1
|
Chang CC, Jiang SS, Tsai FY, Hsu PJ, Hsieh CC, Wang LT, Yen ML, Yen BL. Targeting Conserved Pathways in 3D Spheroid Formation of Diverse Cell Types for Translational Application: Enhanced Functional and Antioxidant Capacity. Cells 2023; 12:2050. [PMID: 37626861 PMCID: PMC10453086 DOI: 10.3390/cells12162050] [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: 07/08/2023] [Revised: 08/03/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
Three-dimensional (3D) in vitro spheroid/organoid culture increasingly appears to better mimic physiological states than standard 2D systems. The biological consequence of 3D spheroids, however, differs for different cell types: for pluripotent embryonic stem cells (ESCs), differentiation and loss of stemness occur, while the converse is true for somatic and cancer cells. Despite such diverse consequences, there are likely conserved mechanisms governing 3D spheroid formation across cell types that are unknown but could be efficiently targeted for translational application. To elucidate such processes, we performed transcriptome analysis with functional validation on 2D- and 3D-cultured mouse ESCs, mesenchymal stromal/stem cells (MSCs), and cancer cells. At both the transcriptomic and functional levels, 3D spheroid formation resulted in commitment towards known cell-specific functional outcomes. Surprisingly in all cell types, downregulation of the cholesterol synthesis pathway was found during 3D spheroid formation, with modulation concomitantly affecting 3D spheroid formation and cell-specific consequences; similar results were seen with human cell types. Furthermore, improved antioxidant capacity after 3D spheroid formation across cell types was further enhanced with modulation of the pathway. These findings demonstrate the profound cell-specific consequences and the translational value of understanding conserved mechanisms across diverse cell types after 3D spheroid formation.
Collapse
Affiliation(s)
- Chia-Chi Chang
- Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), Taipei 114, Taiwan
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 350, Taiwan
| | | | - Fang-Yu Tsai
- National Institute of Cancer Research, NHRI, Zhunan 350, Taiwan
| | - Pei-Ju Hsu
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 350, Taiwan
| | - Chen-Chan Hsieh
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 350, Taiwan
| | - Li-Tzu Wang
- Department of Obstetrics/Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, Taipei 100, Taiwan
| | - Men-Luh Yen
- Department of Obstetrics/Gynecology, National Taiwan University (NTU) Hospital & College of Medicine, Taipei 100, Taiwan
| | - B. Linju Yen
- Graduate Institute of Life Sciences, National Defense Medical Center (NDMC), Taipei 114, Taiwan
- Regenerative Medicine Research Group, Institute of Cellular & System Medicine, National Health Research Institutes (NHRI), Zhunan 350, Taiwan
| |
Collapse
|
2
|
Gould NR, Torre OM, Leser JM, Stains JP. The cytoskeleton and connected elements in bone cell mechano-transduction. Bone 2021; 149:115971. [PMID: 33892173 PMCID: PMC8217329 DOI: 10.1016/j.bone.2021.115971] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/30/2021] [Accepted: 04/17/2021] [Indexed: 02/07/2023]
Abstract
Bone is a mechano-responsive tissue that adapts to changes in its mechanical environment. Increases in strain lead to increased bone mass acquisition, whereas decreases in strain lead to a loss of bone mass. Given that mechanical stress is a regulator of bone mass and quality, it is important to understand how bone cells sense and transduce these mechanical cues into biological changes to identify druggable targets that can be exploited to restore bone cell mechano-sensitivity or to mimic mechanical load. Many studies have identified individual cytoskeletal components - microtubules, actin, and intermediate filaments - as mechano-sensors in bone. However, given the high interconnectedness and interaction between individual cytoskeletal components, and that they can assemble into multiple discreet cellular structures, it is likely that the cytoskeleton as a whole, rather than one specific component, is necessary for proper bone cell mechano-transduction. This review will examine the role of each cytoskeletal element in bone cell mechano-transduction and will present a unified view of how these elements interact and work together to create a mechano-sensor that is necessary to control bone formation following mechanical stress.
Collapse
Affiliation(s)
- Nicole R Gould
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Olivia M Torre
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jenna M Leser
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joseph P Stains
- Department of Orthopaedics, University of Maryland School of Medicine, Baltimore, MD 21201, USA..
| |
Collapse
|
3
|
Camal Ruggieri IN, Cícero AM, Issa JPM, Feldman S. Bone fracture healing: perspectives according to molecular basis. J Bone Miner Metab 2021; 39:311-331. [PMID: 33151416 DOI: 10.1007/s00774-020-01168-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
Fractures have a great impact on health all around the world and with fracture healing optimization; this problem could be resolved partially. To make a practical contribution to this issue, the knowledge of bone tissue, cellularity, and metabolism is essential, especially cytoskeletal architecture and its transformations according to external pressures. Special physical and chemical characteristics of the extracellular matrix (ECM) allow the transmission of mechanical stimuli from outside the cell to the plasmatic membrane. The osteocyte cytoskeleton is conformed by a complex network of actin and microtubules combined with crosslinker proteins like vinculin and fimbrin, connecting and transmitting outside stimuli through EMC to cytoplasm. Herein, critical signaling pathways like Cx43-depending ones, MAPK/ERK, Wnt, YAP/TAZ, Rho-ROCK, and others are activated due to mechanical stimuli, resulting in osteocyte cytoskeletal changes and ECM remodeling, altering the tissue and, therefore, the bone. In recent years, the osteocyte has gained more interest and value in relation to bone homeostasis as a great coordinator of other cell populations, thanks to its unique functions. By integrating the latest advances in relation to intracellular signaling pathways, mechanotransmission system of the osteocyte and bone tissue engineering, there are promising experimental strategies, while some are ready for clinical trials. This work aims to show clearly and precisely the integration between cytoskeleton and main molecular pathways in relation to mechanotransmission mechanism in osteocytes, and the use of this theoretical knowledge in therapeutic tools for bone fracture healing.
Collapse
Affiliation(s)
- Iván Nadir Camal Ruggieri
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina.
| | - Andrés Mauricio Cícero
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina
| | | | - Sara Feldman
- School of Medicine, LABOATEM (Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory), Biological Chemistry Cat, School of Medicine, Rosario National University, Rosario, Argentina
- Research Council of the Rosario National University (CIUNR) and CONICET, Rosario, Argentina
| |
Collapse
|
4
|
Markov P, Hayes AJ, Zhu H, Boote C, Blain EJ. 3D immuno-confocal image reconstruction of fibroblast cytoskeleton and nucleus architecture. JOURNAL OF BIOPHOTONICS 2021; 14:e202000202. [PMID: 33038053 DOI: 10.1002/jbio.202000202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/28/2020] [Accepted: 10/07/2020] [Indexed: 06/11/2023]
Abstract
Computational models of cellular structures generally rely on simplifying approximations and assumptions that limit biological accuracy. This study presents a comprehensive image processing pipeline for creating unified three-dimensional (3D) reconstructions of the cell cytoskeletal networks and nuclei. Confocal image stacks of these cellular structures were reconstructed to 3D isosurfaces (Imaris), then tessellations were simplified to reduce the number of elements in initial meshes by applying quadric edge collapse decimation with preserved topology boundaries (MeshLab). Geometries were remeshed to ensure uniformity (Instant Meshes) and the resulting 3D meshes exported (ABAQUS) for downstream application. The protocol has been applied successfully to fibroblast cytoskeletal reorganisation in the scleral connective tissue of the eye, under mechanical load that mimics internal eye pressure. While the method herein is specifically employed to reconstruct immunofluorescent confocal imaging data, it is also more widely applicable to other biological imaging modalities where accurate 3D cell structures are required.
