1
|
De Gaudemaris I, Hannoun A, Gauthier R, Attik N, Brizuela L, Mebarek S, Hassler M, Bougault C, Trunfio-Sfarghiu AM. Positive impact of pyrocarbon and mechanical loading on cartilage-like tissue synthesis in a scaffold-free process. J Biosci Bioeng 2025; 139:53-59. [PMID: 39395870 DOI: 10.1016/j.jbiosc.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/14/2024]
Abstract
Aiming to build a tissue analogue engineered cartilage from differentiated chondrocytes, we investigated the potential of a pyrocarbon (PyC)-based and scaffold-free process, under mechanical stimulation. PyC biomaterial has shown promise in arthroplasty and implant strategies, and mechanical stimulation is recognized as an improvement in regeneration strategies. The objective was to maintain the cell phenotype to produce constructs with cartilage-like matrix composition and mechanical properties. Primary murine chondrocytes were deposited in drop form between two biomaterial surfaces expanded to 500 μm and a uniaxial cyclic compression was applied thanks to a handmade tribo-bioreactor (0.5 Hz, 100 μm of amplitude, 17 days). Histology and immunohistochemistry analysis showed that PyC biomaterial promoted expression of cartilage-like matrix components (glycosaminoglycans, type II collagen, aggrecan). Importantly, constructs obtained in dynamic conditions were denser and showed a cohesive and compact shape. The most promising condition was the combined use of PyC and dynamic stimulation, resulting in constructs of low elasticity and high viscosity, thus with an increased damping factor. We verified that no calcium deposits were detectable and that type X collagen was not expressed, suggesting that the cells had not undergone hypertrophic maturation. While most studies focus on the comparison of different biomaterials or on the effect of different mechanical stimuli separately, we demonstrated the value of combining the two approaches to get as close as possible to the biological and mechanical qualities of natural hyaline articular cartilage.
Collapse
Affiliation(s)
| | - Amira Hannoun
- Univ Lyon, CNRS, INSA Lyon, UMR5259, LaMCoS, F-69621, Villeurbanne, France
| | - Rémy Gauthier
- Univ Lyon, CNRS, INSA Lyon, UCBL, UMR5510, MATEIS, F-69621, Villeurbanne, France
| | - Nina Attik
- Universite Claude Bernard Lyon 1, CNRS UMR5615, LMI, F-69622, Lyon, France; Universite Claude Bernard Lyon 1, Faculté d'odontologie, F-69372, Lyon, France
| | - Leyre Brizuela
- Universite Claude Bernard Lyon 1, CNRS UMR5246, ICBMS, F-69622, Lyon, France
| | - Saida Mebarek
- Universite Claude Bernard Lyon 1, CNRS UMR5246, ICBMS, F-69622, Lyon, France
| | - Michel Hassler
- Tornier SAS, 161 rue Lavoisier, F-38330, Montbonnot Saint-Martin, France
| | - Carole Bougault
- Universite Claude Bernard Lyon 1, CNRS UMR5246, ICBMS, F-69622, Lyon, France.
| | | |
Collapse
|
2
|
Ponce A, Ogazon del Toro A, Jimenez L, Roldan ML, Shoshani L. Osmotically Sensitive TREK Channels in Rat Articular Chondrocytes: Expression and Functional Role. Int J Mol Sci 2024; 25:7848. [PMID: 39063089 PMCID: PMC11277475 DOI: 10.3390/ijms25147848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Articular chondrocytes are the primary cells responsible for maintaining the integrity and functionality of articular cartilage, which is essential for smooth joint movement. A key aspect of their role involves mechanosensitive ion channels, which allow chondrocytes to detect and respond to mechanical forces encountered during joint activity; nonetheless, the variety of mechanosensitive ion channels involved in this process has not been fully resolved so far. Because some members of the two-pore domain potassium (K2P) channel family have been described as mechanosensors in other cell types, in this study, we investigate whether articular chondrocytes express such channels. RT-PCR analysis reveals the presence of TREK-1 and TREK-2 channels in these cells. Subsequent protein expression assessments, including Western blotting and immunohistochemistry, confirm the presence of TREK-1 in articular cartilage samples. Furthermore, whole-cell patch clamp assays demonstrate that freshly isolated chondrocytes exhibit currents attributable to TREK-1 channels, as evidenced by activation by arachidonic acid (AA) and ml335 and further inhibition by spadin. Additionally, exposure to hypo-osmolar shock activates currents, which can be attributed to the presence of TREK-1 channels, as indicated by their inhibition with spadin. Therefore, these findings highlight the expression of TREK channels in rat articular chondrocytes and suggest their potential involvement in regulating the integrity of cartilage extracellular matrix.
Collapse
Affiliation(s)
- Arturo Ponce
- Department of Physiology, Biophysics and Neurosciences, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Ciudad de México 07360, Mexico; (A.O.d.T.); (L.J.); (M.L.R.); (L.S.)
| | | | | | | | | |
Collapse
|
3
|
Urdeitx P, Mousavi SJ, Avril S, Doweidar MH. Computational modeling of multiple myeloma interactions with resident bone marrow cells. Comput Biol Med 2023; 153:106458. [PMID: 36599211 DOI: 10.1016/j.compbiomed.2022.106458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/08/2022] [Accepted: 12/19/2022] [Indexed: 12/27/2022]
Abstract
The interaction of multiple myeloma with bone marrow resident cells plays a key role in tumor progression and the development of drug resistance. The tumor cell response involves contact-mediated and paracrine interactions. The heterogeneity of myeloma cells and bone marrow cells makes it difficult to reproduce this environment in in-vitro experiments. The use of in-silico established tools can help to understand these complex problems. In this article, we present a computational model based on the finite element method to define the interactions of multiple myeloma cells with resident bone marrow cells. This model includes cell migration, which is controlled by stress-strain equilibrium, and cell processes such as proliferation, differentiation, and apoptosis. A series of computational experiments were performed to validate the proposed model. Cell proliferation by the growth factor IGF-1 is studied for different concentrations ranging from 0-10 ng/mL. Cell motility is studied for different concentrations of VEGF and fibronectin in the range of 0-100 ng/mL. Finally, cells were simulated under a combination of IGF-1 and VEGF stimuli whose concentrations are considered to be dependent on the cancer-associated fibroblasts in the extracellular matrix. Results show a good agreement with previous in-vitro results. Multiple myeloma growth and migration are shown to correlate linearly to the IGF-1 stimuli. These stimuli are coupled with the mechanical environment, which also improves cell growth. Moreover, cell migration depends on the fiber and VEGF concentration in the extracellular matrix. Finally, our computational model shows myeloma cells trigger mesenchymal stem cells to differentiate into cancer-associated fibroblasts, in a dose-dependent manner.
Collapse
Affiliation(s)
- Pau Urdeitx
- School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, 50018, Spain; Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, 50018, Spain
| | - S Jamaleddin Mousavi
- Mines Saint-Étienne, University of Lyon, University of Jean Monnet, INSERM, Saint-Etienne, 42023, France
| | - Stephane Avril
- Mines Saint-Étienne, University of Lyon, University of Jean Monnet, INSERM, Saint-Etienne, 42023, France; Institute for Mechanics of Materials and Structures, TU Wien-Vienna University of Technology, Vienna, 1040, Austria
| | - Mohamed H Doweidar
- School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, 50018, Spain; Aragon Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, 50018, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, 50018, Spain.
| |
Collapse
|
4
|
Murphy P, Rolfe RA. Building a Co-ordinated Musculoskeletal System: The Plasticity of the Developing Skeleton in Response to Muscle Contractions. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:81-110. [PMID: 37955772 DOI: 10.1007/978-3-031-38215-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The skeletal musculature and the cartilage, bone and other connective tissues of the skeleton are intimately co-ordinated. The shape, size and structure of each bone in the body is sculpted through dynamic physical stimuli generated by muscle contraction, from early development, with onset of the first embryo movements, and through repair and remodelling in later life. The importance of muscle movement during development is shown by congenital abnormalities where infants that experience reduced movement in the uterus present a sequence of skeletal issues including temporary brittle bones and joint dysplasia. A variety of animal models, utilising different immobilisation scenarios, have demonstrated the precise timing and events that are dependent on mechanical stimulation from movement. This chapter lays out the evidence for skeletal system dependence on muscle movement, gleaned largely from mouse and chick immobilised embryos, showing the many aspects of skeletal development affected. Effects are seen in joint development, ossification, the size and shape of skeletal rudiments and tendons, including compromised mechanical function. The enormous plasticity of the skeletal system in response to muscle contraction is a key factor in building a responsive, functional system. Insights from this work have implications for our understanding of morphological evolution, particularly the challenging concept of emergence of new structures. It is also providing insight for the potential of physical therapy for infants suffering the effects of reduced uterine movement and is enhancing our understanding of the cellular and molecular mechanisms involved in skeletal tissue differentiation, with potential for informing regenerative therapies.
Collapse
Affiliation(s)
- Paula Murphy
- School of Natural Sciences, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland.
| | - Rebecca A Rolfe
- School of Natural Sciences, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| |
Collapse
|
5
|
Poillot P, Snuggs JW, Le Maitre CL, Huyghe JM. L-type Voltage-Gated calcium channels partly mediate Mechanotransduction in the intervertebral disc. JOR Spine 2022; 5:e1213. [PMID: 36601377 PMCID: PMC9799080 DOI: 10.1002/jsp2.1213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/18/2022] [Accepted: 06/09/2022] [Indexed: 01/07/2023] Open
Abstract
Background Intervertebral disc (IVD) degeneration continues to be a major global health challenge, with strong links to lower back pain, while the pathogenesis of this disease is poorly understood. In cartilage, much more is known about mechanotransduction pathways involving the strain-generated potential (SGP) and function of voltage-gated ion channels (VGICs) in health and disease. This evidence implicates a similar important role for VGICs in IVD matrix turnover. However, the field of VGICs, and to a lesser extent the SGP, remains unexplored in the IVD. Methods A two-step process was utilized to investigate the role of VGICs in the IVD. First, immunohistochemical staining was used to identify and localize several different VGICs in bovine and human IVDs. Second, a pilot study was conducted on the function of L-type voltage gated calcium channels (VGCCs) by inhibiting these channels with nifedipine (Nf) and measuring calcium influx in monolayer or gene expression from 3D cell-embedded alginate constructs subject to dynamic compression. Results Several VGICs were identified at the protein level, one of which, Cav2.2, appears to be upregulated with the onset of human IVD degeneration. Inhibiting L-type VGCCs with Nf supplementation led to an altered cell calcium influx in response to osmotic loading as well as downregulation of col 1a, aggrecan and ADAMTS-4 during dynamic compression. Conclusions This study demonstrates the presence of several VGICs in the IVD, with evidence supporting a role for L-type VGCCs in mechanotransduction. These findings highlight the importance of future detailed studies in this area to fully elucidate IVD mechanotransduction pathways and better inform treatment strategies for IVD degeneration.
Collapse
Affiliation(s)
| | - Joseph W. Snuggs
- Biomolecular Sciences Research CentreSheffield Hallam UniversitySheffieldUK
| | | | - Jacques M. Huyghe
- Bernal InstituteUniversity of LimerickLimerickIreland
- Department of Mechanical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
| |
Collapse
|
6
|
Osteogenesis Imperfecta/Ehlers-Danlos Overlap Syndrome and Neuroblastoma-Case Report and Review of Literature. Genes (Basel) 2022; 13:genes13040581. [PMID: 35456387 PMCID: PMC9024599 DOI: 10.3390/genes13040581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 11/21/2022] Open
Abstract
Osteogenesis imperfecta/Ehlers−Danlos (OI/EDS) overlap syndrome is a recently described disorder of connective tissue, characterized by mutation of COL1A1 (17q21.33) or COL1A2 (7q21.3) genes, that are involved in α-1 and α-2 chains of type 1 collagen synthesis. The clinical spectrum of this new clinical entity is broad: patients could present a mixed phenotype that includes features of both osteogenesis imperfecta (bone fragility, long bone fractures, blue sclerae, short stature) and Ehlers−Danlos syndrome (joint hyperextensibility, soft and hyperextensible skin, abnormal wound healing, easy bruising, vascular fragility). We reported the case of a young Caucasian girl with severe short stature and a previous history of neuroblastoma, who displayed the compound phenotype of OI/EDS. Next generation sequencing was applied to the proband and her parent genome. Our patient presented a de novo heterozygous COL1A1 variant (c.3235G>A, p.Gly1079Ser), whose presence might be indicative of diagnosis of OI/EDS overlap syndrome. We also hypothesize that the association with the previous history of neuroblastoma could be influenced by the presence of COL1A1 mutation, whose role has been already described in the behavior and progression of some cancers.
