferretti
- Biomechanics and Tissue Engineering Laboratory, The Ohio State Univ., 305 West 12th Ave., Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
31
|
Kisiday JD, Kurz B, DiMicco MA, Grodzinsky AJ. Evaluation of medium supplemented with insulin-transferrin-selenium for culture of primary bovine calf chondrocytes in three-dimensional hydrogel scaffolds. ACTA ACUST UNITED AC 2005; 11:141-51. [PMID: 15738669 DOI: 10.1089/ten.2005.11.141] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Insulin-transferrin-selenium (ITS) was investigated as a complete or partial replacement for fetal bovine serum (FBS) during in vitro culture of bovine calf chondrocytes in hydrogel scaffolds. Chondrocyte-seeded agarose and self-assembling peptide hydrogels were maintained in Dulbecco's modified Eagle's medium plus 10% FBS, 1% ITS plus 0.2% FBS, or 1% ITS and evaluated for biosynthesis, cell division, and surface outgrowth of fibroblastic-like cells and fibrous capsule formation over several weeks of culture. In peptide hydrogels, cells cultured in ITS plus 0.2% FBS medium exhibited high rates of biosynthesis and showed similar cell division trends as seen in 10% FBS cultures. ITS medium alone did not support glycosaminoglycan accumulation beyond 5 days of culture, and cell division was less than that in both serum-containing cultures. Extensive cellular outgrowth and fibrous capsule formation were observed in 10% FBS medium, whereas little outgrowth was observed in ITS plus 0.2% FBS and none was seen in ITS medium alone. In agarose hydrogels, chondrocyte biosynthesis and cell division in ITS medium were similar to that in 10% serum culture over 5 weeks, and cellular outgrowth was eliminated. Taken together, ITS was suitable as a partial (peptide) or complete (agarose) substitute for serum, and also provided the benefit of reducing or eliminating cell outgrowth and fibrous capsule formation on the hydrogel surface.
Collapse
Affiliation(s)
- John D Kisiday
- Biological Engineering Division, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | |
Collapse
|
32
|
Dai SM, Shan ZZ, Nishioka K, Yudoh K. Implication of interleukin 18 in production of matrix metalloproteinases in articular chondrocytes in arthritis: direct effect on chondrocytes may not be pivotal. Ann Rheum Dis 2005; 64:735-42. [PMID: 15834055 PMCID: PMC1755478 DOI: 10.1136/ard.2004.026088] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To clarify the effect of interleukin (IL) 18 on cartilage degeneration by studying the profile of IL18 receptor (IL18R) on chondrocytes and the direct effect of IL18 on production of matrix metalloproteinases (MMPs), aggrecanases, and tissue inhibitors of metalloproteinases (TIMPs) in articular chondrocytes. METHODS Monolayer cultured human articular chondrocytes were isolated from non-arthritic subjects and patients with rheumatoid arthritis or osteoarthritis. Gene expression of IL18, IL18Ralpha, IL18Rbeta, MMPs, and aggrecanases was detected by RT-PCR. Protein levels of IL18Ralpha were analysed by flow cytometry. Protein levels of IL18, MMPs, and TIMPs were measured by ELISA. Aggrecanase-2 mRNA expression was quantitatively analysed by real time RT-PCR. Protein levels of signalling molecules were assayed by western blotting. RESULTS IL18 mRNA was constitutively expressed in chondrocytes, and was enhanced by IL1beta stimulation. Flow cytometric analysis showed that IL1beta, tumour necrosis factor alpha, and IL18 up regulated IL18Ralpha expression levels. The level of IL18Rbeta mRNA was much lower than that of IL18Ralpha, and was slightly up regulated by IL1beta. In chondrocytes responding to IL18, IL18 (1-100 ng/ml) slightly increased the production of MMP-1, MMP-3, and MMP-13, which was blocked by NF-kappaB inhibitor and p38 mitogen activated protein kinase inhibitor. IL18 up regulated mRNA expression of aggrecanase-2, but not aggrecanase-1. IL18 also slightly stimulated TIMP-1 production?through extracellular signal regulated kinase activation. CONCLUSION IL18 induces production of MMPs from chondrocytes in inflammatory arthritis. Although the direct effect of IL18 on chondrocytes may not be pivotal for the induction of cartilage degeneration, IL18 seems to play some part in the degradation of articular cartilage in arthritis.