Collapse
Affiliation(s)
- Petar Markov
- School of Biosciences, Cardiff University, Cardiff, Wales, UK
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
- School of Engineering, Cardiff University, Cardiff, Wales, UK
| | - Anthony J Hayes
- Bioimaging Research Hub, School of Biosciences, Cardiff University, Cardiff, Wales, UK
| | - Hanxing Zhu
- School of Engineering, Cardiff University, Cardiff, Wales, UK
| | - Craig Boote
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, Wales, UK
| | - Emma J Blain
- School of Biosciences, Cardiff University, Cardiff, Wales, UK
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, Wales, UK
| |
Collapse
|
5
|
Kihara T, Umezu C, Sawada K, Furutani Y. Osteogenic cells form mineralized particles, a few μm in size, in a 3D collagen gel culture. PeerJ 2019; 7:e7889. [PMID: 31660270 PMCID: PMC6815190 DOI: 10.7717/peerj.7889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 09/13/2019] [Indexed: 02/03/2023] Open
Abstract
Osteogenic cells form mineralized matrices in vitro, as well as in vivo. The formation and shape of the mineralized matrices are highly regulated by the cells. In vitro formation of mineralized matrices by osteogenic cells can be a model for in vivo osteogenesis. In this study, using a three-dimensional (3D) collagen gel culture system, we developed a new in vitro model for the formation of mineralized particles, a few µm in size, by the osteogenic cells. Human osteosarcoma (HOS) cells formed spherical mineralized matrices (about 12 µm) at approximately 7 days when cultured with β-glycerophosphate (β-GP)-containing culture media on 2D tissue culture plates. Alternately, when they were cultured in a 3D collagen gel containing β-GP, they formed mineralized particles with about 1.7 µm in the gel at approximately 3 days. Calcium precipitation in the gel was evaluated by measuring the gel turbidity. This type of mineralization of HOS cells, which formed mineralized particles inside the gel, was also observed in a peptide-based hydrogel culture. The mineralized particles were completely diminished by inhibiting the activity of Pit-1, phosphate cotransporter, of the HOS cells. When mouse osteoblast-like MC3T3-E1 cells, which form large and flat mineralized matrices in 2D osteogenic conditions at approximately 3 weeks of culture, were cultured in a 3D collagen gel, they also formed mineralized particles in the gel, similar to those in HOS cells, at approximately 18 days. Thus, osteogenic cells cultured in the 3D collagen gel form mineralized particles over a shorter period, and the mineralization could be easily determined by gel turbidity. This 3D gel culture system of osteogenic cells acts as a useful model for cells forming particle-type mineralized matrices, and we assume that the mineralized particles in the 3D hydrogel are calcospherulites, which are derived from matrix vesicles secreted by osteogenic cells.
Collapse
Affiliation(s)
- Takanori Kihara
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, Kitakyushu, Fukuoka, Japan
| | - Chiya Umezu
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, Kitakyushu, Fukuoka, Japan
| | - Karin Sawada
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, Kitakyushu, Fukuoka, Japan
| | - Yukari Furutani
- Department of Life and Environment Engineering, Faculty of Environmental Engineering, The University of Kitakyushu, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
6
|
Alternating Differentiation and Dedifferentiation between Mature Osteoblasts and Osteocytes. Sci Rep 2019; 9:13842. [PMID: 31554848 PMCID: PMC6761144 DOI: 10.1038/s41598-019-50236-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/09/2019] [Indexed: 12/22/2022] Open
Abstract
Osteocytes are terminally differentiated osteoblasts embedded in the bone matrix. Evidence indicates that cells in the mesenchymal lineage possess plasticity. However, whether or not osteocytes have the capacity to dedifferentiate back into osteoblasts is unclear. This study aimed to clarify the dedifferentiation potential of osteocytes. Mouse calvarial osteoblasts were isolated and maintained in normal two-dimensional (2D) or collagen gel three-dimensional (3D) cultures. In 2D cultures, osteoblasts exhibited a typical fibroblast-like shape with high Alpl and minimal Sost, Fgf23, and Dmp1 expression and osteoblasts formed mineralised nodules. When these osteoblasts were transferred into 3D cultures, they showed a stellate shape with diminished cytoplasm and numerous long processes and expression of Alpl decreased while Sost, Fgf23, and Dmp1 were significantly increased. These cells were in cell cycle arrest and showed suppressed mineralisation, indicating that they were osteocytes. When these osteocytes were recovered from 3D cultures and cultured two-dimensionally again, they regained adequate cytoplasm and lost the long processes, resulting in a fibroblast-like shape. These cells showed high Alpl and low Sost, Fgf23, and Dmp1 expression with a high mineralisation capability, indicating that they were osteoblasts. This report shows that osteocytes possess the capacity to dedifferentiate back into mature osteoblasts without gene manipulation.
Collapse
|
7
|
Murshid S, Takano-Yamamoto T, Kamioka H. Differential distribution of microtubules in immature osteocytes in vivo. J Oral Biosci 2018. [DOI: 10.1016/j.job.2018.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
8
|
Morrell AE, Brown GN, Robinson ST, Sattler RL, Baik AD, Zhen G, Cao X, Bonewald LF, Jin W, Kam LC, Guo XE. Mechanically induced Ca 2+ oscillations in osteocytes release extracellular vesicles and enhance bone formation. Bone Res 2018; 6:6. [PMID: 29581909 PMCID: PMC5859015 DOI: 10.1038/s41413-018-0007-x] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 11/23/2017] [Indexed: 02/01/2023] Open
Abstract
The vast osteocytic network is believed to orchestrate bone metabolic activity in response to mechanical stimuli through production of sclerostin, RANKL, and osteoprotegerin (OPG). However, the mechanisms of osteocyte mechanotransduction remain poorly understood. We've previously shown that osteocyte mechanosensitivity is encoded through unique intracellular calcium (Ca2+) dynamics. Here, by simultaneously monitoring Ca2+ and actin dynamics in single cells exposed to fluid shear flow, we detected actin network contractions immediately upon onset of flow-induced Ca2+ transients, which were facilitated by smooth muscle myosin and further confirmed in native osteocytes ex vivo. Actomyosin contractions have been linked to the secretion of extracellular vesicles (EVs), and our studies demonstrate that mechanical stimulation upregulates EV production in osteocytes through immunostaining for the secretory vesicle marker Lysosomal-associated membrane protein 1 (LAMP1) and quantifying EV release in conditioned medium, both of which are blunted when Ca2+ signaling was inhibited by neomycin. Axial tibia compression was used to induce anabolic bone formation responses in mice, revealing upregulated LAMP1 and expected downregulation of sclerostin in vivo. This load-related increase in LAMP1 expression was inhibited in neomycin-injected mice compared to vehicle. Micro-computed tomography revealed significant load-related increases in both trabecular bone volume fraction and cortical thickness after two weeks of loading, which were blunted by neomycin treatment. In summary, we found mechanical stimulation of osteocytes activates Ca2+-dependent contractions and enhances the production and release of EVs containing bone regulatory proteins. Further, blocking Ca2+ signaling significantly attenuates adaptation to mechanical loading in vivo, suggesting a critical role for Ca2+-mediated signaling in bone adaptation.