Collapse
|
7
|
Retrograde Analysis of Calcium Signaling by CaMPARI2 Shows Cytosolic Calcium in Chondrocytes Is Unaffected by Parabolic Flights. Biomedicines 2022; 10:biomedicines10010138. [PMID: 35052817 PMCID: PMC8773224 DOI: 10.3390/biomedicines10010138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 02/04/2023] Open
Abstract
Calcium (Ca2+) elevation is an essential secondary messenger in many cellular processes, including disease progression and adaptation to external stimuli, e.g., gravitational load. Therefore, mapping and quantifying Ca2+ signaling with a high spatiotemporal resolution is a key challenge. However, particularly on microgravity platforms, experiment time is limited, allowing only a small number of replicates. Furthermore, experiment hardware is exposed to changes in gravity levels, causing experimental artifacts unless appropriately controlled. We introduce a new experimental setup based on the fluorescent Ca2+ reporter CaMPARI2, onboard LED arrays, and subsequent microscopic analysis on the ground. This setup allows for higher throughput and accuracy due to its retrograde nature. The excellent performance of CaMPARI2 was demonstrated with human chondrocytes during the 75th ESA parabolic flight campaign. CaMPARI2 revealed a strong Ca2+ response triggered by histamine but was not affected by the alternating gravitational load of a parabolic flight.
Collapse
|
8
|
Zhang K, Wang L, Liu Z, Geng B, Teng Y, Liu X, Yi Q, Yu D, Chen X, Zhao D, Xia Y. Mechanosensory and mechanotransductive processes mediated by ion channels in articular chondrocytes: Potential therapeutic targets for osteoarthritis. Channels (Austin) 2021; 15:339-359. [PMID: 33775217 PMCID: PMC8018402 DOI: 10.1080/19336950.2021.1903184] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Articular cartilage consists of an extracellular matrix including many proteins as well as embedded chondrocytes. Articular cartilage formation and function are influenced by mechanical forces. Hind limb unloading or simulated microgravity causes articular cartilage loss, suggesting the importance of the healthy mechanical environment in articular cartilage homeostasis and implying a significant role of appropriate mechanical stimulation in articular cartilage degeneration. Mechanosensitive ion channels participate in regulating the metabolism of articular chondrocytes, including matrix protein production and extracellular matrix synthesis. Mechanical stimuli, including fluid shear stress, stretch, compression and cell swelling and decreased mechanical conditions (such as simulated microgravity) can alter the membrane potential and regulate the metabolism of articular chondrocytes via transmembrane ion channel-induced ionic fluxes. This process includes Ca2+ influx and the resulting mobilization of Ca2+ that is due to massive released Ca2+ from stores, intracellular cation efflux and extracellular cation influx. This review brings together published information on mechanosensitive ion channels, such as stretch-activated channels (SACs), voltage-gated Ca2+ channels (VGCCs), large conductance Ca2+-activated K+ channels (BKCa channels), Ca2+-activated K+ channels (SKCa channels), voltage-activated H+ channels (VAHCs), acid sensing ion channels (ASICs), transient receptor potential (TRP) family channels, and piezo1/2 channels. Data based on epithelial sodium channels (ENaCs), purinergic receptors and N-methyl-d-aspartate (NMDA) receptors are also included. These channels mediate mechanoelectrical physiological processes essential for converting physical force signals into biological signals. The primary channel-mediated effects and signaling pathways regulated by these mechanosensitive ion channels can influence the progression of osteoarthritis during the mechanosensory and mechanoadaptive process of articular chondrocytes.
Collapse
Affiliation(s)
- Kun Zhang
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Lifu Wang
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Zhongcheng Liu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Bin Geng
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Yuanjun Teng
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Xuening Liu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Qiong Yi
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Dechen Yu
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Xiangyi Chen
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Dacheng Zhao
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| | - Yayi Xia
- Department of Orthopedics, Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou Gansu, China
| |
Collapse
|
9
|
Dieterle MP, Husari A, Rolauffs B, Steinberg T, Tomakidi P. Integrins, cadherins and channels in cartilage mechanotransduction: perspectives for future regeneration strategies. Expert Rev Mol Med 2021; 23:e14. [PMID: 34702419 PMCID: PMC8724267 DOI: 10.1017/erm.2021.16] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023]
Abstract
Articular cartilage consists of hyaline cartilage, is a major constituent of the human musculoskeletal system and has critical functions in frictionless joint movement and articular homoeostasis. Osteoarthritis (OA) is an inflammatory disease of articular cartilage, which promotes joint degeneration. Although it affects millions of people, there are no satisfying therapies that address this disease at the molecular level. Therefore, tissue regeneration approaches aim at modifying chondrocyte biology to mitigate the consequences of OA. This requires appropriate biochemical and biophysical stimulation of cells. Regarding the latter, mechanotransduction of chondrocytes and their precursor cells has become increasingly important over the last few decades. Mechanotransduction is the transformation of external biophysical stimuli into intracellular biochemical signals, involving sensor molecules at the cell surface and intracellular signalling molecules, so-called mechano-sensors and -transducers. These signalling events determine cell behaviour. Mechanotransducing ion channels and gap junctions additionally govern chondrocyte physiology. It is of great scientific and medical interest to induce a specific cell behaviour by controlling these mechanotransduction pathways and to translate this knowledge into regenerative clinical therapies. This review therefore focuses on the mechanotransduction properties of integrins, cadherins and ion channels in cartilaginous tissues to provide perspectives for cartilage regeneration.
Collapse
Affiliation(s)
- Martin Philipp Dieterle
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106Freiburg, Germany
| | - Ayman Husari
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106Freiburg, Germany
- Department of Orthodontics, Center for Dental Medicine, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106Freiburg, Germany
| | - Bernd Rolauffs
- Department of Orthopedics and Trauma Surgery, G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Medical Center – Albert-Ludwigs-University of Freiburg, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, 79085Freiburg im Breisgau, Germany
| | - Thorsten Steinberg
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106Freiburg, Germany
| | - Pascal Tomakidi
- Division of Oral Biotechnology, Center for Dental Medicine, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106Freiburg, Germany
| |
Collapse
|
10
|
Li X, Wang Y, Cai Z, Zhou Q, Li L, Fu P. Exosomes from human umbilical cord mesenchymal stem cells inhibit ROS production and cell apoptosis in human articular chondrocytes via the miR-100-5p/NOX4 axis. Cell Biol Int 2021; 45:2096-2106. [PMID: 34197004 DOI: 10.1002/cbin.11657] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/23/2021] [Accepted: 06/17/2021] [Indexed: 11/05/2022]
Abstract
Cyclic strain-induced chondrocyte damage is actively involved in the pathogenesis of osteoarthritis and arthritis. MicroRNAs (miRNAs) carried by exosomes have been implicated in various diseases. However, the role of miR-100-5p in cyclic strain-induced chondrocyte damage remains to be elucidated. miR-100-5p and NADPH oxidase 4 (NOX4) were silenced or overexpressed in human primary articular chondrocytes. PKH-67 Dye was used to trace exosome endocytosis. Reactive oxygen species (ROS) production was monitored using DCFH-DA. Cell apoptosis was measured using a flow cytometer. Quantitative RT-PCR and Western blots were used to evaluate gene expression. Cyclic strain promoted ROS production and apoptosis in primary articular chondrocytes in a time-dependent manner. HucMSCs-derived exosomal miR-100-5p inhibited cyclic strain-induced ROS production and apoptosis in primary articular chondrocytes. miR-100-5p directly targeted NOX4. Overexpressing NOX4 attenuated hucMSCs-derived exosomes-mediated protective effects in primary articular chondrocytes. Cyclic strain promotes ROS production and apoptosis in primary articular chondrocytes, which was abolished by hucMSCs-derived exosomal miR-100-5p through its target NOX4. The findings highlight the importance of miR-100-5p/NOX4 axis in primary articular chondrocytes injury and provide new insights into therapeutic strategies for articular chondrocytes injury and osteoarthritis.
Collapse
Affiliation(s)
- Xiang Li
- Department of Joint Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuanyuan Wang
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, China
| | - Zhuyun Cai
- Department of Joint Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Qi Zhou
- Department of Joint Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Lexiang Li
- Department of Joint Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Peiliang Fu
- Department of Joint Surgery, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
11
|
Urdeitx P, Doweidar MH. Enhanced Piezoelectric Fibered Extracellular Matrix to Promote Cardiomyocyte Maturation and Tissue Formation: A 3D Computational Model. BIOLOGY 2021; 10:biology10020135. [PMID: 33572184 PMCID: PMC7914718 DOI: 10.3390/biology10020135] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 12/26/2022]
Abstract
Mechanical and electrical stimuli play a key role in tissue formation, guiding cell processes such as cell migration, differentiation, maturation, and apoptosis. Monitoring and controlling these stimuli on in vitro experiments is not straightforward due to the coupling of these different stimuli. In addition, active and reciprocal cell-cell and cell-extracellular matrix interactions are essential to be considered during formation of complex tissue such as myocardial tissue. In this sense, computational models can offer new perspectives and key information on the cell microenvironment. Thus, we present a new computational 3D model, based on the Finite Element Method, where a complex extracellular matrix with piezoelectric properties interacts with cardiac muscle cells during the first steps of tissue formation. This model includes collective behavior and cell processes such as cell migration, maturation, differentiation, proliferation, and apoptosis. The model has employed to study the initial stages of in vitro cardiac aggregate formation, considering cell-cell junctions, under different extracellular matrix configurations. Three different cases have been purposed to evaluate cell behavior in fibered, mechanically stimulated fibered, and mechanically stimulated piezoelectric fibered extra-cellular matrix. In this last case, the cells are guided by the coupling of mechanical and electrical stimuli. Accordingly, the obtained results show the formation of more elongated groups and enhancement in cell proliferation.
Collapse
Affiliation(s)
- Pau Urdeitx
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, 50018 Zaragoza, Spain;
- Aragon Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 50018 Zaragoza, Spain
| | - Mohamed H. Doweidar
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, 50018 Zaragoza, Spain;
- Aragon Institute of Engineering Research (I3A), University of Zaragoza, 50018 Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 50018 Zaragoza, Spain
- Correspondence:
| |
Collapse
|
12
|
A Computational Model for Cardiomyocytes Mechano-Electric Stimulation to Enhance Cardiac Tissue Regeneration. MATHEMATICS 2020. [DOI: 10.3390/math8111875] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Electrical and mechanical stimulations play a key role in cell biological processes, being essential in processes such as cardiac cell maturation, proliferation, migration, alignment, attachment, and organization of the contractile machinery. However, the mechanisms that trigger these processes are still elusive. The coupling of mechanical and electrical stimuli makes it difficult to abstract conclusions. In this sense, computational models can establish parametric assays with a low economic and time cost to determine the optimal conditions of in-vitro experiments. Here, a computational model has been developed, using the finite element method, to study cardiac cell maturation, proliferation, migration, alignment, and organization in 3D matrices, under mechano-electric stimulation. Different types of electric fields (continuous, pulsating, and alternating) in an intensity range of 50–350 Vm−1, and extracellular matrix with stiffnesses in the range of 10–40 kPa, are studied. In these experiments, the group’s morphology and cell orientation are compared to define the best conditions for cell culture. The obtained results are qualitatively consistent with the bibliography. The electric field orientates the cells and stimulates the formation of elongated groups. Group lengthening is observed when applying higher electric fields in lower stiffness extracellular matrix. Groups with higher aspect ratios can be obtained by electrical stimulation, with better results for alternating electric fields.
Collapse
|
13
|
Maleckar MM, Martín-Vasallo P, Giles WR, Mobasheri A. Physiological Effects of the Electrogenic Current Generated by the Na +/K + Pump in Mammalian Articular Chondrocytes. Bioelectricity 2020; 2:258-268. [PMID: 34471850 PMCID: PMC8370340 DOI: 10.1089/bioe.2020.0036] [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] [Indexed: 12/11/2022] Open
Abstract
Background: Although the chondrocyte is a nonexcitable cell, there is strong interest in gaining detailed knowledge of its ion pumps, channels, exchangers, and transporters. In combination, these transport mechanisms set the resting potential, regulate cell volume, and strongly modulate responses of the chondrocyte to endocrine agents and physicochemical alterations in the surrounding extracellular microenvironment. Materials and Methods: Mathematical modeling was used to assess the functional roles of energy-requiring active transport, the Na+/K+ pump, in chondrocytes. Results: Our findings illustrate plausible physiological roles for the Na+/K+ pump in regulating the resting membrane potential and suggest ways in which specific molecular components of pump can respond to the unique electrochemical environment of the chondrocyte. Conclusion: This analysis provides a basis for linking chondrocyte electrophysiology to metabolism and yields insights into novel ways of manipulating or regulating responsiveness to external stimuli both under baseline conditions and in chronic diseases such as osteoarthritis.