Collapse
Affiliation(s)
- S-M Dai
- Department of Bioregulation, Institute of Medical Science, St Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa 216-8512, Japan
| | | | | | | |
Collapse
|
33
|
Onodera K, Takahashi I, Sasano Y, Bae JW, Mitani H, Kagayama M, Mitani H. Stepwise mechanical stretching inhibits chondrogenesis through cell-matrix adhesion mediated by integrins in embryonic rat limb-bud mesenchymal cells. Eur J Cell Biol 2005; 84:45-58. [PMID: 15724815 DOI: 10.1016/j.ejcb.2004.09.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Biomechanical forces are major epigenetic factors that determine the form and differentiation of skeletal tissues, and may be transduced through cell adhesion to the intracellular biochemical signaling pathway. To test the hypothesis that stepwise stretching is translated to molecular signals during early chondrogenesis, we developed a culture system to study the proliferation and differentiation of chondrocytes. Rat embryonic day-12 limb buds were microdissected and dissociated into cells, which were then micromass cultured on a silicone membrane and maintained for up to 7 days. Stepwise-increased stretching was applied to the silicone membrane, which exerted shearing stress on the cultures on day 4 after the initiation of chondrogenesis. Under stretched conditions, type II collagen expression was significantly inhibited by 44% on day 1 and by 67% on day 2, and this difference in type II collagen reached 80% after 3 days of culture. Accumulation of type II collagen protein and the size of the chondrogenic nodules had decreased by 50% on day 3. On the other hand, expression of the non-chondrogenic marker fibronectin was significantly upregulated by 1.8-fold on day 3, while the up-regulation of type I collagen was minimal, even by day 3. The downregulation in the expression of chondrogenic markers was completely recovered when cell-extracellular matrix attachment was inhibited by Gly-Arg-Gly-Asp-Ser-Pro-Lys peptide or by the application of blocking antibodies for alpha2, alpha5 or beta1 integrins. We conclude that shearing stress generated by stepwise stretching inhibits chondrogenesis through integrins, and propose that signal transduction from biomechanical stimuli may be mediated by cell-extracellular matrix adhesion.
Collapse
Affiliation(s)
- Kazuyuki Onodera
- Tohoku University Graduate School of Dentistry, Aoba-ku 980-8575, Sendai, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Szafranski JD, Grodzinsky AJ, Burger E, Gaschen V, Hung HH, Hunziker EB. Chondrocyte mechanotransduction: effects of compression on deformation of intracellular organelles and relevance to cellular biosynthesis. Osteoarthritis Cartilage 2004; 12:937-46. [PMID: 15564060 DOI: 10.1016/j.joca.2004.08.004] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2004] [Accepted: 08/11/2004] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The effects of mechanical deformation of intact cartilage tissue on chondrocyte biosynthesis in situ have been well documented, but the mechanotransduction pathways that regulate such phenomena have not been elucidated completely. The goal of this study was to examine the effects of tissue deformation on the morphology of a range of intracellular organelles which play a major role in cell biosynthesis and metabolism. DESIGN Using chemical fixation, high pressure freezing, and electron microscopy, we imaged chondrocytes within mechanically compressed cartilage explants at high magnification and quantitatively and qualitatively assessed changes in organelle volume and shape caused by graded levels of loading. RESULTS Compression of the tissue caused a concomitant reduction in the volume of the extracellular matrix (ECM), chondrocyte, nucleus, rough endoplasmic reticulum, and mitochondria. Interestingly, however, the Golgi apparatus was able to resist loss of intraorganelle water and retain a portion of its volume relative to the remainder of the cell. These combined results suggest that a balance between intracellular mechanical and osmotic gradients govern the changes in shape and volume of the organelles as the tissue is compressed. CONCLUSIONS Our results lead to the interpretive hypothesis that organelle volume changes appear to be driven mainly by osmotic interactions while shape changes are mediated by structural factors, such as cytoskeletal interactions that may be linked to extracellular matrix deformations. The observed volume and shape changes of the chondrocyte organelles and the differential behavior between organelles during tissue compression provide evidence for an important mechanotransduction pathway linking translational and post-translational events (e.g., elongation and sulfation of glycosaminoglycans (GAGs) in the Golgi) to cell deformation.