Collapse
Affiliation(s)
- Andrea E. Morrell
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Genevieve N. Brown
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Samuel T. Robinson
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Rachel L. Sattler
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Andrew D. Baik
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Gehua Zhen
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, MD USA
| | - Xu Cao
- Center for Musculoskeletal Research, Department of Orthopaedic Surgery, Johns Hopkins University, Baltimore, MD USA
| | - Lynda F. Bonewald
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN USA
| | - Weiyang Jin
- Microscale Biocomplexity Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - Lance C. Kam
- Microscale Biocomplexity Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY USA
| | - X. Edward Guo
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, NY USA
| |
Collapse
|
9
|
Cao R, Xiao W, Wu X, Sun L, Pan F. Quantitative observations on cytoskeleton changes of osteocytes at different cell parts using digital holographic microscopy. BIOMEDICAL OPTICS EXPRESS 2018; 9:72-85. [PMID: 29359088 PMCID: PMC5772590 DOI: 10.1364/boe.9.000072] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/30/2017] [Accepted: 11/30/2017] [Indexed: 05/10/2023]
Abstract
Cytoskeletons such as F-actin have different distributions in different cell parts and they are the cause of different degrees of cell collapse when the F-actin is disrupted. It is challenging to use conventional methods such as fluorescence microscopy and atomic force microscopy to conduct real-time and three-dimensional observations on the dynamic processes at different cell parts due to the slow measuring speed and the need for live-cell staining. In this study, the morphological variations of different bone cell parts caused by F-actin disruption are dynamically measured by using digital holographic microscopy (DHM). We separately analyze local parameters (cell height and cell width) and global parameters (cell projected area and cell volume) of cells to address variations of specific cell areas and quantify the changing process of the whole cell. We found significant differences in temporal variations of both local and global cell parameters between the cell body and cell process, which is consistent with the qualitative observation by fluorescence staining. Our study not only validates the unique ability of DHM to simultaneously investigate the dynamic process at different cell parts, but also provides sufficient experimental bases for exploring the mechanism for F-actin disruption.
Collapse
Affiliation(s)
- Runyu Cao
- Key Laboratory of Precision Opto-Mechatronics Technology of Ministry of Education, School of Instrumentation Science & Optoelectronics Engineering, Beihang University, Beijing, 100191, China
| | - Wen Xiao
- Key Laboratory of Precision Opto-Mechatronics Technology of Ministry of Education, School of Instrumentation Science & Optoelectronics Engineering, Beihang University, Beijing, 100191, China
| | - Xintong Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Lianwen Sun
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Feng Pan
- Key Laboratory of Precision Opto-Mechatronics Technology of Ministry of Education, School of Instrumentation Science & Optoelectronics Engineering, Beihang University, Beijing, 100191, China
| |
Collapse
|
10
|
Jacobs EE, Gronowicz G, Hurley MM, Kuhn LT. Biomimetic calcium phosphate/polyelectrolyte multilayer coatings for sequential delivery of multiple biological factors. J Biomed Mater Res A 2017; 105:1500-1509. [PMID: 28002652 DOI: 10.1002/jbm.a.35985] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 11/01/2016] [Accepted: 12/14/2016] [Indexed: 12/12/2022]
Abstract
Combinations of growth factors synergistically enhance tissue regeneration, but typically require sequential, rather than co-delivery from biomaterials for maximum efficacy. Polyelectrolyte multilayer (PEM) coatings can deliver multiple factors without loss of activity; however, sequential delivery from PEM has been limited due to interlayer diffusion that results in co-delivery of the factors. This study shows that addition of a biomimetic calcium phosphate (bCaP) barrier layer to a PEM coating effectively prevents interlayer diffusion and enables sequential delivery of two different biomolecules via direct cell access. A simulated body fluid method was used to deposit a layer of bCaP followed by 30 bilayers of PEM made with poly-l-Lysine (+) and poly l-Glutamic acid (-) (bCaP-PEM). Measurements of MC3T3-E1 proliferation and viability over time on bCaP-PEM were used to demonstrate the sequential delivery kinetics of a proliferative factor [fibroblast growth factor-2 (FGF-2)] followed by a cytotoxic factor (antimycin A, AntiA). FGF-2 and AntiA both retained their bioactivity within bCaP-PEM, yet no release of FGF-2 or AntiA from bCaP-PEM was observed when cells were absent indicating a cell-mediated, local delivery process. This coating technique is useful for a variety of applications that would benefit from highly localized, sequential delivery of multiple biomolecules governed by cell initiated degradation that avoids off-target effects associated with diffusion-based release. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 1500-1509, 2017.
Collapse
Affiliation(s)
- E E Jacobs
- Reconstructive Sciences, University of Connecticut Health, Farmington, Connecticut.,Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| | - G Gronowicz
- Department of Surgery, University of Connecticut Health, Farmington, Connecticut
| | - M M Hurley
- Department of Medicine, University of Connecticut Health, Farmington, Connecticut
| | - L T Kuhn
- Reconstructive Sciences, University of Connecticut Health, Farmington, Connecticut.,Biomedical Engineering, University of Connecticut, Storrs, Connecticut
| |
Collapse
|
11
|
Brown GN, Sattler RL, Guo XE. Experimental studies of bone mechanoadaptation: bridging in vitro and in vivo studies with multiscale systems. Interface Focus 2016; 6:20150071. [PMID: 26855756 DOI: 10.1098/rsfs.2015.0071] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Despite advancements in technology and science over the last century, the mechanisms underlying Wolff's law-bone structure adaptation in response to physical stimuli-remain poorly understood, limiting the ability to effectively treat and prevent skeletal diseases. A challenge to overcome in the study of the underlying mechanisms of this principle is the multiscale nature of mechanoadaptation. While there exist in silico systems that are capable of studying across these scales, experimental studies are typically limited to interpretation at a single dimension or time point. For instance, studies of single-cell responses to defined physical stimuli offer only a limited prediction of the whole bone response, while overlapping pathways or compensatory mechanisms complicate the ability to isolate critical targets in a whole animal model. Thus, there exists a need to develop experimental systems capable of bridging traditional experimental approaches and informing existing multiscale theoretical models. The purpose of this article is to review the process of mechanoadaptation and inherent challenges in studying its underlying mechanisms, discuss the limitations of traditional experimental systems in capturing the many facets of this process and highlight three multiscale experimental systems which bridge traditional approaches and cover relatively understudied time and length scales in bone adaptation.
Collapse
Affiliation(s)
- Genevieve N Brown
- Bone Bioengineering Laboratory, Department of Biomedical Engineering , Columbia University , New York, NY 10027 , USA
| | - Rachel L Sattler
- Bone Bioengineering Laboratory, Department of Biomedical Engineering , Columbia University , New York, NY 10027 , USA
| | - X Edward Guo
- Bone Bioengineering Laboratory, Department of Biomedical Engineering , Columbia University , New York, NY 10027 , USA
| |
Collapse
|
12
|
Poellmann MJ, Estrada JB, Boudou T, Berent ZT, Franck C, Wagoner Johnson AJ. Differences in Morphology and Traction Generation of Cell Lines Representing Different Stages of Osteogenesis. J Biomech Eng 2015; 137:124503. [DOI: 10.1115/1.4031848] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Indexed: 01/10/2023]
Abstract
Osteogenesis is the process by which mesenchymal stem cells differentiate to osteoblasts and form bone. The morphology and root mean squared (RMS) traction of four cell types representing different stages of osteogenesis were quantified. Undifferentiated D1, differentiated D1, MC3T3-E1, and MLO-A5 cell types were evaluated using both automated image analysis of cells stained for F-actin and by traction force microscopy (TFM). Undifferentiated mesenchymal stem cell lines were small, spindly, and exerted low traction, while differentiated osteoblasts were large, had multiple processes, and exerted higher traction. Size, shape, and traction all correlated with the differentiation stage. Thus, cell morphology evolved and RMS traction increased with differentiation. The results provide a foundation for further work with these cell lines to study the mechanobiology of bone formation.