Collapse
Affiliation(s)
| | - Pablo Martín-Vasallo
- UD of Biochemistry and Molecular Biology, Universidad de La Laguna, San Cristóbal de La Laguna, Spain.,Instituto de Tecnologías Biomédicas de Canarias, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Wayne R Giles
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Canada
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.,Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania.,Department of Orthopedics, Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
14
|
K + and Ca 2+ Channels Regulate Ca 2+ Signaling in Chondrocytes: An Illustrated Review. Cells 2020; 9:cells9071577. [PMID: 32610485 PMCID: PMC7408816 DOI: 10.3390/cells9071577] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/23/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022] Open
Abstract
An improved understanding of fundamental physiological principles and progressive pathophysiological processes in human articular joints (e.g., shoulders, knees, elbows) requires detailed investigations of two principal cell types: synovial fibroblasts and chondrocytes. Our studies, done in the past 8–10 years, have used electrophysiological, Ca2+ imaging, single molecule monitoring, immunocytochemical, and molecular methods to investigate regulation of the resting membrane potential (ER) and intracellular Ca2+ levels in human chondrocytes maintained in 2-D culture. Insights from these published papers are as follows: (1) Chondrocyte preparations express a number of different ion channels that can regulate their ER. (2) Understanding the basis for ER requires knowledge of (a) the presence or absence of ligand (ATP/histamine) stimulation and (b) the extraordinary ionic composition and ionic strength of synovial fluid. (3) In our chondrocyte preparations, at least two types of Ca2+-activated K+ channels are expressed and can significantly hyperpolarize ER. (4) Accounting for changes in ER can provide insights into the functional roles of the ligand-dependent Ca2+ influx through store-operated Ca2+ channels. Some of the findings are illustrated in this review. Our summary diagram suggests that, in chondrocytes, the K+ and Ca2+ channels are linked in a positive feedback loop that can augment Ca2+ influx and therefore regulate lubricant and cytokine secretion and gene transcription.
Collapse
|
15
|
Carter RT. Reinforcement of the larynx and trachea in echolocating and non-echolocating bats. J Anat 2020; 237:495-503. [PMID: 32319086 DOI: 10.1111/joa.13204] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 11/29/2022] Open
Abstract
The synchronization of flight mechanics with respiration and echolocation call emission by bats, while economizing these behaviors, presumably puts compressive loads on the cartilaginous rings that hold open the respiratory tract. Previous work has shown that during postnatal development of Artibeus jamaicensis (Phyllostomidae), the onset of adult echolocation call emission rate coincides with calcification of the larynx, and the development of flight coincides with tracheal ring calcification. In the present study, I assessed the level of reinforcement of the respiratory system in 13 bat species representing six families that use stereotypical modes of echolocation (i.e. duty cycle % and intensity). Using computed tomography, the degree of mineralization or ossification of the tracheal rings, cricoid, thyroid and arytenoid cartilages were determined for non-echolocators, tongue clicking, low-duty cycle low-intensity, low-duty cycle high-intensity, and high-duty cycle high-intensity echolocating bats. While all bats had evidence of cervical tracheal ring mineralization, about half the species had evidence of thoracic tracheal ring calcification. Larger bats (Phyllostomus hastatus and Pterpodidae sp.) exhibited more extensive tracheal ring mineralization, suggesting an underlying cause independent of laryngeal echolocation. Within most of the laryngeally echolocating species, the degree of mineralization or ossification of the larynx was dependent on the mode of echolocation system used. Low-duty cycle low-intensity bats had extensively mineralized cricoids, and zero to very minor mineralization of the thyroids and arytenoids. Low-duty cycle high-intensity bats had extensively mineralized cricoids, and patches of thyroid and arytenoid mineralization. The high-duty cycle high-intensity rhinolophids and hipposiderid had extensively ossified cricoids, large patches of ossification on the thyroids, and heavily ossified arytenoids. The high-duty cycle high-intensity echolocator, Pteronotus parnellii, had mineralization patterns and laryngeal morphology very similar to the other low-duty cycle high-intensity mormoopid species, perhaps suggesting relatively recent evolution of high-duty cycle echolocation in P. parnellii compared with the Old World high-duty cycle echolocators (Rhinolophidae and Hipposideridae). All laryngeal echolocators exhibited mineralized or ossified lateral expansions of the cricoid for articulation with the inferior horn of the thyroid, these were most prominent in the high-duty cycle high-intensity rhinolophids and hipposiderid, and least prominent in the low-duty cycle low-intensity echolocators. The non-laryngeal echolocators had extensively ossified cricoid and thyroid cartilages, and no evidence of mineralization/ossification of the arytenoids or lateral expansions of the cricoid. While the non-echolocators had extensive ossification of the larynx, it was inconsistent with that seen in the laryngeal echolocators.
Collapse
|
16
|
URDEITX PAU, FARZANEH SOLMAZ, MOUSAVI SJAMALEDDIN, DOWEIDAR MOHAMEDH. ROLE OF OXYGEN CONCENTRATION IN THE OSTEOBLASTS BEHAVIOR: A FINITE ELEMENT MODEL. J MECH MED BIOL 2020. [DOI: 10.1142/s0219519419500647] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Oxygen concentration plays a key role in cell survival and viability. Besides, it has important effects on essential cellular biological processes such as cell migration, differentiation, proliferation and apoptosis. Therefore, the prediction of the cellular response to the alterations of the oxygen concentration can help significantly in the advances of cell culture research. Here, we present a 3D computational mechanotactic model to simulate all the previously mentioned cell processes under different oxygen concentrations. With this model, three cases have been studied. Starting with mesenchymal stem cells within an extracellular matrix with mechanical properties suitable for its differentiation into osteoblasts, and under different oxygen conditions to evaluate their behavior under normoxia, hypoxia and anoxia. The obtained results, which are consistent with the experimental observations, indicate that cells tend to migrate toward zones with higher oxygen concentration where they accelerate their differentiation and proliferation. This technique can be employed to control cell migration toward fracture zones to accelerate the healing process. Besides, as expected, to avoid cell apoptosis under conditions of anoxia and to avoid the inhibition of the differentiation and proliferation processes under conditions of hypoxia, the state of normoxia should be maintained throughout the entire cell-culture process.
Collapse
Affiliation(s)
- PAU URDEITX
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, Spain
- Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| | - SOLMAZ FARZANEH
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059, Sainbiose, Centre CIS, F - 42023, Saint-Etienne, France
| | - S. JAMALEDDIN MOUSAVI
- Mines Saint-Etienne, Univ Lyon, Univ Jean Monnet, INSERM, U 1059, Sainbiose, Centre CIS, F - 42023, Saint-Etienne, France
| | - MOHAMED H. DOWEIDAR
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, Spain
- Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| |
Collapse
|
17
|
Atsuta Y, Tomizawa RR, Levin M, Tabin CJ. L-type voltage-gated Ca 2+ channel Ca V1.2 regulates chondrogenesis during limb development. Proc Natl Acad Sci U S A 2019; 116:21592-21601. [PMID: 31591237 PMCID: PMC6815189 DOI: 10.1073/pnas.1908981116] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
All cells, including nonexcitable cells, maintain a discrete transmembrane potential (Vmem), and have the capacity to modulate Vmem and respond to their own and neighbors' changes in Vmem Spatiotemporal variations have been described in developing embryonic tissues and in some cases have been implicated in influencing developmental processes. Yet, how such changes in Vmem are converted into intracellular inputs that in turn regulate developmental gene expression and coordinate patterned tissue formation, has remained elusive. Here we document that the Vmem of limb mesenchyme switches from a hyperpolarized to depolarized state during early chondrocyte differentiation. This change in Vmem increases intracellular Ca2+ signaling through Ca2+ influx, via CaV1.2, 1 of L-type voltage-gated Ca2+ channels (VGCCs). We find that CaV1.2 activity is essential for chondrogenesis in the developing limbs. Pharmacological inhibition by an L-type VGCC specific blocker, or limb-specific deletion of CaV1.2, down-regulates expression of genes essential for chondrocyte differentiation, including Sox9, Col2a1, and Agc1, and thus disturbs proper cartilage formation. The Ca2+-dependent transcription factor NFATc1, which is a known major transducer of intracellular Ca2+ signaling, partly rescues Sox9 expression. These data reveal instructive roles of CaV1.2 in limb development, and more generally expand our understanding of how modulation of membrane potential is used as a mechanism of developmental regulation.
Collapse
Affiliation(s)
- Yuji Atsuta
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Allen Discovery Center at Tufts University, Tufts University, Medford, MA 02155
| | - Reiko R Tomizawa
- Department of Genetics, Harvard Medical School, Boston, MA 02115
- Allen Discovery Center at Tufts University, Tufts University, Medford, MA 02155
| | - Michael Levin
- Allen Discovery Center at Tufts University, Tufts University, Medford, MA 02155
- Department of Biology, Tufts University, Medford, MA 02155
| | - Clifford J Tabin
- Department of Genetics, Harvard Medical School, Boston, MA 02115;
- Allen Discovery Center at Tufts University, Tufts University, Medford, MA 02155
| |
Collapse
|
18
|
Parisi C, Chandaria VV, Nowlan NC. Blocking mechanosensitive ion channels eliminates the effects of applied mechanical loading on chick joint morphogenesis. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2017.0317. [PMID: 30249769 PMCID: PMC6158207 DOI: 10.1098/rstb.2017.0317] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2018] [Indexed: 11/12/2022] Open
Abstract
Abnormalities in joint shape are increasingly considered a critical risk factor for developing osteoarthritis in life. It has been shown that mechanical forces during prenatal development, particularly those due to fetal movements, play a fundamental role in joint morphogenesis. However, how mechanical stimuli are sensed or transduced in developing joint tissues is unclear. Stretch-activated and voltage-gated calcium ion channels have been shown to be involved in the mechanoregulation of chondrocytes in vitro. In this study, we analyse, for the first time, how blocking these ion channels influences the effects of mechanical loading on chick joint morphogenesis. Using in vitro culture of embryonic chick hindlimb explants in a mechanostimulation bioreactor, we block stretch-activated and voltage-gated ion channels using, respectively, gadolinium chloride and nifedipine. We find that the administration of high doses of either drug largely removed the effects of mechanical stimulation on growth and shape development in vitro, while neither drug had any effect in static cultures. This study demonstrates that, during joint morphogenesis, mechanical cues are transduced—at least in part—through mechanosensitive calcium ion channels, advancing our understanding of cartilage development and mechanotransduction. This article is part of the Theo Murphy meeting issue ‘Mechanics of development’.
Collapse
Affiliation(s)
- Cristian Parisi
- Department of Bioengineering, Faculty of Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Vikesh V Chandaria
- Department of Bioengineering, Faculty of Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| | - Niamh C Nowlan
- Department of Bioengineering, Faculty of Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK
| |
Collapse
|
19
|
Maleckar MM, Clark RB, Votta B, Giles WR. The Resting Potential and K + Currents in Primary Human Articular Chondrocytes. Front Physiol 2018; 9:974. [PMID: 30233381 PMCID: PMC6131720 DOI: 10.3389/fphys.2018.00974] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/03/2018] [Indexed: 11/23/2022] Open
Abstract
Human transplant programs provide significant opportunities for detailed in vitro assessments of physiological properties of selected tissues and cell types. We present a semi-quantitative study of the fundamental electrophysiological/biophysical characteristics of human chondrocytes, focused on K+ transport mechanisms, and their ability to regulate to the resting membrane potential, Em. Patch clamp studies on these enzymatically isolated human chondrocytes reveal consistent expression of at least three functionally distinct K+ currents, as well as transient receptor potential (TRP) currents. The small size of these cells and their exceptionally low current densities present significant technical challenges for electrophysiological recordings. These limitations have been addressed by parallel development of a mathematical model of these K+ and TRP channel ion transfer mechanisms in an attempt to reveal their contributions to Em. In combination, these experimental results and simulations yield new insights into: (i) the ionic basis for Em and its expected range of values; (ii) modulation of Em by the unique articular joint extracellular milieu; (iii) some aspects of TRP channel mediated depolarization-secretion coupling; (iv) some of the essential biophysical principles that regulate K+ channel function in “chondrons.” The chondron denotes the chondrocyte and its immediate extracellular compartment. The presence of discrete localized surface charges and associated zeta potentials at the chondrocyte surface are regulated by cell metabolism and can modulate interactions of chondrocytes with the extracellular matrix. Semi-quantitative analysis of these factors in chondrocyte/chondron function may yield insights into progressive osteoarthritis.