Collapse
Affiliation(s)
- Jon D Szafranski
- Biological Engineering Division and Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | | | | | | | | |
Collapse
|
35
|
Schmidt TA, Schumacher BL, Klein TJ, Voegtline MS, Sah RL. Synthesis of proteoglycan 4 by chondrocyte subpopulations in cartilage explants, monolayer cultures, and resurfaced cartilage cultures. ACTA ACUST UNITED AC 2004; 50:2849-57. [PMID: 15457453 DOI: 10.1002/art.20480] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To quantify the levels of proteoglycan 4 (PRG4) expression by subpopulations of chondrocytes from superficial, middle, and deep layers of normal bovine calf cartilage in various culture systems. METHODS Bovine calf articular cartilage discs or isolated cells were used in 1 of 3 systems of chondrocyte culture: explant, monolayer, or transplant, for 1-9 days. PRG4 expression was quantified by enzyme-linked immunosorbent assay of spent medium and localized by immunohistochemistry at the articular surface and within chondrocytes in explants and cultured cells. RESULTS Superficial chondrocytes secreted much more PRG4 than did middle and deep chondrocytes in all cultures. The pattern of PRG4 secretion into superficial culture medium varied with the duration of culture, decreasing with time in explant culture (from approximately 25 microg/cm(2)/day on days 0-1 to approximately 3 microg/cm(2)/day on days 5-9), while increasing in monolayer culture (from approximately 1 pg/cell/day on days 0-1 to approximately 7 pg/cell/day on days 7-9) and tending to increase in transplant culture (reaching approximately 2 microg/cm(2)/day by days 7-9). In all of the culture systems, inclusion of ascorbic acid stimulated PRG4 secretion, and the source of PRG4 was immunolocalized to superficial cells. CONCLUSION The results described here indicate that the phenotype of PRG4 secretion by chondrocytes in culture is generally maintained, in that PRG4 is expressed to a much greater degree by chondrocytes from the superficial zone than by those from the middle and deep zones. The marked up-regulation of PRG4 synthesis by ascorbic acid may have implications for cartilage homeostasis and prevention of osteoarthritic disease. Transplanting specialized cells that secrete PRG4 to a surface may impart functional lubrication and be generally applicable to many tissues in the body.