Collapse
Affiliation(s)
- Michael J. Poellmann
- Mem. ASME Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | | | - Thomas Boudou
- Laboratory of Materials and Physical Engineering, Grenoble Institute of Technology, Grenoble 38016, France
| | - Zachary T. Berent
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Christian Franck
- Mem. ASME School of Engineering, Brown University, Providence, RI 02912 e-mail:
| | - Amy J. Wagoner Johnson
- Mem. ASME Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, 1206 W. Green Street, Urbana, IL 61801 e-mail:
| |
Collapse
|
13
|
Preclinical models for in vitro mechanical loading of bone-derived cells. BONEKEY REPORTS 2015; 4:728. [PMID: 26331007 DOI: 10.1038/bonekey.2015.97] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 05/29/2015] [Indexed: 02/06/2023]
Abstract
It is well established that bone responds to mechanical stimuli whereby physical forces are translated into chemical signals between cells, via mechanotransduction. It is difficult however to study the precise cellular and molecular responses using in vivo systems. In vitro loading models, which aim to replicate forces found within the bone microenvironment, make the underlying processes of mechanotransduction accessible to the researcher. Direct measurements in vivo and predictive modeling have been used to define these forces in normal physiological and pathological states. The types of mechanical stimuli present in the bone include vibration, fluid shear, substrate deformation and compressive loading, which can all be applied in vitro to monolayer and three-dimensional (3D) cultures. In monolayer, vibration can be readily applied to cultures via a low-magnitude, high-frequency loading rig. Fluid shear can be applied to cultures in multiwell plates via a simple rocking platform to engender gravitational fluid movement or via a pump to cells attached to a slide within a parallel-plate flow chamber, which may be micropatterned for use with osteocytes. Substrate strain can be applied via the vacuum-driven FlexCell system or via a four-point loading jig. 3D cultures better replicate the bone microenvironment and can also be subjected to the same forms of mechanical stimuli as monolayer, including vibration, fluid shear via perfusion flow, strain or compression. 3D cocultures that more closely replicate the bone microenvironment can be used to study the collective response of several cell types to loading. This technical review summarizes the methods for applying mechanical stimuli to bone cells in vitro.
Collapse
|
14
|
Lai X, Price C, Modla S, Thompson WR, Caplan J, Kirn-Safran CB, Wang L. The dependences of osteocyte network on bone compartment, age, and disease. Bone Res 2015; 3. [PMID: 26213632 PMCID: PMC4511381 DOI: 10.1038/boneres.2015.9] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Osteocytes, the most abundant bone cells, form an interconnected network in the lacunar-canalicular pore system (LCS) buried within the mineralized matrix, which allows osteocytes to obtain nutrients from the blood supply, sense external mechanical signals, and communicate among themselves and with other cells on bone surfaces. In this study, we examined key features of the LCS network including the topological parameter and the detailed structure of individual connections and their variations in cortical and cancellous compartments, at different ages, and in two disease conditions with altered mechanosensing (perlecan deficiency and diabetes). LCS network showed both topological stability, in terms of conservation of connectivity among osteocyte lacunae (similar to the "nodes" in a computer network), and considerable variability the pericellular annular fluid gap surrounding lacunae and canaliculi (similar to the "bandwidth" of individual links in a computer network). Age, in the range of our study (15-32 weeks), affected only the pericellular fluid annulus in cortical bone but not in cancellous bone. Diabetes impacted the spacing of the lacunae, while the perlecan deficiency had a profound influence on the pericellular fluid annulus. The LCS network features play important roles in osteocyte signaling and regulation of bone growth and adaptation.
Collapse
Affiliation(s)
- Xiaohan Lai
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Christopher Price
- Biomedical Engineering Program, University of Delaware, Newark, DE, USA
| | - Shannon Modla
- DBI Bioimaging Center, University of Delaware, Newark, DE, USA
| | - William R Thompson
- Department of Physical Therapy, Indiana University, Indianapolis, IN, USA
| | - Jeffrey Caplan
- DBI Bioimaging Center, University of Delaware, Newark, DE, USA
| | | | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| |
Collapse
|
15
|
Hong MH, Choi HJ, Ko YM, Lee YK. Engineered microstructure granules for tailored drug release rate. Biotechnol Bioeng 2015; 112:1936-47. [DOI: 10.1002/bit.25595] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/24/2015] [Accepted: 03/04/2015] [Indexed: 12/19/2022]
Affiliation(s)
- Min-Ho Hong
- Department of Materials Science and Engineering; Yonsei University; Seodaemun-gu Seoul Korea
| | - Heon-Jin Choi
- Department of Materials Science and Engineering; Yonsei University; Seodaemun-gu Seoul Korea
| | - Yeong-Mu Ko
- Research Center for Oral Disease Regulation of the Aged; Chosun University School of Dentistry; 309 Pilmun-daero Dong-gu Gwangju 501-759 Korea
| | - Yong-Keun Lee
- Research Center for Oral Disease Regulation of the Aged; Chosun University School of Dentistry; 309 Pilmun-daero Dong-gu Gwangju 501-759 Korea
| |
Collapse
|
16
|
Fuse M, Hayakawa T, Hashizume-Takizawa T, Takeuchi R, Kurita-Ochiai T, Fujita-Yoshigaki J, Fukumoto M. MC3T3-E1 Cell Assay on Collagen or Fibronectin Immobilized Poly (Lactic Acid-ε-Caprolactone) Film. J HARD TISSUE BIOL 2015. [DOI: 10.2485/jhtb.24.249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Megumi Fuse
- Department of Laboratory Medicine for Dentistry, Nihon University School of Dentistry at Matsudo
| | - Tohru Hayakawa
- Department of Dental Engineering, Tsurumi University School of Dental Medicine
| | | | - Reiri Takeuchi
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo
| | | | | | - Masahiko Fukumoto
- Department of Laboratory Medicine for Dentistry, Nihon University School of Dentistry at Matsudo
| |
Collapse
|
17
|
Klein-Nulend J, van Oers RFM, Bakker AD, Bacabac RG. Bone cell mechanosensitivity, estrogen deficiency, and osteoporosis. J Biomech 2014; 48:855-65. [PMID: 25582356 DOI: 10.1016/j.jbiomech.2014.12.007] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2014] [Indexed: 11/26/2022]
Abstract
Adaptation of bone to mechanical stresses normally produces a bone architecture that combines a proper resistance against failure with a minimal use of material. This adaptive process is governed by mechanosensitive osteocytes that transduce the mechanical signals into chemical responses, i.e. the osteocytes release signaling molecules, which orchestrate the recruitment and activity of bone forming osteoblasts and/or bone resorbing osteoclasts. Computer models have shown that the maintenance of a mechanically-efficient bone architecture depends on the intensity and spatial distribution of the mechanical stimulus as well as on the osteocyte response. Osteoporosis is a condition characterized by a reduced bone mass and a compromized resistance of bone against mechanical loads, which has led us to hypothesize that mechanotransduction by osteocytes is altered in osteoporosis. One of the major causal factors for osteoporosis is the loss of estrogen, the major hormonal regulator of bone metabolism. Loss of estrogen may increase osteocyte-mediated activation of bone remodeling, resulting in impaired bone mass and architecture. In this review we highlight current insights on how osteocytes perceive mechanical stimuli placed on whole bones. Particular emphasis is placed on the role of estrogen in signaling pathway activation by mechanical stimuli, and on computer simulation in combination with cell biology to unravel biological processes contributing to bone strength.