Collapse
Affiliation(s)
- Mary M Maleckar
- Simula Research Laboratory, Center for Biomedical Computing and Center for Cardiological Innovation, Oslo, Norway.,Allen Institute for Cell Science, Seattle, WA, United States
| | - Robert B Clark
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
| | | | - Wayne R Giles
- Faculties of Kinesiology and Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
20
|
Chen CH, Kuo CY, Chen JP. Effect of Cyclic Dynamic Compressive Loading on Chondrocytes and Adipose-Derived Stem Cells Co-Cultured in Highly Elastic Cryogel Scaffolds. Int J Mol Sci 2018; 19:370. [PMID: 29373507 PMCID: PMC5855592 DOI: 10.3390/ijms19020370] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/20/2018] [Accepted: 01/22/2018] [Indexed: 12/23/2022] Open
Abstract
In this study, we first used gelatin/chondroitin-6-sulfate/hyaluronan/chitosan highly elastic cryogels, which showed total recovery from large strains during repeated compression cycles, as 3D scaffolds to study the effects of cyclic dynamic compressive loading on chondrocyte gene expression and extracellular matrix (ECM) production. Dynamic culture of porcine chondrocytes was studied at 1 Hz, 10% to 40% strain and 1 to 9 h/day stimulation duration, in a mechanical-driven multi-chamber bioreactor for 14 days. From the experimental results, we could identify the optimum dynamic culture condition (20% and 3 h/day) to enhance the chondrocytic phenotype of chondrocytes from the expression of marker (Col I, Col II, Col X, TNF-α, TGF-β1 and IGF-1) genes by quantitative real-time polymerase chain reactions (qRT-PCR) and production of ECM (GAGs and Col II) by biochemical analysis and immunofluorescence staining. With up-regulated growth factor (TGF-β1 and IGF-1) genes, co-culture of chondrocytes with porcine adipose-derived stem cells (ASCs) was employed to facilitate chondrogenic differentiation of ASCs during dynamic culture in cryogel scaffolds. By replacing half of the chondrocytes with ASCs during co-culture, we could obtain similar production of ECM (GAGs and Col II) and expression of Col II, but reduced expression of Col I, Col X and TNF-α. Subcutaneous implantation of cells/scaffold constructs in nude mice after mono-culture (chondrocytes or ASCs) or co-culture (chondrocytes + ASCs) and subject to static or dynamic culture condition in vitro for 14 days was tested for tissue-engineering applications. The constructs were retrieved 8 weeks post-implantation for histological analysis by Alcian blue, Safranin O and Col II immunohistochemical staining. The most abundant ectopic cartilage tissue was found for the chondrocytes and chondrocytes + ASCs groups using dynamic culture, which showed similar neo-cartilage formation capability with half of the chondrocytes replaced by ASCs for co-culture. This combined co-culture/dynamic culture strategy is expected to cut down the amount of donor chondrocytes needed for cartilage-tissue engineering.
Collapse
Affiliation(s)
- Chih-Hao Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan.
| | - Chang-Yi Kuo
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan.
| | - Jyh-Ping Chen
- Department of Chemical and Materials Engineering, Chang Gung University, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Plastic and Reconstructive Surgery and Craniofacial Research Center, Chang Gung Memorial Hospital, Linkou, Chang Gung University School of Medicine, Kwei-San, Taoyuan 33305, Taiwan.
- Research Center for Chinese Herbal Medicine, Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-San, Taoyuan 33302, Taiwan.
- Department of Materials Engineering, Ming Chi University of Technology, Tai-Shan, New Taipei City 24301, Taiwan.
| |
Collapse
|
21
|
Abstract
During embryogenesis, the musculoskeletal system develops while containing within itself a force generator in the form of the musculature. This generator becomes functional relatively early in development, exerting an increasing mechanical load on neighboring tissues as development proceeds. A growing body of evidence indicates that such mechanical forces can be translated into signals that combine with the genetic program of organogenesis. This unique situation presents both a major challenge and an opportunity to the other tissues of the musculoskeletal system, namely bones, joints, tendons, ligaments and the tissues connecting them. Here, we summarize the involvement of muscle-induced mechanical forces in the development of various vertebrate musculoskeletal components and their integration into one functional unit.
Collapse
Affiliation(s)
- Neta Felsenthal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Elazar Zelzer
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
22
|
Teo A, Morshedi A, Wang JC, Zhou Y, Lim M. Enhancement of Cardiomyogenesis in Murine Stem Cells by Low-Intensity Ultrasound. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2017; 36:1693-1706. [PMID: 28439945 DOI: 10.7863/ultra.16.12042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 10/19/2016] [Indexed: 05/15/2023]
Abstract
OBJECTIVES Low-intensity ultrasound (LIUS) has been shown to enhance bone and cartilage regeneration from stem cells. The ease of its incorporation makes it an attractive mechanical stimulus for not only osteogenesis and chondrogenesis, but also cardiomyogenesis. However, to date, no study has investigated its effects on cardiomyogenesis from embryonic stem cells. METHODS In this study, murine embryonic stem cells were differentiated via embryoid body formation and plating, and after 3 days they were subjected to daily 10 minutes of LIUS treatment with various conditions: (1) low-pulsed (21 mW/cm2 , 20% duty cycle), (2) low-continuous, (3) high-pulsed (147 mW/cm2 , 20% duty cycle), and (4) high-continuous LIUS. RESULTS Low-pulsed and high-continuous LIUS had improved beating rates of contractile areas as well as increased late cardiac gene expressions, such as α- and β-myosin heavy chain and cardiac troponin T, showing its benefits on cardiomyocyte differentiation. Meanwhile, an early endodermal marker, α-fetoprotein, was significantly attenuated after LIUS treatments. CONCLUSIONS With these observations, it is demonstrated that LIUS simulation could enhance cardiomyogenesis from embryonic stem cells and increase its selectivity toward cardiomyocytes by reducing spontaneous differentiation.
Collapse
Affiliation(s)
- Ailing Teo
- Schools of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Amir Morshedi
- Schools of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Jen-Chieh Wang
- Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore
| | - Yufeng Zhou
- Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore
| | - Mayasari Lim
- Schools of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| |
Collapse
|
23
|
Lee JK, Huwe LW, Paschos N, Aryaei A, Gegg CA, Hu JC, Athanasiou KA. Tension stimulation drives tissue formation in scaffold-free systems. NATURE MATERIALS 2017; 16:864-873. [PMID: 28604717 PMCID: PMC5532069 DOI: 10.1038/nmat4917] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 05/04/2017] [Indexed: 05/04/2023]
Abstract
Scaffold-free systems have emerged as viable approaches for engineering load-bearing tissues. However, the tensile properties of engineered tissues have remained far below the values for native tissue. Here, by using self-assembled articular cartilage as a model to examine the effects of intermittent and continuous tension stimulation on tissue formation, we show that the application of tension alone, or in combination with matrix remodelling and synthesis agents, leads to neocartilage with tensile properties approaching those of native tissue. Implantation of tension-stimulated tissues results in neotissues that are morphologically reminiscent of native cartilage. We also show that tension stimulation can be translated to a human cell source to generate anisotropic human neocartilage with enhanced tensile properties. Tension stimulation, which results in nearly sixfold improvements in tensile properties over unstimulated controls, may allow the engineering of mechanically robust biological replacements of native tissue.
Collapse
Affiliation(s)
- Jennifer K. Lee
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Le W. Huwe
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Nikolaos Paschos
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Ashkan Aryaei
- Department of Biomedical Engineering, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Courtney A. Gegg
- Department of Bioengineering, Stanford University, 443 Via Ortega, Stanford, CA, 94305
| | - Jerry C. 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
- Department of Orthopaedic Surgery, University of California, Davis, One Shields Avenue, Davis, CA, 95616, USA
- Correspondence and reprint requests should be addressed to: KA Athanasiou, Tel.: (530) 754-6645, Fax: (530) 754-5739, , Department of Biomedical Engineering, University of California, Davis, One Shields Ave, Davis, CA 95616, USA
| |
Collapse
|
24
|
Tanaka N, Ohno S, Honda K, Tanimoto K, Doi T, Ohno-Nakahara M, Tafolla E, Kapila S, Tanne K. Cyclic Mechanical Strain Regulates the PTHrP Expression in Cultured Chondrocytes via Activation of the Ca2+ Channel. J Dent Res 2016; 84:64-8. [PMID: 15615878 DOI: 10.1177/154405910508400111] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The association between mechanical stimulation and chondrocyte homeostasis has been reported. However, the participation of PTHrP (parathyroid-hormone-related protein) in the mechano-regulation of chondrocyte metabolism remains unclear. We determined whether mechanical stimulation of chondrocytes induces the expression of PTHrP and, further, whether the mechano-modulation of PTHrP is dependent on the maturational status of chondrocytes. Cyclic mechanical strain was applied to rat growth plate chondrocytes at the proliferating, matrix-forming, and hypertrophic stages at 30 cycles/min. Cyclic mechanical strain significantly increased PTHrP mRNA levels in chondrocytes at the proliferating and matrix-forming stages only. The induction of PTHrP was dependent on loading magnitude at the proliferating stage. Using specific ion channel blockers, we determined that mechano-induction of PTHrP was inhibited by nifedipine, a Ca2+ channel blocker. These results suggest that mechanical induction of PTHrP possibly provides the environment for greater chondrocyte replication and matrix formation that would subsequently affect cartilage formation.
Collapse
Affiliation(s)
- N Tanaka
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Graduate School of Biomedical Sciences, 1-2-3 Kasumi, Minami-Ku, Hiroshima 734-8553, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Chow SKH, Leung KS, Qin J, Guo A, Sun M, Qin L, Cheung WH. Mechanical stimulation enhanced estrogen receptor expression and callus formation in diaphyseal long bone fracture healing in ovariectomy-induced osteoporotic rats. Osteoporos Int 2016; 27:2989-3000. [PMID: 27155884 DOI: 10.1007/s00198-016-3619-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 04/27/2016] [Indexed: 12/28/2022]
Abstract
UNLABELLED Estrogen receptor (ER) in ovariectomy-induced osteoporotic fracture was reported to exhibit delayed expression. Mechanical stimulation enhanced ER-α expression in osteoporotic fracture callus at the tissue level. ER was also found to be required for the effectiveness of vibrational mechanical stimulation treatment in osteoporotic fracture healing. INTRODUCTION Estrogen receptor(ER) is involved in mechanical signal transduction in bone metabolism. Its expression was reported to be delayed in osteoporotic fracture healing. The purpose of this study was to investigate the roles played by ER during osteoporotic fracture healing enhanced with mechanical stimulation. METHODS Ovariectomy-induced osteoporotic SD rats that received closed femoral fractures were divided into five groups, (i) SHAM, (ii) SHAM-VT, (iii) OVX, (iv) OVX-VT, and (v) OVX-VT-ICI, where VT stands for whole-body vibration treatment and ICI for ER antagonization by ICI 182,780. Callus formation and gene expression were assessed at 2, 4, and 8 weeks postfracture. In vitro osteoblastic differentiation, mineralization, and ER-α expression were assessed. RESULTS The delayed ER expression was found to be enhanced by vibration treatment. Callus formation enhancement was shown by callus morphometry and micro-CT analysis. Enhancement effects by vibration were partially abolished when ER was modulated by ICI 182,780, in terms of callus formation capacity at 2-4 weeks and ER gene and protein expression at all time points. In vitro, ER expression in osteoblasts was not enhanced by VT treatment, but osteoblastic differentiation and mineralization were enhanced under estrogen-deprived condition. When osteoblastic cells were modulated by ICI 182,780, enhancement effects of VT were eliminated. CONCLUSIONS Vibration was able to enhance ER expression in ovariectomy-induced osteoporotic fracture healing. ER was essential in mechanical signal transduction and enhancement in callus formation effects during osteoporotic fracture healing enhanced by vibration. The enhancement of ER-α expression by mechanical stimulation was not likely to be related to the increased expression in osteoblastic cells but rather to the systemic enhancement in recruitment of ER-expressing progenitor cells through increased blood flow and neo-angiogenesis. This finding might explain the observed difference in mechanical sensitivity of osteoporotic fracture to mechanical stimulation.