Collapse
Affiliation(s)
- Tannin A Schmidt
- Whitaker Institute of Biomedical Engineering, University of California, San Diego, CA 92093, USA
| | | | | | | | | |
Collapse
|
36
|
Grümmer R, Hewitt SW, Traub O, Korach KS, Winterhager E. Different Regulatory Pathways of Endometrial Connexin Expression: Preimplantation Hormonal-Mediated Pathway Versus Embryo Implantation-Initiated Pathway1. Biol Reprod 2004; 71:273-81. [PMID: 15028626 DOI: 10.1095/biolreprod.103.024067] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Transformation of the endometrium into the receptive phase is under the control of ovarian steroid hormones and is modulated by embryonic signals during implantation. We have previously shown that this differentiation process is accompanied by a suppression of gap junction connexins (Cx) 26 and 43 before implantation followed by a local induction of both connexins in the implantation chamber. In the present study, we demonstrate that connexin gene expression in the rodent endometrium is regulated via two distinct signaling pathways during these different stages of early pregnancy. During preimplantation, transcription of connexins can be induced by estrogen via an estrogen receptor (ER)-dependent pathway. Additionally, Cx26 and Cx43 are induced by embryonic signals during implantation and delayed implantation as well as during artificially induced decidualization. In contrast to the estrogen-induced expression, this embryonic/decidual-associated induction of Cx26 and Cx43 could not be blocked by antiestrogen, thus pointing to another regulatory pathway independent of the ER. Studies in ERalpha and ERbeta knockout mice confirmed these different pathways, demonstrating that in the endometrium, estrogen-mediated Cx26 gene induction, but not induction during decidualization, is dependent on functional ERalpha. To evaluate potential embryonic signals regulating Cx26 expression, uteri of pseudopregnant animals were incubated with different mediators in an organ-culture model, showing that catechol estrogen and mediators of the inflammatory cascade such as prostaglandin F(2alpha) and interleukin-1beta are able to induce Cx26 expression through the ER-independent pathway. Thus, the present study demonstrates that endometrial expression of Cx26 and Cx43 is induced via estrogen and ERalpha during preimplantation but then utilizes an ER-independent signaling pathway during embryo implantation and decidualization.
Collapse
Affiliation(s)
- R Grümmer
- Institute of Anatomy, University Hospital, 45122 Essen, Germany.
| | | | | | | | | |
Collapse
|
37
|
Fitzgerald JB, Jin M, Dean D, Wood DJ, Zheng MH, Grodzinsky AJ. Mechanical Compression of Cartilage Explants Induces Multiple Time-dependent Gene Expression Patterns and Involves Intracellular Calcium and Cyclic AMP. J Biol Chem 2004; 279:19502-11. [PMID: 14960571 DOI: 10.1074/jbc.m400437200] [Citation(s) in RCA: 173] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Chondrocytes are influenced by mechanical forces to remodel cartilage extracellular matrix. Previous studies have demonstrated the effects of mechanical forces on changes in biosynthesis and mRNA levels of particular extracellular matrix molecules, and have identified certain signaling pathways that may be involved. However, the broad extent and kinetics of mechano-regulation of gene transcription has not been studied in depth. We applied static compressive strains to bovine cartilage explants for periods between 1 and 24 h and measured the response of 28 genes using real time PCR. Compression time courses were also performed in the presence of an intracellular calcium chelator or an inhibitor of cyclic AMP-activated protein kinase A. Cluster analysis of the data revealed four main expression patterns: two groups containing either transiently up-regulated or duration-enhanced expression profiles could each be subdivided into genes that did or did not require intracellular calcium release and cyclic AMP-activated protein kinase A for their mechano-regulation. Transcription levels for aggrecan, type II collagen, and link protein were up-regulated approximately 2-3-fold during the first 8 h of 50% compression and subsequently down-regulated to levels below that of free-swelling controls by 24 h. Transcription levels of matrix metalloproteinases-3, -9, and -13, aggrecanase-1, and the matrix protease regulator cyclooxygenase-2 increased with the duration of 50% compression 2-16-fold by 24 h. Thus, transcription of proteins involved in matrix remodeling and catabolism dominated over anabolic matrix proteins as the duration of static compression increased. Immediate early genes c-fos and c-jun were dramatically up-regulated 6-30-fold, respectively, during the first 8 h of 50% compression and remained up-regulated after 24 h.
Collapse
Affiliation(s)
- Jonathan B Fitzgerald
- Biological Engineering Division, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | | | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- William L Lowe
- Division of Endocrinology, Metabolism, and Molecular Medicine, Northwestern University, Feinberg School of Medicine, Tarry 15-703, 303 East Chicago Avenue, Chicago, Illinois 60611, USA.
| |
Collapse
|
39
|
|