Collapse
Affiliation(s)
- Jenneke Klein-Nulend
- Department of Oral Cell Biology, ACTA-University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands.
| | - René F M van Oers
- Department of Oral Cell Biology, ACTA-University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands; Department of Dental Materials Science, ACTA-University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Astrid D Bakker
- Department of Oral Cell Biology, ACTA-University of Amsterdam and VU University Amsterdam, MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Rommel G Bacabac
- Department of Physics, Medical Biophysics Group, University of San Carlos, Cebu City, Philippines
| |
Collapse
|
18
|
Vazquez M, Evans BAJ, Riccardi D, Evans SL, Ralphs JR, Dillingham CM, Mason DJ. A new method to investigate how mechanical loading of osteocytes controls osteoblasts. Front Endocrinol (Lausanne) 2014; 5:208. [PMID: 25538684 PMCID: PMC4260042 DOI: 10.3389/fendo.2014.00208] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 11/18/2014] [Indexed: 01/08/2023] Open
Abstract
Mechanical loading, a potent stimulator of bone formation, is governed by osteocyte regulation of osteoblasts. We developed a three-dimensional (3D) in vitro co-culture system to investigate the effect of loading on osteocyte-osteoblast interactions. MLO-Y4 cells were embedded in type I collagen gels and MC3T3-E1(14) or MG63 cells layered on top. Ethidium homodimer staining of 3D co-cultures showed 100% osteoblasts and 86% osteocytes were viable after 7 days. Microscopy revealed osteoblasts and osteocytes maintain their respective ovoid/pyriform and dendritic morphologies in 3D co-cultures. Reverse-transcriptase quantitative polymerase chain reaction (RT-qPCR) of messenger ribonucleic acid (mRNA) extracted separately from osteoblasts and osteocytes, showed that podoplanin (E11), osteocalcin, and runt-related transcription factor 2 mRNAs were expressed in both cell types. Type I collagen (Col1a1) mRNA expression was higher in osteoblasts (P < 0.001), whereas, alkaline phosphatase mRNA was higher in osteocytes (P = 0.001). Immunohistochemistry revealed osteoblasts and osteocytes express E11, type I pro-collagen, and connexin 43 proteins. In preliminary experiments to assess osteogenic responses, co-cultures were treated with human recombinant bone morphogenetic protein 2 (BMP-2) or mechanical loading using a custom built loading device. BMP-2 treatment significantly increased osteoblast Col1a1 mRNA synthesis (P = 0.031) in MLO-Y4/MG63 co-cultures after 5 days treatment. A 16-well silicone plate, loaded (5 min, 10 Hz, 2.5 N) to induce 4000-4500 με cyclic compression within gels increased prostaglandin E2 (PGE2) release 0.5 h post-load in MLO-Y4 cells pre-cultured in 3D collagen gels for 48, 72 h, or 7 days. Mechanical loading of 3D co-cultures increased type I pro-collagen release 1 and 5 days later. These methods reveal a new osteocyte-osteoblast co-culture model that may be useful for investigating mechanically induced osteocyte control of osteoblast bone formation.
Collapse
Affiliation(s)
- Marisol Vazquez
- Arthritis Research UK Biomechanics and Bioengineering Centre, School of Biosciences, Cardiff University, Cardiff, UK
| | - Bronwen A. J. Evans
- Institute of Molecular and Experimental Medicine, School of Medicine, Cardiff University, Cardiff, UK
| | - Daniela Riccardi
- Division of Pathophysiology and Repair, School of Biosciences, Cardiff University, Cardiff, UK
| | - Sam L. Evans
- Institute of Mechanical and Manufacturing Engineering, School of Engineering, Cardiff University, Cardiff, UK
| | - Jim R. Ralphs
- Division of Pathophysiology and Repair, School of Biosciences, Cardiff University, Cardiff, UK
| | | | - Deborah J. Mason
- Division of Pathophysiology and Repair, School of Biosciences, Cardiff University, Cardiff, UK
| |
Collapse
|
19
|
Solberg LB, Stang E, Brorson SH, Andersson G, Reinholt FP. Tartrate-resistant acid phosphatase (TRAP) co-localizes with receptor activator of NF-KB ligand (RANKL) and osteoprotegerin (OPG) in lysosomal-associated membrane protein 1 (LAMP1)-positive vesicles in rat osteoblasts and osteocytes. Histochem Cell Biol 2014; 143:195-207. [PMID: 25201349 PMCID: PMC4298672 DOI: 10.1007/s00418-014-1272-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2014] [Indexed: 12/19/2022]
Abstract
Tartrate-resistant acid phosphatase (TRAP) is well known as an osteoclast marker; however, a recent study from our group demonstrated enhanced number of TRAP + osteocytes as well as enhanced levels of TRAP located to intracellular vesicles in osteoblasts and osteocytes in experimental osteoporosis in rats. Such vesicles were especially abundant in osteoblasts and osteocytes in cancellous bone as well as close to bone surface and intracortical remodeling sites. To further investigate TRAP in osteoblasts and osteocytes, long bones from young, growing rats were examined. Immunofluorescence confocal microscopy displayed co-localization of TRAP with receptor activator of NF-KB ligand (RANKL) and osteoprotegerin (OPG) in hypertrophic chondrocytes and diaphyseal osteocytes with Pearson's correlation coefficient ≥0.8. Transmission electron microscopy showed co-localization of TRAP and RANKL in lysosomal-associated membrane protein 1 (LAMP1) + vesicles in osteoblasts and osteocytes supporting the results obtained by confocal microscopy. Recent in vitro data have demonstrated OPG as a traffic regulator for RANKL to LAMP1 + secretory lysosomes in osteoblasts and osteocytes, which seem to serve as temporary storage compartments for RANKL. Our in situ observations indicate that TRAP is located to RANKL-/OPG-positive secretory lysosomes in osteoblasts and osteocytes, which may have implications for osteocyte regulation of osteoclastogenesis.
Collapse
Affiliation(s)
- L B Solberg
- Department of Pathology, The Core Facility for Advanced Electron Microscopy, Oslo University Hospital, Rikshospitalet, P.O. Box 4950, Nydalen, 0424, Oslo, Norway,
| | | | | | | | | |
Collapse
|
20
|
Honma M, Ikebuchi Y, Kariya Y, Hayashi M, Hayashi N, Aoki S, Suzuki H. RANKL subcellular trafficking and regulatory mechanisms in osteocytes. J Bone Miner Res 2013; 28:1936-49. [PMID: 23529793 DOI: 10.1002/jbmr.1941] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 03/13/2013] [Accepted: 03/20/2013] [Indexed: 11/07/2022]
Abstract
The receptor activator of the NF-κB ligand (RANKL) is the central player in the regulation of osteoclastogenesis, and the quantity of RANKL presented to osteoclast precursors is an important factor determining the magnitude of osteoclast formation. Because osteoblastic cells are thought to be a major source of RANKL, the regulatory mechanisms of RANKL subcellular trafficking have been studied in osteoblastic cells. However, recent reports showed that osteocytes are a major source of RANKL presentation to osteoclast precursors, prompting a need to reinvestigate RANKL subcellular trafficking in osteocytes. Investigation of molecular mechanisms in detail needs well-designed in vitro experimental systems. Thus, we developed a novel co-culture system of osteoclast precursors and osteocytes embedded in collagen gel. Experiments using this model revealed that osteocytic RANKL is provided as a membrane-bound form to osteoclast precursors through osteocyte dendritic processes and that the contribution of soluble RANKL to the osteoclastogenesis supported by osteocytes is minor. Moreover, the regulation of RANKL subcellular trafficking, such as OPG-mediated transport of newly synthesized RANKL molecules to lysosomal storage compartments, and the release of RANKL to the cell surface upon stimulation with RANK are confirmed to be functional in osteocytes. These results provide a novel understanding of the regulation of osteoclastogenesis.