Collapse
Affiliation(s)
- S K H Chow
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Hong Kong Special Administrative Region, The Chinese University of Hong Kong, Hong Kong, The People's Republic of China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, The People's Republic of China
| | - K S Leung
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Hong Kong Special Administrative Region, The Chinese University of Hong Kong, Hong Kong, The People's Republic of China
- Translational Medicine Research and Development Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, The People's Republic of China
| | - J Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Hong Kong Special Administrative Region, The Chinese University of Hong Kong, Hong Kong, The People's Republic of China
| | - A Guo
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Hong Kong Special Administrative Region, The Chinese University of Hong Kong, Hong Kong, The People's Republic of China
| | - M Sun
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Hong Kong Special Administrative Region, The Chinese University of Hong Kong, Hong Kong, The People's Republic of China
| | - L Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Hong Kong Special Administrative Region, The Chinese University of Hong Kong, Hong Kong, The People's Republic of China
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, The People's Republic of China
- Translational Medicine Research and Development Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, The People's Republic of China
| | - W H Cheung
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Hong Kong Special Administrative Region, The Chinese University of Hong Kong, Hong Kong, The People's Republic of China.
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, The People's Republic of China.
- Translational Medicine Research and Development Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, The People's Republic of China.
| |
Collapse
|
26
|
Hoyer-Kuhn H, Höbing L, Cassens J, Schoenau E, Semler O. Children with severe Osteogenesis imperfecta and short stature present on average with normal IGF-I and IGFBP-3 levels. J Pediatr Endocrinol Metab 2016; 29:813-8. [PMID: 27089405 DOI: 10.1515/jpem-2015-0385] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 03/07/2016] [Indexed: 11/15/2022]
Abstract
BACKGROUND Osteogenesis imperfecta (OI) is characterized by bone fragility and short stature. Data about IGF-I/IGFBP-3 levels are rare in OI. Therefore IGF-I/IGFBP-3 levels in children with different types of OI were investigated. METHODS IGF-I and IGFBP-3 levels of 60 children (male n=38) were assessed in a retrospective cross-sectional setting. RESULTS Height/weight was significant different [height z-score type 3 versus type 4: p=0.0011 and weight (p≤0.0001)] between OI type 3 and 4. Mean IGF-I levels were in the lower normal range (mean±SD level 137.4±109.1 μg/L). Mean IGFBP-3 measurements were in the normal range (mean±SD 3.105±1.175 mg/L). No significant differences between OI type 3 and 4 children have been observed (IGF-I: p=0.0906; IGFBP-3: p=0.2042). CONCLUSIONS Patients with different severities of OI have IGF-I and IGFBP-3 levels in the lower normal range. The type of OI does not significantly influence these growth factors.
Collapse
|
27
|
Mousavi SJ, Doweidar MH. Numerical modeling of cell differentiation and proliferation in force-induced substrates via encapsulated magnetic nanoparticles. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2016; 130:106-117. [PMID: 27208526 DOI: 10.1016/j.cmpb.2016.03.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 03/16/2016] [Accepted: 03/17/2016] [Indexed: 06/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Cell migration, differentiation, proliferation and apoptosis are the main processes in tissue regeneration. Mesenchymal Stem Cells have the potential to differentiate into many cell phenotypes such as tissue- or organ-specific cells to perform special functions. Experimental observations illustrate that differentiation and proliferation of these cells can be regulated according to internal forces induced within their Extracellular Matrix. The process of how exactly they interpret and transduce these signals is not well understood. METHODS A previously developed three-dimensional (3D) computational model is here extended and employed to study how force-free substrates and force-induced substrate control cell differentiation and/or proliferation during the mechanosensing process. Consistent with experimental observations, it is assumed that cell internal deformation (a mechanical signal) in correlation with the cell maturation state directly triggers cell differentiation and/or proliferation. The Extracellular Matrix is modeled as Neo-Hookean hyperelastic material assuming that cells are cultured within 3D nonlinear hydrogels. RESULTS In agreement with well-known experimental observations, the findings here indicate that within neurogenic (0.1-1kPa), chondrogenic (20-25kPa) and osteogenic (30-45kPa) substrates, Mesenchymal Stem Cells differentiation and proliferation can be precipitated by inducing the substrate with an internal force. Therefore, cells require a longer time to grow and maturate within force-free substrates than within force-induced substrates. In the instance of Mesenchymal Stem Cells differentiation into a compatible phenotype, the magnitude of the net traction force increases within chondrogenic and osteogenic substrates while it reduces within neurogenic substrates. This is consistent with experimental studies and numerical works recently published by the same authors. However, in all cases the magnitude of the net traction force considerably increases at the instant of cell proliferation because of cell-cell interaction. CONCLUSIONS The present model provides new perspectives to delineate the role of force-induced substrates in remotely controlling the cell fate during cell-matrix interaction, which open the door for new tissue regeneration methodologies.
Collapse
Affiliation(s)
- Seyed Jamaleddin Mousavi
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, Spain; Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain
| | - Mohamed Hamdy Doweidar
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, Spain; Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Zaragoza, Spain.
| |
Collapse
|
28
|
Asmar A, Barrett-Jolley R, Werner A, Kelly R, Stacey M. Membrane channel gene expression in human costal and articular chondrocytes. Organogenesis 2016; 12:94-107. [PMID: 27116676 PMCID: PMC4981366 DOI: 10.1080/15476278.2016.1181238] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Chondrocytes are the uniquely resident cells found in all types of cartilage and key to their function is the ability to respond to mechanical loads with changes of metabolic activity. This mechanotransduction property is, in part, mediated through the activity of a range of expressed transmembrane channels; ion channels, gap junction proteins, and porins. Appropriate expression of ion channels has been shown essential for production of extracellular matrix and differential expression of transmembrane channels is correlated to musculoskeletal diseases such as osteoarthritis and Albers-Schönberg. In this study we analyzed the consistency of gene expression between channelomes of chondrocytes from human articular and costal (teenage and fetal origin) cartilages. Notably, we found 14 ion channel genes commonly expressed between articular and both types of costal cartilage chondrocytes. There were several other ion channel genes expressed only in articular (6 genes) or costal chondrocytes (5 genes). Significant differences in expression of BEST1 and KCNJ2 (Kir2.1) were observed between fetal and teenage costal cartilage. Interestingly, the large Ca2+ activated potassium channel (BKα, or KCNMA1) was very highly expressed in all chondrocytes examined. Expression of the gap junction genes for Panx1, GJA1 (Cx43) and GJC1 (Cx45) was also observed in chondrocytes from all cartilage samples. Together, this data highlights similarities between chondrocyte membrane channel gene expressions in cells derived from different anatomical sites, and may imply that common electrophysiological signaling pathways underlie cellular control. The high expression of a range of mechanically and metabolically sensitive membrane channels suggest that chondrocyte mechanotransduction may be more complex than previously thought.
Collapse
Affiliation(s)
- A Asmar
- a Frank Reidy Research Center for Bioelectrics, Old Dominion University , Norfolk , VA , USA
| | - R Barrett-Jolley
- b Department of Musculoskeletal Biology , University of Liverpool , England , UK
| | - A Werner
- c Department of Pathology , Eastern Virginia Medical School and Med Director of Laboratories, Children's Hospital of The King's Daughters , Norfolk , VA , USA
| | - R Kelly
- d Department of Surgery , Eastern Virginia Medical School and Pediatric Surgery Division, Children's Hospital of the King's Daughters , Norfolk , VA , USA
| | - M Stacey
- a Frank Reidy Research Center for Bioelectrics, Old Dominion University , Norfolk , VA , USA
| |
Collapse
|
29
|
Finlay S, Seedhom BB, Carey DO, Bulpitt AJ, Treanor DE, Kirkham J. In Vitro Engineering of High Modulus Cartilage-Like Constructs. Tissue Eng Part C Methods 2016; 22:382-97. [PMID: 26850081 PMCID: PMC4827287 DOI: 10.1089/ten.tec.2015.0351] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
To date, the outcomes of cartilage repair have been inconsistent and have frequently yielded mechanically inferior fibrocartilage, thereby increasing the chances of damage recurrence. Implantation of constructs with biochemical composition and mechanical properties comparable to natural cartilage could be advantageous for long-term repair. This study attempted to create such constructs, in vitro, using tissue engineering principles. Bovine synoviocytes were seeded on nonwoven polyethylene terephthalate fiber scaffolds and cultured in chondrogenic medium for 4 weeks, after which uniaxial compressive loading was applied using an in-house bioreactor for 1 h per day, at a frequency of 1 Hz, for a further 84 days. The initial loading conditions, determined from the mechanical properties of the immature constructs after 4 weeks in chondrogenic culture, were strains ranging between 13% and 23%. After 56 days (sustained at 84 days) of loading, the constructs were stained homogenously with Alcian blue and for type-II collagen. Dynamic compressive moduli were comparable to the high end values for native cartilage and proportional to Alcian blue staining intensity. We suggest that these high moduli values were attributable to the bioreactor setup, which caused the loading regime to change as the constructs developed, that is, the applied stress and strain increased with construct thickness and stiffness, providing continued sufficient cell stimulation as further matrix was deposited. Constructs containing cartilage-like matrix with response to load similar to that of native cartilage could produce long-term effective cartilage repair when implanted.
Collapse
Affiliation(s)
- Scott Finlay
- 1 Division of Oral Biology, School of Dentistry, University of Leeds , Leeds, United Kingdom
| | - Bahaa B Seedhom
- 1 Division of Oral Biology, School of Dentistry, University of Leeds , Leeds, United Kingdom
| | - Duane O Carey
- 2 School of Computing, University of Leeds , Leeds, United Kingdom
| | - Andy J Bulpitt
- 2 School of Computing, University of Leeds , Leeds, United Kingdom
| | - Darren E Treanor
- 3 Department of Pathology, Leeds Institute of Cancer and Pathology, University of Leeds , Leeds, United Kingdom .,4 Leeds Teaching Hospitals NHS Trust , Leeds, United Kingdom
| | - Jennifer Kirkham
- 1 Division of Oral Biology, School of Dentistry, University of Leeds , Leeds, United Kingdom
| |
Collapse
|
30
|
Abstract
Chondrocytes, the single cell type in adult articular cartilage, have conventionally been considered to be non-excitable cells. However, recent evidence suggests that their resting membrane potential (RMP) is less negative than that of excitable cells, and they are fully equipped with channels that control ion, water and osmolyte movement across the chondrocyte membrane. Amongst calcium-specific ion channels, members of the voltage-dependent calcium channel (VDCC) family are expressed in chondrocytes where they are functionally active. L-type VDCC inhibitors such as nifedipine and verapamil have contributed to our understanding of the roles of these ion channels in chondrogenesis, chondrocyte signalling and mechanotransduction. In this narrative review, we discuss published data indicating that VDCC function is vital for chondrocyte health, especially in regulating proliferation and maturation. We also highlight the fact that activation of VDCC function appears to accompany various inflammatory aspects of osteoarthritis (OA) and, based on in vitro data, the application of nifedipine and/or verapamil may be a promising approach for ameliorating OA severity. However, very few studies on clinical outcomes are available regarding the influence of calcium antagonists, which are used primarily for treating cardiovascular conditions in OA patients. This review is intended to stimulate further research on the chondrocyte 'channelome', contribute to the development of novel therapeutic strategies and facilitate the retargeting and repositioning of existing pharmacological agents currently used for other comorbidities for the treatment of OA.