Collapse
Affiliation(s)
- Masashi Honma
- Department of Pharmacy, University of Tokyo Hospital, Faculty of Medicine, University of Tokyo, Tokyo, Japan. mhonma‐
| | | | | | | | | | | | | |
Collapse
|
21
|
Baik AD, Qiu J, Hillman EMC, Dong C, Guo XE. Simultaneous tracking of 3D actin and microtubule strains in individual MLO-Y4 osteocytes under oscillatory flow. Biochem Biophys Res Commun 2013; 431:718-23. [PMID: 23352617 DOI: 10.1016/j.bbrc.2013.01.052] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Accepted: 01/14/2013] [Indexed: 01/21/2023]
Abstract
Osteocytes in vivo experience complex fluid shear flow patterns to activate mechanotransduction pathways. The actin and microtubule (MT) cytoskeletons have been shown to play an important role in the osteocyte's biochemical response to fluid shear loading. The dynamic nature of physiologically relevant fluid flow profiles (i.e., 1Hz oscillatory flow) impedes the ability to image and study both actin and MT cytoskeletons simultaneously in the same cell with high spatiotemporal resolution. To overcome these limitations, a multi-channel quasi-3D microscopy technique was developed to track the actin and MT networks simultaneously under steady and oscillatory flow. Cells displayed high intercellular variability and intracellular cytoskeletal variability in strain profiles. Shear Exz was the predominant strain in both steady and oscillatory flows in the form of viscoelastic creep and elastic oscillations, respectively. Dramatic differences were seen in oscillatory flow, however. The actin strains displayed an oscillatory strain profile more often than the MT networks in all the strains tested and had a higher peak-to-trough strain magnitude. Taken together, the actin networks are the more responsive cytoskeletal networks in osteocytes under oscillatory flow and may play a bigger role in mechanotransduction pathway activation and regulation.
Collapse
Affiliation(s)
- Andrew D Baik
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, NY 10027, USA
| | | | | | | | | |
Collapse
|
22
|
Schweitzer MH, Zheng W, Cleland TP, Bern M. Molecular analyses of dinosaur osteocytes support the presence of endogenous molecules. Bone 2013; 52:414-23. [PMID: 23085295 DOI: 10.1016/j.bone.2012.10.010] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 10/05/2012] [Accepted: 10/06/2012] [Indexed: 11/15/2022]
Abstract
The discovery of soft, transparent microstructures in dinosaur bone consistent in morphology with osteocytes was controversial. We hypothesize that, if original, these microstructures will have molecular features in common with extant osteocytes. We present immunological and mass spectrometry evidence for preservation of proteins comprising extant osteocytes (Actin, Tubulin, PHEX, Histone H4) in osteocytes recovered from two non-avian dinosaurs. Furthermore, antibodies to DNA show localized binding to these microstructures, which also react positively with DNA intercalating stains propidium iodide (PI) and 4',6'-diamidino-2-phenylindole dihydrochloride (DAPI). Each antibody binds dinosaur cells in patterns similar to extant cells. These data are the first to support preservation of multiple proteins and to present multiple lines of evidence for material consistent with DNA in dinosaurs, supporting the hypothesis that these structures were part of the once living animals. We propose mechanisms for preservation of cells and component molecules, and discuss implications for dinosaurian cellular biology.
Collapse
Affiliation(s)
- Mary Higby Schweitzer
- Department of Marine, Earth and Atmospheric Sciences, North Carolina State University, Raleigh, NC 27695, USA.
| | | | | | | |
Collapse
|
23
|
Stern AR, Stern MM, Van Dyke ME, Jähn K, Prideaux M, Bonewald LF. Isolation and culture of primary osteocytes from the long bones of skeletally mature and aged mice. Biotechniques 2012; 52:361-73. [PMID: 22668415 PMCID: PMC3612989 DOI: 10.2144/0000113876] [Citation(s) in RCA: 148] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 05/18/2012] [Indexed: 12/20/2022] Open
Abstract
The purpose of this work was to establish a methodology to enable the isolation and study of osteocytes from skeletally mature young (4-month-old) and old (22-month-old) mice. The location of osteocytes deep within bone is ideal for their function as mechanosensors. However, this location makes the observation and study of osteocytes in vivo technically difficult. Osteocytes were isolated from murine long bones through a process of extended collagenase digestions combined with EDTA-based decalcification. A tissue homogenizer was used to reduce the remaining bone fragments to a suspension of bone particles, which were placed in culture to yield an outgrowth of osteocyte-like cells. All of the cells obtained from this outgrowth that displayed an osteocyte-like morphology stained positive for the osteocyte marker E11/GP38. The osteocyte phenotype was further confirmed by a lack of staining for alkaline phosphatase and the absence of collagen1a1 expression. The outgrowth of osteocytes also expressed additional osteocyte-specific genes such as Sost and Mepe. This technique facilitates the isolation of osteocytes from skeletally mature bone. This novel enabling methodology should prove useful in advancing our understanding of the roles mature osteocytes play in bone health and disease.
Collapse
Affiliation(s)
- Amber Rath Stern
- Mechanical Engineering and Oral Biology, University of Missouri Kansas City, Kansas City, MO, USA.
| | | | | | | | | | | |
Collapse
|
24
|
Yang N, Schindeler A, McDonald MM, Seto JT, Houweling PJ, Lek M, Hogarth M, Morse AR, Raftery JM, Balasuriya D, MacArthur DG, Berman Y, Quinlan KGR, Eisman JA, Nguyen TV, Center JR, Prince RL, Wilson SG, Zhu K, Little DG, North KN. α-Actinin-3 deficiency is associated with reduced bone mass in human and mouse. Bone 2011; 49:790-8. [PMID: 21784188 DOI: 10.1016/j.bone.2011.07.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 06/28/2011] [Accepted: 07/07/2011] [Indexed: 10/18/2022]
Abstract
Bone mineral density (BMD) is a complex trait that is the single best predictor of the risk of osteoporotic fractures. Candidate gene and genome-wide association studies have identified genetic variations in approximately 30 genetic loci associated with BMD variation in humans. α-Actinin-3 (ACTN3) is highly expressed in fast skeletal muscle fibres. There is a common null-polymorphism R577X in human ACTN3 that results in complete deficiency of the α-actinin-3 protein in approximately 20% of Eurasians. Absence of α-actinin-3 does not cause any disease phenotypes in muscle because of compensation by α-actinin-2. However, α-actinin-3 deficiency has been shown to be detrimental to athletic sprint/power performance. In this report we reveal additional functions for α-actinin-3 in bone. α-Actinin-3 but not α-actinin-2 is expressed in osteoblasts. The Actn3(-/-) mouse displays significantly reduced bone mass, with reduced cortical bone volume (-14%) and trabecular number (-61%) seen by microCT. Dynamic histomorphometry indicated this was due to a reduction in bone formation. In a cohort of postmenopausal Australian women, ACTN3 577XX genotype was associated with lower BMD in an additive genetic model, with the R577X genotype contributing 1.1% of the variance in BMD. Microarray analysis of cultured osteoprogenitors from Actn3(-/-) mice showed alterations in expression of several genes regulating bone mass and osteoblast/osteoclast activity, including Enpp1, Opg and Wnt7b. Our studies suggest that ACTN3 likely contributes to the regulation of bone mass through alterations in bone turnover. Given the high frequency of R577X in the general population, the potential role of ACTN3 R577X as a factor influencing variations in BMD in elderly humans warrants further study.