Collapse
|
31
|
Abstract
The development of the vertebrate skeleton reflects its evolutionary history. Cartilage formation came before biomineralization and a head skeleton evolved before the formation of axial and appendicular skeletal structures. This review describes the processes that result in endochondral and intramembranous ossification, the important roles of growth and transcription factors, and the consequences of mutations in some of the genes involved. Following a summary of the origin of cartilage, muscle, and tendon cell lineages in the axial skeleton, we discuss the role of muscle forces in the formation of skeletal architecture and assembly of musculoskeletal functional units. Finally, ontogenetic patterning of bones in response to mechanical loading is reviewed.This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Agnes D Berendsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard University, USA
| | - Bjorn R Olsen
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard University, USA.
| |
Collapse
|
32
|
Shea CA, Rolfe RA, Murphy P. The importance of foetal movement for co-ordinated cartilage and bone development in utero : clinical consequences and potential for therapy. Bone Joint Res 2015; 4:105-16. [PMID: 26142413 PMCID: PMC4602203 DOI: 10.1302/2046-3758.47.2000387] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Construction of a functional skeleton is accomplished
through co-ordination of the developmental processes of chondrogenesis,
osteogenesis, and synovial joint formation. Infants whose movement in
utero is reduced or restricted and who subsequently suffer
from joint dysplasia (including joint contractures) and thin hypo-mineralised
bones, demonstrate that embryonic movement is crucial for appropriate
skeletogenesis. This has been confirmed in mouse, chick, and zebrafish
animal models, where reduced or eliminated movement consistently yields
similar malformations and which provide the possibility of experimentation
to uncover the precise disturbances and the mechanisms by which
movement impacts molecular regulation. Molecular genetic studies have
shown the important roles played by cell communication signalling
pathways, namely Wnt, Hedgehog, and transforming growth factor-beta/bone
morphogenetic protein. These pathways regulate cell behaviours such
as proliferation and differentiation to control maturation of the
skeletal elements, and are affected when movement is altered. Cell
contacts to the extra-cellular matrix as well as the cytoskeleton
offer a means of mechanotransduction which could integrate mechanical
cues with genetic regulation. Indeed, expression of cytoskeletal
genes has been shown to be affected by immobilisation. In addition
to furthering our understanding of a fundamental aspect of cell control
and differentiation during development, research in this area is
applicable to the engineering of stable skeletal tissues from stem
cells, which relies on an understanding of developmental mechanisms
including genetic and physical criteria. A deeper understanding
of how movement affects skeletogenesis therefore has broader implications
for regenerative therapeutics for injury or disease, as well as
for optimisation of physical therapy regimes for individuals affected
by skeletal abnormalities. Cite this article: Bone Joint Res 2015;4:105–116
Collapse
Affiliation(s)
- C A Shea
- Trinity College Dublin, College Green, Dublin, D2, Ireland
| | | | - P Murphy
- Trinity College Dublin, College Green, Dublin, D2, Ireland
| |
Collapse
|
33
|
Kurita T, Yamamura H, Suzuki Y, Giles WR, Imaizumi Y. The ClC-7 Chloride Channel Is Downregulated by Hypoosmotic Stress in Human Chondrocytes. Mol Pharmacol 2015; 88:113-20. [PMID: 25943117 DOI: 10.1124/mol.115.098160] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 05/05/2015] [Indexed: 02/14/2025] Open
Abstract
Articular chondrocytes in osteoarthritis (OA) patients are exposed to hypoosmotic stress because the osmolality of this synovial fluid is significantly decreased. Hypoosmotic stress can cause an efflux of Cl(-) and an associated decrease of cell volume. We have previously reported that a Cl(-) conductance contributes to the regulation of resting membrane potential and thus can alter intracellular Ca(2+) concentration ([Ca(2+)]i) in human chondrocytes. The molecular identity and pathologic function of these Cl(-) channels, however, remained to be determined. Here, we show that the ClC-7 Cl(-) channel is strongly expressed in a human chondrocyte cell line (OUMS-27) and that it is responsible for Cl(-) currents that are activated by extracellular acidification (pH 5.0). These acid-sensitive currents are inhibited by 4,4'-diisothiocyanatostilbene-2,2'-disulfonic acid (DIDS; IC50 = 13 μM) and are markedly reduced by small-interfering RNA-induced knockdown of ClC-7. DIDS hyperpolarized these chondrocytes, and this was followed by an increase in [Ca(2+)]i. ClC-7 knockdown caused a similar hyperpolarization of the membrane potential. Short-term culture (48 hours) in hypoosmotic medium (270 mOsm) reduced the expression of ClC-7 and decreased the acid-sensitive currents. Interestingly, these hypoosmotic culture conditions, or ClC-7 knockdown, resulted in enhanced cell death. Taken together, our results show that the significant hyperpolarization due to ClC-7 impairment in chondrocytes can significantly increase [Ca(2+)]i and cell death. Thus, downregulation of ClC-7 channels during the hypoosmotic stress that accompanies OA progression is one important concept of the complex etiology of OA. These findings suggest novel targets for therapeutic intervention(s) and drug development for OA.
Collapse
Affiliation(s)
- Takashi Kurita
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., H.Y., Y.S., Y.I.); and Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada (W.R.G.)
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., H.Y., Y.S., Y.I.); and Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada (W.R.G.)
| | - Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., H.Y., Y.S., Y.I.); and Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada (W.R.G.)
| | - Wayne R Giles
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., H.Y., Y.S., Y.I.); and Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada (W.R.G.)
| | - Yuji Imaizumi
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan (T.K., H.Y., Y.S., Y.I.); and Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada (W.R.G.)
| |
Collapse
|
34
|
Mousavi SJ, Hamdy Doweidar M. Role of Mechanical Cues in Cell Differentiation and Proliferation: A 3D Numerical Model. PLoS One 2015; 10:e0124529. [PMID: 25933372 PMCID: PMC4416758 DOI: 10.1371/journal.pone.0124529] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/16/2015] [Indexed: 12/28/2022] Open
Abstract
Cell differentiation, proliferation and migration are essential processes in tissue regeneration. Experimental evidence confirms that cell differentiation or proliferation can be regulated according to the extracellular matrix stiffness. For instance, mesenchymal stem cells (MSCs) can differentiate to neuroblast, chondrocyte or osteoblast within matrices mimicking the stiffness of their native substrate. However, the precise mechanisms by which the substrate stiffness governs cell differentiation or proliferation are not well known. Therefore, a mechano-sensing computational model is here developed to elucidate how substrate stiffness regulates cell differentiation and/or proliferation during cell migration. In agreement with experimental observations, it is assumed that internal deformation of the cell (a mechanical signal) together with the cell maturation state directly coordinates cell differentiation and/or proliferation. Our findings indicate that MSC differentiation to neurogenic, chondrogenic or osteogenic lineage specifications occurs within soft (0.1-1 kPa), intermediate (20-25 kPa) or hard (30-45 kPa) substrates, respectively. These results are consistent with well-known experimental observations. Remarkably, when a MSC differentiate to a compatible phenotype, the average net traction force depends on the substrate stiffness in such a way that it might increase in intermediate and hard substrates but it would reduce in a soft matrix. However, in all cases the average net traction force considerably increases at the instant of cell proliferation because of cell-cell interaction. Moreover cell differentiation and proliferation accelerate with increasing substrate stiffness due to the decrease in the cell maturation time. Thus, the model provides insights to explain the hypothesis that substrate stiffness plays a key role in regulating cell fate during mechanotaxis.
Collapse
Affiliation(s)
- Seyed Jamaleddin Mousavi
- Group of Structural Mechanics and Materials Modeling (GEMM), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Spain
| | - Mohamed Hamdy Doweidar
- Group of Structural Mechanics and Materials Modeling (GEMM), Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Spain
- Mechanical Engineering Department, School of Engineering and Architecture (EINA), University of Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Zaragoza, Spain
- * E-mail:
| |
Collapse
|
35
|
Inayama M, Suzuki Y, Yamada S, Kurita T, Yamamura H, Ohya S, Giles WR, Imaizumi Y. Orai1-Orai2 complex is involved in store-operated calcium entry in chondrocyte cell lines. Cell Calcium 2015; 57:337-47. [PMID: 25769459 DOI: 10.1016/j.ceca.2015.02.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Revised: 02/01/2015] [Accepted: 02/11/2015] [Indexed: 12/18/2022]
Abstract
Ca(2+) influx via store-operated Ca(2+) entry (SOCE) plays critical roles in many essential cellular functions. The Ca(2+) release-activated Ca(2+) (CRAC) channel complex, consisting of Orai and STIM, is one of the major components of store-operated Ca(2+) (SOC) channels. Our previous study demonstrated that histamine can cause sustained Ca(2+) entry through SOC channels in OUMS-27 cells derived from human chondrosarcoma. This SOCE was increased by low- and decreased by high-concentrations of 2-aminoethoxydiphenyl borate. Quantitative reverse transcription PCR and Western blot analyses revealed abundant expressions of Orai1, Orai2 and STIM1. Introduction of dominant negative mutant of Orai1, or siOrai1 knockdown significantly attenuated SOCE. Following histamine application, single molecule imaging using total internal reflection fluorescence (TIRF) microscopy demonstrated punctate Orai1-STIM1 complex formation in plasma membrane. In contrast, knockdown or over-expression of Orai2 resulted in an increase or a decrease in SOCE, respectively. Finally, TIRF imaging revealed direct coupling between Orai1 and Orai2, and suggested that Orai2 reduces Orai1 function by formation of a hetero-tetramer. These results provide substantial evidence that Orai1, Orai2 and STIM1 form functional CRAC channels in OUMS-27 cells and that these complexes are responsible for sustained Ca(2+) entry in response to agonist stimulation.
Collapse
Affiliation(s)
- Munenori Inayama
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Yoshiaki Suzuki
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Satoshi Yamada
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Takashi Kurita
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Hisao Yamamura
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Susumu Ohya
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; Department of Pharmacology, Division of Pathological Sciences, Kyoto Pharmaceutical University, Kyoto 607-8414, Japan
| | - Wayne R Giles
- Faculty of Kinesiology, University of Calgary, Calgary, Alberta T2N 1N4, Canada
| | - Yuji Imaizumi
- Department of Molecular & Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan.
| |
Collapse
|
36
|
Rais Y, Reich A, Simsa-Maziel S, Moshe M, Idelevich A, Kfir T, Miosge N, Monsonego-Ornan E. The growth plate's response to load is partially mediated by mechano-sensing via the chondrocytic primary cilium. Cell Mol Life Sci 2015; 72:597-615. [PMID: 25084815 PMCID: PMC11114052 DOI: 10.1007/s00018-014-1690-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 07/20/2014] [Accepted: 07/21/2014] [Indexed: 02/03/2023]
Abstract
Mechanical load plays a significant role in bone and growth-plate development. Chondrocytes sense and respond to mechanical stimulation; however, the mechanisms by which those signals exert their effects are not fully understood. The primary cilium has been identified as a mechano-sensor in several cell types, including renal epithelial cells and endothelium, and accumulating evidence connects it to mechano-transduction in chondrocytes. In the growth plate, the primary cilium is involved in several regulatory pathways, such as the non-canonical Wnt and Indian Hedgehog. Moreover, it mediates cell shape, orientation, growth, and differentiation in the growth plate. In this work, we show that mechanical load enhances ciliogenesis in the growth plate. This leads to alterations in the expression and localization of key members of the Ihh-PTHrP loop resulting in decreased proliferation and an abnormal switch from proliferation to differentiation, together with abnormal chondrocyte morphology and organization. Moreover, we use the chondrogenic cell line ATDC5, a model for growth-plate chondrocytes, to understand the mechanisms mediating the participation of the primary cilium, and in particular KIF3A, in the cell's response to mechanical stimulation. We show that this key component of the cilium mediates gene expression in response to mechanical stimulation.
Collapse
Affiliation(s)
- Yoach Rais
- Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, P.O. Box 12, 76100, Rehovot, Israel
| | - Adi Reich
- Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, P.O. Box 12, 76100, Rehovot, Israel
- Bone and Extracellular Matrix Branch, National Institute of Child Health and Human Development, Bethesda, 20892-1830, MD, USA
| | - Stav Simsa-Maziel
- Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, P.O. Box 12, 76100, Rehovot, Israel
| | - Maya Moshe
- Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, P.O. Box 12, 76100, Rehovot, Israel
| | - Anna Idelevich
- Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, P.O. Box 12, 76100, Rehovot, Israel
| | - Tal Kfir
- Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, P.O. Box 12, 76100, Rehovot, Israel
| | - Nicolai Miosge
- Department of Prosthodontics, Oral Biology and Tissue Regeneration Work Group, Medical Faculty, Georg-August-University, 37075, Goettingen, Germany
| | - Efrat Monsonego-Ornan
- Institute of Biochemistry and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University, P.O. Box 12, 76100, Rehovot, Israel.
| |
Collapse
|
37
|
Brady MA, Waldman SD, Ethier CR. The Application of Multiple Biophysical Cues to Engineer Functional Neocartilage for Treatment of Osteoarthritis. Part I: Cellular Response. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:1-19. [DOI: 10.1089/ten.teb.2013.0757] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Mariea A. Brady
- Department of Bioengineering, Imperial College London, South Kensington, London, United Kingdom
| | | | - C. Ross Ethier
- Department of Bioengineering, Imperial College London, South Kensington, London, United Kingdom
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
38
|
Guevara JM, Moncayo MA, Vaca-González JJ, Gutiérrez ML, Barrera LA, Garzón-Alvarado DA. Growth plate stress distribution implications during bone development: a simple framework computational approach. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2015; 118:59-68. [PMID: 25453383 DOI: 10.1016/j.cmpb.2014.10.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 09/22/2014] [Accepted: 10/08/2014] [Indexed: 06/04/2023]
Abstract
Mechanical stimuli play a significant role in the process of long bone development as evidenced by clinical observations and in vivo studies. Up to now approaches to understand stimuli characteristics have been limited to the first stages of epiphyseal development. Furthermore, growth plate mechanical behavior has not been widely studied. In order to better understand mechanical influences on bone growth, we used Carter and Wong biomechanical approximation to analyze growth plate mechanical behavior, and explore stress patterns for different morphological stages of the growth plate. To the best of our knowledge this work is the first attempt to study stress distribution on growth plate during different possible stages of bone development, from gestation to adolescence. Stress distribution analysis on the epiphysis and growth plate was performed using axisymmetric (3D) finite element analysis in a simplified generic epiphyseal geometry using a linear elastic model as the first approximation. We took into account different growth plate locations, morphologies and widths, as well as different epiphyseal developmental stages. We found stress distribution during bone development established osteogenic index patterns that seem to influence locally epiphyseal structures growth and coincide with growth plate histological arrangement.