Collapse
Affiliation(s)
- Nan Yang
- Institute for Neuroscience and Muscle Research, The Children's Hospital at Westmead, Sydney 2145, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Waters KM, Jacobs JM, Gritsenko MA, Karin NJ. Regulation of gene expression and subcellular protein distribution in MLO-Y4 osteocytic cells by lysophosphatidic acid: Relevance to dendrite outgrowth. Bone 2011; 48:1328-35. [PMID: 21356339 PMCID: PMC3095666 DOI: 10.1016/j.bone.2011.02.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 02/18/2011] [Accepted: 02/23/2011] [Indexed: 01/19/2023]
Abstract
Osteoblastic and osteocytic cells are highly responsive to the lipid growth factor lysophosphatidic acid (LPA) but the mechanisms by which LPA alters bone cell functions are largely unknown. A major effect of LPA on osteocytic cells is the stimulation of dendrite membrane outgrowth, a process that we predicted to require changes in gene expression and protein distribution. We employed DNA microarrays for global transcriptional profiling of MLO-Y4 osteocytic cells grown for 6 and 24h in the presence or absence of LPA. We identified 932 transcripts that displayed statistically significant changes in abundance of at least 1.25-fold in response to LPA treatment. Gene ontology (GO) analysis revealed that the regulated gene products were linked to diverse cellular processes, including DNA repair, response to unfolded protein, ossification, protein-RNA complex assembly, and amine biosynthesis. Gene products associated with the regulation of actin microfilament dynamics displayed the most robust expression changes, and LPA-induced dendritogenesis in vitro was blocked by the stress fiber inhibitor cytochalasin D. Mass spectrometry-based proteomic analysis of MLO-Y4 cells revealed significant LPA-induced changes in the abundance of 284 proteins at 6h and 844 proteins at 24h. GO analysis of the proteomic data linked the effects of LPA to cell processes that control of protein distribution and membrane outgrowth, including protein localization, protein complex assembly, Golgi vesicle transport, cytoskeleton-dependent transport, and membrane invagination/endocytosis. Dendrites were isolated from LPA-treated MLO-Y4 cells and subjected to proteomic analysis to quantitatively assess the subcellular distribution of proteins. Sets of 129 and 36 proteins were enriched in the dendrite fraction as compared to whole cells after 6h and 24h of LPA exposure, respectively. Protein markers indicated that membranous organelles were largely excluded from the dendrites. Highly represented among the proteins with elevated abundances in dendrites were molecules that regulate cytoskeletal function, cell motility and membrane adhesion. Our combined transcriptomic/proteomic analysis of the response of MLO-Y4 osteocytic cells to LPA indicates that dendritogenesis is a membrane- and cytoskeleton-driven process with actin dynamics playing a particularly critical role.
Collapse
Affiliation(s)
- Katrina M. Waters
- Computational Biology and Bioinformatics, Pacific Northwest National Laboratory, Richland WA 99352, USA
| | - Jon M. Jacobs
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Marina A. Gritsenko
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Norman J. Karin
- Cell Biology and Biochemistry, Pacific Northwest National Laboratory, Richland WA 99352, USA
- Corresponding author: Norman J. Karin, Ph.D., Cell Biology and Biochemistry, Pacific Northwest National Laboratory, P.O. Box 999, J4-02, Richland, WA 99352, Tel: (509) 371-7303, Fax: (509) 371-7304,
| |
Collapse
|
26
|
Mota C, Puppi D, Dinucci D, Errico C, Bártolo P, Chiellini F. Dual-Scale Polymeric Constructs as Scaffolds for Tissue Engineering. MATERIALS 2011; 4:527-542. [PMID: 28880003 PMCID: PMC5448499 DOI: 10.3390/ma4030527] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2011] [Accepted: 02/25/2011] [Indexed: 11/16/2022]
Abstract
This research activity was aimed at the development of dual-scale scaffolds consisting of three-dimensional constructs of aligned poly(ε-caprolactone) (PCL) microfilaments and electrospun poly(lactic-co-glycolic acid) (PLGA) fibers. PCL constructs composed by layers of parallel microsized filaments (0/90° lay-down pattern), with a diameter of around 365 μm and interfilament distance of around 191 μm, were produced using a melt extrusion-based additive manufacturing technique. PLGA electrospun fibers with a diameter of around 1 μm were collected on top of the PCL constructs with different thicknesses, showing a certain degree of alignment. Cell culture experiments employing the MC3T3 murine preosteoblast cell line showed good cell viability and adhesion on the dual-scale scaffolds. In particular, the influence of electrospun fibers on cell morphology and behavior was evident, as well as in creating a structural bridging for cell colonization in the interfilament gap.
Collapse
Affiliation(s)
- Carlos Mota
- Laboratory of Bioactive Polymeric Materials for Biomedical and Environmental Applications (BIOlab), Department of Chemistry and Industrial Chemistry, University of Pisa, via Vecchia Livornese 1291, 56010 San Piero a Grado (Pi), Italy.
| | - Dario Puppi
- Laboratory of Bioactive Polymeric Materials for Biomedical and Environmental Applications (BIOlab), Department of Chemistry and Industrial Chemistry, University of Pisa, via Vecchia Livornese 1291, 56010 San Piero a Grado (Pi), Italy.
| | - Dinuccio Dinucci
- Laboratory of Bioactive Polymeric Materials for Biomedical and Environmental Applications (BIOlab), Department of Chemistry and Industrial Chemistry, University of Pisa, via Vecchia Livornese 1291, 56010 San Piero a Grado (Pi), Italy.
| | - Cesare Errico
- Laboratory of Bioactive Polymeric Materials for Biomedical and Environmental Applications (BIOlab), Department of Chemistry and Industrial Chemistry, University of Pisa, via Vecchia Livornese 1291, 56010 San Piero a Grado (Pi), Italy.
| | - Paulo Bártolo
- Centre for Rapid and Sustainable Product Development, Centro Empresarial da Marinha Grande, Rua de Portugal-Zona Industrial, 2430-028 Marinha Grande, Portugal.
| | - Federica Chiellini
- Laboratory of Bioactive Polymeric Materials for Biomedical and Environmental Applications (BIOlab), Department of Chemistry and Industrial Chemistry, University of Pisa, via Vecchia Livornese 1291, 56010 San Piero a Grado (Pi), Italy.
| |
Collapse
|
27
|
Application of Bioimaging to Osteocyte Biology. Clin Rev Bone Miner Metab 2010. [DOI: 10.1007/s12018-010-9077-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
28
|
Perrier A, Dumas V, Linossier MT, Fournier C, Jurdic P, Rattner A, Vico L, Guignandon A. Apatite content of collagen materials dose-dependently increases pre-osteoblastic cell deposition of a cement line-like matrix. Bone 2010; 47:23-33. [PMID: 20303420 DOI: 10.1016/j.bone.2010.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 03/08/2010] [Accepted: 03/11/2010] [Indexed: 11/26/2022]
Abstract
Bone matrix, mainly composed of type I collagen and apatite, is constantly modified during the bone remodeling process, which exposes bone cells to various proportions of mineralized collagen within bone structural units. Collagen-mineralized substrates have been shown to increase osteoblast activities. We hypothesized that such effects may be explained by a rapid secretion of specific growth factors and/or deposition of specific matrix proteins. Using MC3T3-E1 seeded for 32h on collagen substrates complexed with various apatite contents, we found that pre-osteoblasts in contact with mineralized collagen gave rise to a dose-dependent deposit of Vascular Endothelial Growth Factor-A (VEGF-A) and RGD-containing proteins such as osteopontin (OPN) and fibronectin (FN). This RGD-matrix deposition reinforced the cell adhesion to collagen-mineralized substrates. It was also observed that, on these substrates, this matrix was elaborated concomitantly to an increased cell migration, allowing a homogeneous coverage of the sample. This particular surface activation was probably done firstly to reinforce cell survival (VEGF-A) and adhesion (OPN, FN) and secondly to recruit and prepare surfaces for subsequent bone cell activity.