Collapse
Affiliation(s)
- J M Guevara
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - M A Moncayo
- Biomimetics Laboratory and Numerical Methods and Modeling Research Group (GNUM), Instituto de Biotecnología (IBUN), Universidad Nacional de Colombia, Bogotá, Colombia
| | - J J Vaca-González
- Biomimetics Laboratory and Numerical Methods and Modeling Research Group (GNUM), Instituto de Biotecnología (IBUN), Universidad Nacional de Colombia, Bogotá, Colombia
| | - M L Gutiérrez
- Biomimetics Laboratory and Numerical Methods and Modeling Research Group (GNUM), Instituto de Biotecnología (IBUN), Universidad Nacional de Colombia, Bogotá, Colombia
| | - L A Barrera
- Institute for the Study of Inborn Errors of Metabolism, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - D A Garzón-Alvarado
- Biomimetics Laboratory and Numerical Methods and Modeling Research Group (GNUM), Instituto de Biotecnología (IBUN), Universidad Nacional de Colombia, Bogotá, Colombia.
| |
Collapse
|
39
|
Tanne K, Okamoto Y, Su SC, Mitsuyoshi T, Asakawa-Tanne Y, Tanimoto K. Current status of temporomandibular joint disorders and the therapeutic system derived from a series of biomechanical, histological, and biochemical studies. APOS TRENDS IN ORTHODONTICS 2014. [DOI: 10.4103/2321-1407.148014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
This article was designed to report the current status of temporomandibular joint disorders (TMDs) and the therapeutic system on the basis of a series of clinical, biomechanical, histological and biochemical studies in our research groups. In particular, we have focused on the association of degenerative changes of articular cartilage in the mandibular condyle and the resultant progressive condylar resorption with mechanical stimuli acting on the condyle during the stomatognathic function. In a clinical aspect, the nature and prevalence of TMDs, association of malocclusion with TMDs, association of condylar position with TMDs, association of craniofacial morphology with TMDs, and influences of TMDs, TMJ-osteoarthritis (TMJ-OA) in particular, were examined. In a biomechanical aspect, the nature of stress distribution in the TMJ from maximum clenching was analyzed with finite element method. In addition, the pattern of stress distribution was examined in association with varying vertical discrepancies of the craniofacial skeleton and friction between the articular disk and condyle. The results demonstrated an induction of large compressive stresses in the anterior and lateral areas on the condyle by the maximum clenching and the subsequent prominent increases in the same areas of the mandibular condyle as the vertical skeletal discrepancy became more prominent. Increase of friction at the articular surface was also indicated as a cause of larger stresses and the relevant disk displacement, which further induced an increase in stresses in the tissues posterior to the disks, indicating an important role of TMJ disks as a stress absorber. In a histological or biological aspect, increase in TMJ loading simulated by vertical skeletal discrepancy, which has already been revealed by the preceding finite element analysis or represented by excessive mouth opening, produced a decrease in the thickness of cartilage layers, an increase in the numbers of chondroblasts and osteoclasts and the subsequent degenerative changes in the condylar cartilage associated with the expression of bone resorption-related factors. In a biochemical or molecular and cellular aspect, excessive mechanical stimuli, irrespective of compressive or tensile stress, induced HA fragmentation, expression of proinflammatory cytokines, an imbalance between matrix metalloproteinases and the tissue inhibitors, all of which are assumed to induce lower resistance to external stimuli and degenerative changes leading to bone and cartilage resorption. Excessive mechanical stimuli also reduced the synthesis of superficial zone protein in chondrocytes, which exerts an important role in the protection of cartilage and bone layers from the degenerative changes. It is also revealed that various cytoskeletal changes induced by mechanical stimuli are transmitted through a stretch-activated or Ca2+channel. Finally, on the basis of the results from a series of studies, it is demonstrated that optimal intra-articular environment can be achieved by splint therapy, if indicated, followed by occlusal reconstruction with orthodontic approach in patients with myalgia of the masticatory muscles, and TMJ internal derangement or anterior disk displacement with or without reduction. It is thus shown that orthodontic treatment is available for the treatment of TMDs and the long-term stability after treatment.
Collapse
Affiliation(s)
- Kazuo Tanne
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuki Okamoto
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Shao-Ching Su
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Tomomi Mitsuyoshi
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yuki Asakawa-Tanne
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| | - Kotaro Tanimoto
- Department of Orthodontics and Craniofacial Developmental Biology, Hiroshima University Institute of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
40
|
Chondrosenescence: definition, hallmarks and potential role in the pathogenesis of osteoarthritis. Maturitas 2014; 80:237-44. [PMID: 25637957 DOI: 10.1016/j.maturitas.2014.12.003] [Citation(s) in RCA: 166] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 12/06/2014] [Indexed: 01/07/2023]
Abstract
Aging and inflammation are major contributing factors to the development and progression of arthritic and musculoskeletal diseases. "Inflammaging" refers to low-grade inflammation that occurs during physiological aging. In this paper we review the published literature on cartilage aging and propose the term "chondrosenescence" to define the age-dependent deterioration of chondrocyte function and how it undermines cartilage function in osteoarthritis. We propose the concept that a small number of senescent chondrocytes may be able to take advantage of the inflammatory tissue microenvironment and the inflammaging and immunosenescence that is concurrently occurring in the arthritic joint, further contributing to the age-related degradation of articular cartilage, subchondral bone, synovium and other tissues. In this new framework "chondrosenescence" is intimately linked with inflammaging and the disturbed interplay between autophagy and inflammasomes, thus contributing to the age-related increase in the prevalence of osteoarthritis and a decrease in the efficacy of articular cartilage repair. A better understanding of the basic mechanisms underlying chondrosenescence and its modification by drugs, weight loss, improved nutrition and physical exercise could lead to the development of new therapeutic and preventive strategies for osteoarthritis and a range of other age-related inflammatory joint diseases. Aging is inevitable but age-related diseases may be modifiable.
Collapse
|
41
|
Brady MA, Waldman SD, Ethier CR. The application of multiple biophysical cues to engineer functional neocartilage for treatment of osteoarthritis. Part II: signal transduction. TISSUE ENGINEERING PART B-REVIEWS 2014; 21:20-33. [PMID: 25065615 DOI: 10.1089/ten.teb.2013.0760] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The unique mechanoelectrochemical environment of cartilage has motivated researchers to investigate the effect of multiple biophysical cues, including mechanical, magnetic, and electrical stimulation, on chondrocyte biology. It is well established that biophysical stimuli promote chondrocyte proliferation, differentiation, and maturation within "biological windows" of defined dose parameters, including mode, frequency, magnitude, and duration of stimuli (see companion review Part I: Cellular Response). However, the underlying molecular mechanisms and signal transduction pathways activated in response to multiple biophysical stimuli remain to be elucidated. Understanding the mechanisms of biophysical signal transduction will deepen knowledge of tissue organogenesis, remodeling, and regeneration and aiding in the treatment of pathologies such as osteoarthritis. Further, this knowledge will provide the tissue engineer with a potent toolset to manipulate and control cell fate and subsequently develop functional replacement cartilage. The aim of this article is to review chondrocyte signal transduction pathways in response to mechanical, magnetic, and electrical cues. Signal transduction does not occur along a single pathway; rather a number of parallel pathways appear to be activated, with calcium signaling apparently common to all three types of stimuli, though there are different modes of activation. Current tissue engineering strategies, such as the development of "smart" functionalized biomaterials that enable the delivery of growth factors or integration of conjugated nanoparticles, may further benefit from targeting known signal transduction pathways in combination with external biophysical cues.
Collapse
Affiliation(s)
- Mariea A Brady
- 1 Department of Bioengineering, Imperial College London , London, United Kingdom
| | | | | |
Collapse
|
42
|
Yang X, Trehan SK, Guan Y, Sun C, Moore DC, Jayasuriya CT, Chen Q. Matrilin-3 inhibits chondrocyte hypertrophy as a bone morphogenetic protein-2 antagonist. J Biol Chem 2014; 289:34768-79. [PMID: 25331953 DOI: 10.1074/jbc.m114.583104] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Increased chondrocyte hypertrophy is often associated with cartilage joint degeneration in human osteoarthritis patients. Matrilin-3 knock-out (Matn3 KO) mice exhibit these features. However, the underlying mechanism is unknown. In this study, we sought a molecular explanation for increased chondrocyte hypertrophy in the mice prone to cartilage degeneration. We analyzed the effects of Matn3 on chondrocyte hypertrophy and bone morphogenetic protein (Bmp) signaling by quantifying the hypertrophic marker collagen type X (Col X) gene expression and Smad1 activity in Matn3 KO mice in vivo and in Matn3-overexpressing chondrocytes in vitro. The effect of Matn3 and its specific domains on BMP activity were quantified by Col X promoter activity containing the Bmp-responsive element. Binding of MATN3 with BMP-2 was determined by immunoprecipitation, solid phase binding, and surface plasmon resonance assays. In Matn3 KO mice, Smad1 activity was increased more in growth plate chondrocytes than in wild-type mice. Conversely, Matn3 overexpression in hypertrophic chondrocytes led to inhibition of Bmp-2-stimulated, BMP-responsive element-dependent Col X expression and Smad1 activity. MATN3 bound BMP-2 in a dose-dependent manner. Multiple epidermal growth factor (EGF)-like domains clustered together by the coiled coil of Matn3 is required for Smad1 inhibition. Hence, as a novel BMP-2-binding protein and antagonist in the cartilage extracellular matrix, MATN3 may have the inherent ability to inhibit premature chondrocyte hypertrophy by suppressing BMP-2/Smad1 activity.
Collapse
Affiliation(s)
- Xu Yang
- From the Cell and Molecular Biology Laboratory, Department of Orthopaedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island 02903
| | - Samir K Trehan
- From the Cell and Molecular Biology Laboratory, Department of Orthopaedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island 02903
| | - Yingjie Guan
- From the Cell and Molecular Biology Laboratory, Department of Orthopaedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island 02903
| | - Changqi Sun
- From the Cell and Molecular Biology Laboratory, Department of Orthopaedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island 02903
| | - Douglas C Moore
- From the Cell and Molecular Biology Laboratory, Department of Orthopaedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island 02903
| | - Chathuraka T Jayasuriya
- From the Cell and Molecular Biology Laboratory, Department of Orthopaedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island 02903
| | - Qian Chen
- From the Cell and Molecular Biology Laboratory, Department of Orthopaedics, Warren Alpert Medical School, Brown University/Rhode Island Hospital, Providence, Rhode Island 02903
| |
Collapse
|
43
|
Löfgren M, Ekman S, Svala E, Lindahl A, Ley C, Skiöldebrand E. Cell and matrix modulation in prenatal and postnatal equine growth cartilage, zones of Ranvier and articular cartilage. J Anat 2014; 225:548-68. [PMID: 25175365 DOI: 10.1111/joa.12232] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2014] [Indexed: 11/30/2022] Open
Abstract
Formation of synovial joints includes phenotypic changes of the chondrocytes and the organisation of their extracellular matrix is regulated by different factors and signalling pathways. Increased knowledge of the normal processes involved in joint development may be used to identify similar regulatory mechanisms during pathological conditions in the joint. Samples of the distal radius were collected from prenatal and postnatal equine growth plates, zones of Ranvier and articular cartilage with the aim of identifying Notch signalling components and cells with stem cell-like characteristics and to follow changes in matrix protein localisation during joint development. The localisation of the Notch signalling components Notch1, Delta4, Hes1, Notch dysregulating protein epidermal growth factor-like domain 7 (EGFL7), the stem cell-indicating factor Stro-1 and the matrix molecules cartilage oligomeric matrix protein (COMP), fibromodulin, matrilin-1 and chondroadherin were studied using immunohistochemistry. Spatial changes in protein localisations during cartilage maturation were observed for Notch signalling components and matrix molecules, with increased pericellular localisation indicating new synthesis and involvement of these proteins in the formation of the joint. However, it was not possible to characterise the phenotype of the chondrocytes based on their surrounding matrix during normal chondrogenesis. The zone of Ranvier was identified in all horses and characterised as an area expressing Stro-1, EGFL7 and chondroadherin with an absence of COMP and Notch signalling. Stro-1 was also present in cells close to the perichondrium, in the articular cartilage and in the fetal resting zone, indicating stem cell-like characteristics of these cells. The presence of stem cells in the articular cartilage will be of importance for the repair of damaged cartilage. Perivascular chondrocytes and hypertrophic cells of the cartilage bone interface displayed positive staining for EGFL7, which is a novel finding and suggests a role of EGFL7 in the vascular infiltration of growth cartilage.
Collapse
Affiliation(s)
- Maria Löfgren
- Section of Pathology, Department of Biomedical Sciences and Veterinary Public Health, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | | | | | | | | |
Collapse
|
44
|
Guan Y, Yang X, Yang W, Charbonneau C, Chen Q. Mechanical activation of mammalian target of rapamycin pathway is required for cartilage development. FASEB J 2014; 28:4470-81. [PMID: 25002119 DOI: 10.1096/fj.14-252783] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mechanical stress regulates development by modulating cell signaling and gene expression. However, the cytoplasmic components mediating mechanotransduction remain unclear. In this study, elimination of muscle contraction during chicken embryonic development resulted in a reduction in the activity of mammalian target of rapamycin (mTOR) in the cartilaginous growth plate. Inhibition of mTOR activity led to significant inhibition of chondrocyte proliferation, cartilage tissue growth, and expression of chondrogenic genes, including Indian hedgehog (Ihh), a critical mediator of mechanotransduction. Conversely, cyclic loading (1 Hz, 5% matrix deformation) of embryonic chicken growth plate chondrocytes in 3-dimensional (3D) collagen scaffolding induced sustained activation of mTOR. Mechanical activation of mTOR occurred in serum-free medium, indicating that it is independent of growth factor or nutrients. Treatment of chondrocytes with Rapa abolished mechanical activation of cell proliferation and Ihh gene expression. Cyclic loading of chondroprogenitor cells deficient in SH2-containing protein tyrosine phosphatase 2 (Shp2) further enhanced mechanical activation of mTOR, cell proliferation, and chondrogenic gene expression. This result suggests that Shp2 is an antagonist of mechanotransduction through inhibition of mTOR activity. Our data demonstrate that mechanical activation of mTOR is necessary for cell proliferation, chondrogenesis, and cartilage growth during bone development, and that mTOR is an essential mechanotransduction component modulated by Shp2 in the cytoplasm.
Collapse
Affiliation(s)
- Yingjie Guan
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, Rhode Island, USA; and
| | - Xu Yang
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, Rhode Island, USA; and Department of Orthopaedics, Affiliated Hospital of Medical College of Qingdao University, Qingdao, China
| | - Wentian Yang
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, Rhode Island, USA; and
| | - Cherie Charbonneau
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, Rhode Island, USA; and
| | - Qian Chen
- Cell and Molecular Biology Laboratory, Department of Orthopaedics, Alpert Medical School of Brown University/Rhode Island Hospital, Providence, Rhode Island, USA; and
| |
Collapse
|
45
|
Li X, Liu C, Liang W, Ye H, Chen W, Lin R, Li Z, Liu X, Wu M. Millimeter wave promotes the synthesis of extracellular matrix and the proliferation of chondrocyte by regulating the voltage-gated K+ channel. J Bone Miner Metab 2014; 32:367-77. [PMID: 24202060 DOI: 10.1007/s00774-013-0513-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 08/07/2013] [Indexed: 10/26/2022]
Abstract
Previously, we reported that millimeter wave promoted the chondrocyte proliferation by pushing cell cycle progression. Activation of K(+) channels plays an essential role in the stimulating of extracellular matrix (ECM) synthesis and the cell proliferation in chondrocytes. While it is unclear if millimeter wave enhances ECM synthesis and proliferation of chondrocytes by regulating K(+) channel activity, we here investigated the effects of millimeter waves on ECM synthesis, chondrocyte proliferation and ion channels in the primary chondrocyte culture. We found that millimeter waves led to the increase of chondrocyte viability, the morphological changes of chondrocyte, and the F-actin distortion and remodeling. Ultrastructural analysis showed that treated chondrocytes contained an expansion of mitochondria and granular endoplasmic reticulum, and a high number of cytoplasmic vesicles in the cytoplasm compared to untreated cells, suggesting millimeter waves increased the energy metabolism and protein synthesis of chondrocytes. The analysis of differential ion channels' genes expression further showed an obvious increase of Kcne1, Kcnj3 and Kcnq2. To determine the role of voltage-gated K(+) channel in chondrocyte, we blocked the voltage-gated K(+) channel with 10 mM tetraethylammonium (TEA) and treated chondrocytes with millimeter waves. The results indicated that TEA significantly negated the promotion of millimeter waves for the ECM synthesis and chondrocyte proliferation. Our results support the hypothesis that millimeter waves promote the synthesis of ECM and the proliferation of chondrocyte by regulating the voltage-gated K(+) channel.
Collapse
Affiliation(s)
- Xihai Li
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Cuesta A, Del Valle ME, García-Suárez O, Viña E, Cabo R, Vázquez G, Cobo JL, Murcia A, Alvarez-Vega M, García-Cosamalón J, Vega JA. Acid-sensing ion channels in healthy and degenerated human intervertebral disc. Connect Tissue Res 2014; 55:197-204. [PMID: 24432912 DOI: 10.3109/03008207.2014.884083] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Acid-sensing ion channels (ASICs) are a family of H(+)-gated voltage-insensitive ion channels that respond to extracellular acidification by regulating transmembrane Ca(2+) flux. Moreover, ASICs can also be gated by mechanical forces and may function as mechanosensors. The cells of the intervertebral disc (IVD) have an unusual acidic and hyperosmotic microenvironment. Changes in the pH and osmolarity determine the viability of IVD cells and the composition of the extracellular matrix, and both are the basis of IVD degeneration. In this study, the expression of ASICs (ASIC1, ASIC2, ASIC3 and ASIC4) mRNAs and proteins in human healthy and degenerated IVD was evaluated by quantitative reverse transcription-quantitative polymerase chain reaction and Western blot. The distribution of ASIC proteins was determined by immunohistochemistry. The mRNAs for all ASICs were detected in normal human IVD, and significantly increased levels were found in degenerated IVD. Western blots demonstrated the presence of proteins with estimated molecular weights of approximately 68-72 kDa. In both the annulus fibrosus (AF) and nucleus pulposus (NP) of normal IVD, ASIC2 is the most frequently expressed ASIC followed by ASIC3, ASIC1 and ASIC4. In the AF of degenerated IVD, there was a significant increase in the number of ASIC1 and ASIC4 positive cells, whereas in the NP, we found significant increase of expression of ASIC1, ASIC2 and ASIC3. These results describe the occurrence and localization of different ASICs in human healthy IVD, and their increased expression in degenerated IVD, thus suggesting that ASICs may be involved in IVD degeneration.
Collapse
|
47
|
Crocetti S, Beyer C, Unternährer S, Benavides Damm T, Schade-Kampmann G, Hebeisen M, Di Berardino M, Fröhlich J, Franco-Obregón A. Impedance flow cytometry gauges proliferative capacity by detecting TRPC1 expression. Cytometry A 2014; 85:525-36. [DOI: 10.1002/cyto.a.22461] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Revised: 12/10/2013] [Accepted: 03/03/2014] [Indexed: 01/13/2023]
Affiliation(s)
| | - Christian Beyer
- Electromagnetics in Medicine and Biology Group, Laboratory for Electromagnetic Fields and Microwave Electronics; ETH Zürich Switzerland
| | | | - Tatiana Benavides Damm
- Institute for Biomechanics; ETH Zürich Switzerland
- CC Aerospace Biomedical Science & Technology, Space Biology Group, Luzern University of Applied Sciences and Arts; Hergiswil Switzerland
| | | | - Monika Hebeisen
- Leister Process Technologies; Axetris Division; Kaegiswil Switzerland
| | | | - Jürg Fröhlich
- Electromagnetics in Medicine and Biology Group, Laboratory for Electromagnetic Fields and Microwave Electronics; ETH Zürich Switzerland
| | - Alfredo Franco-Obregón
- Institute for Biomechanics; ETH Zürich Switzerland
- Department of Surgery; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
| |
Collapse
|
48
|
Flow-perfusion interferes with chondrogenic and hypertrophic matrix production by mesenchymal stem cells. J Biomech 2013; 47:2122-9. [PMID: 24290176 DOI: 10.1016/j.jbiomech.2013.11.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 10/28/2013] [Accepted: 11/06/2013] [Indexed: 12/28/2022]
Abstract
Flow-perfusion is being promoted as a way to grow tissue-engineered cartilage in vitro. Yet, there is a concern that flow-perfusion may induce unwanted mechanical effects on chondrogenesis and terminal differentiation. Therefore, the aim of this study is to evaluate the effect of fluid flow on chondrogenesis and chondrocyte hypertrophy of MSCs in a well-established pellet culture model. Human MSC pellets were mounted into 3D-printed porous scaffolds in basic chondrogenic differentiation medium, containing TGF-β2. Constructs were then allowed to form cartilaginous matrix for 18 days, before they were transferred to a custom-built flow-perfusion system. A continuous flow of 1.22ml min(-1) was applied to the constructs for 10 days. Controls were maintained under static culture conditions. To evaluate chondrogenic and hypertrophic differentiation, RNA was isolated at day 20 and 28 and histology, immunohistochemistry and western blot analyses were performed after 28 days of culture. Abundant matrix was formed in the constructs, but production of chondrogenic and hypertrophic matrix components was affected by flow-perfusion. Although gene expression levels of the (late) hypertrophic and osteogenic marker osteocalcin increased by flow-perfusion, this did not result in more collagen type X protein deposition. Decreased GAG release, in combination with diminished collagen II staining, indicates reduced chondrogenesis in response to flow-perfusion. Caution should thus be taken when applying flow-perfusion to cultures to improve nutrient diffusion. Although we show that it is possible to influence the differentiation of chondrogenic differentiated MSCs by flow-perfusion, effects are inconsistent and strongly donor-dependent.
Collapse
|
49
|
Shwartz Y, Blitz E, Zelzer E. One load to rule them all: Mechanical control of the musculoskeletal system in development and aging. Differentiation 2013; 86:104-11. [DOI: 10.1016/j.diff.2013.07.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 07/01/2013] [Accepted: 07/12/2013] [Indexed: 12/24/2022]
|
50
|
Abstract
Development of the various components of a normal skeleton requires highly regulated signalling systems that co-ordinate spatial and temporal patterns of cell division, cell differentiation, and morphogenesis. Much work in recent decades has revealed cascades of molecular signalling, acting through key transcription factors to regulate, for example, organized chondrogenic and osteogenic differentiation. It is now clear that mechanical stimuli are also required for aspects of skeletogenesis but very little is known about how the mechanical signals are integrated with classic biochemical signalling. Spatially organized differentiation is vital to the production of functionally appropriate tissues contributing to precise, region specific morphologies, for example transient chondrogenesis of long bone skeletal rudiments, which prefigures osteogenic replacement of the cartilage template, compared with the production of permanent cartilage at the sites of articulation. Currently a lack of understanding of how these tissues are differentially regulated hampers efforts to specifically regenerate stable bone and cartilage. Here, we review current research revealing the influence of mechanical stimuli on specific aspects of skeletal development and refer to other developing systems to set the scene for current and future work to uncover the molecular mechanisms involved. We integrate this with a brief overview of the effects of mechanical stimulation on stem cells in culture bringing together developmental and tissue engineering aspects of mechanoregulation of cell behavior. A better understanding of the molecular mechanisms that link mechanical stimuli to transcriptional control guiding cell differentiation will lead to new ideas about how to effectively prime stem cells for tissue engineering and regenerative therapies.
Collapse
Affiliation(s)
- Rebecca Rolfe
- Department of Zoology, School of Natural Sciences, Trinity College, Dublin, Ireland
| | | | | |
Collapse
|