Collapse
Affiliation(s)
- A Perrier
- Université de Lyon, F42023, Saint-Etienne, France
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Krishnan V, Dhurjati R, Vogler EA, Mastro AM. Osteogenesis in vitro: from pre-osteoblasts to osteocytes: a contribution from the Osteobiology Research Group, The Pennsylvania State University. In Vitro Cell Dev Biol Anim 2009; 46:28-35. [PMID: 19826885 DOI: 10.1007/s11626-009-9238-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Accepted: 09/03/2009] [Indexed: 02/02/2023]
Abstract
Murine calvariae pre-osteoblasts (MC3T3-E1), grown in a novel bioreactor, proliferate into a mineralizing 3D osteoblastic tissue that undergoes progressive phenotypic maturation into osteocyte-like cells. Initially, the cells are closely packed (high cell/matrix ratio), but transform into a more mature phenotype (low cell/matrix ratio) after about 5 mo, a process that recapitulates stages of bone development observed in vivo. The cell morphology concomitantly evolves from spindle-shaped pre-osteoblasts through cobblestone-shaped osteoblasts to stellate-shaped osteocyte-like cells interconnected by many intercellular processes. Gene-expression profiles parallel cell morphological changes, up-to-and-including increased expression of osteocyte-associated genes such as E11, DMP1, and sclerostin. X-ray scattering and infrared spectroscopy of contiguous, square centimeter-scale macroscopic mineral deposits are consistent with bone hydroxyapatite, showing that bioreactor conditions can lead to ossification reminiscent of bone formation. Thus, extended-term osteoblast culture (< or =10 mo) in a bioreactor based on the concept of simultaneous growth and dialysis captures the full continuum of bone development otherwise inaccessible with conventional cell culture, resulting in an in vitro model of osteogenesis and a source of terminally differentiated osteocytes that does not require demineralization of fully formed bone.
Collapse
Affiliation(s)
- Venkatesh Krishnan
- Biochemistry and Molecular Biology, Pennsylvania State University, 431, South Frear, University Park, PA 16802, USA
| | | | | | | |
Collapse
|
30
|
Carragher NO. Profiling distinct mechanisms of tumour invasion for drug discovery: imaging adhesion, signalling and matrix turnover. Clin Exp Metastasis 2008; 26:381-97. [PMID: 18958576 DOI: 10.1007/s10585-008-9222-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 10/11/2008] [Indexed: 01/09/2023]
Abstract
Recent advances in microscopic imaging technology, fluorescent reporter reagents, 3-dimensional (3D) cell models and multiparametric image analysis have enhanced our ability to model and understand complex cell physiology. Extension of these approaches to live cell, kinetic studies allows further spatial and temporal understanding of a multitude of dynamic functional events, including tumour cell invasion. Recent in vivo and 3D in vitro studies reveal how tumour cells utilize a diverse variety of mechanisms to permit invasion through 3D tissue environments. Such high degrees of diversity and plasticity between invasion mechanisms present a significant challenge to the successful treatment of malignant cancer. This review examines how advances in time-resolved imaging has contributed to the characterization of distinct modes of invasion and their associated molecular mechanisms. Specifically, we highlight the development of fluorescent reporter molecules and their incorporation into more predictive 3D in vitro and in vivo models, to enhance mechanistic analysis of tumour invasion. We also highlight the latest advances in kinetic imaging instrumentation applicable to in vitro and in vivo models of tumour invasion. We discuss how multiparametric image analysis can be used to interpret image data generated by these approaches. We further discuss how these approaches can be integrated into drug discovery pipelines to facilitate evaluation and selection of candidate drugs and novel pharmaceutical compositions, targeting multiple invasive mechanisms.
Collapse
Affiliation(s)
- Neil O Carragher
- Advanced Science and Technology Laboratory, AstraZeneca Charnwood, Bakewell Road, Loughborough, UK.
| |
Collapse
|
31
|
Hormonal, pH, and calcium regulation of connexin 43-mediated dye transfer in osteocytes in chick calvaria. J Bone Miner Res 2008; 23:350-60. [PMID: 17997713 DOI: 10.1359/jbmr.071102] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED Gap junctional intercellular communication among osteocytes in chick calvaria, their natural 3D environment, was examined using FRAP analysis. Cell-cell communication among osteocytes in chick calvaria was mediated by Cx43 and was regulated by extracellular pH, extracellular calcium ion concentration, and PTH. INTRODUCTION The intercellular network of communication among osteocytes is mediated by gap junctions. Gap junctional intercellular communication (GJIC) is thought to play an important role in integration and synchronization of bone remodeling. We hypothesized that extracellular pH (pH(o)) and extracellular calcium ion concentration ([Ca2+](e)), both of which are dynamically altered by osteoclasts during bone remodeling, affect GJIC among osteocytes. Using fluorescence replacement after photobleaching (FRAP) analysis, we examined the effect of changes in pH(o) and [Ca2+](e) and addition of PTH on GJIC in osteocytes in chick calvaria. Additionally, we examined the role of intracellular calcium on the regulation of GJIC among osteocytes. MATERIALS AND METHODS Anti-Connexin43 (Cx43) immunolabeling was used to localize gap junctions in chick calvaria. GJIC among osteocytes in chick calvariae was assessed using FRAP. RESULTS Cx43 immunoreactivity was detected in most of the osteocyte processes. FRAP analysis showed dye-coupling among osteocytes in chick calvariae. In untreated osteocytes, fluorescence intensity recovered 43.7 +/- 2.2% within 5 min after photobleaching. Pretreatment of osteocytes with 18 alpha-GA, a reversible inhibitor of GJIC, significantly decreased fluorescence recovery to 10.7 +/- 2.2%. When pH(o) was decreased from 7.4 to 6.9, fluorescence recovery significantly decreased from 43.3 +/- 2.9% to 19.7 +/- 2.3%. Conversely, when pH(o) was increased from 7.4 to 8.0, fluorescence recovery was significantly increased to 61.9 +/- 4.5%. When [Ca2+](e) was increased from 1 to 25 mM, fluorescence recovery was significantly decreased from 47.0 +/- 6.1% to 16.1 +/- 2.1%. In bone fragments exposed to 1.0-10 nM rPTH for 3 h, replacement of fluorescence was significantly increased to 60.7 +/- 7.2%. Chelating intracellular calcium ions affected GJIC regulation by [Ca2+](e) and PTH. CONCLUSIONS Our study of cell-cell communication between osteocytes in chick calvaria showed for the first time that GJIC among osteocytes is regulated by the extracellular environment and by hormonal stimulation during bone remodeling. This method may be more biologically relevant to living bone than current methods.
Collapse
|
32
|
Marquette ML, Byerly D, Sognier M. A novel in vitro three-dimensional skeletal muscle model. In Vitro Cell Dev Biol Anim 2007; 43:255-63. [PMID: 17786532 DOI: 10.1007/s11626-007-9054-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Accepted: 07/25/2007] [Indexed: 01/21/2023]
Abstract
A novel three-dimensional (3D) skeletal muscle model composed of C2C12 mouse myoblasts is described. This model was generated by cultivating myoblasts in suspension using the rotary cell culture system (RCCS), a unique culture environment. Single-cell suspensions of myoblasts were seeded at 5 x 10(5)/ml in growth medium without exogenous support structures or substrates. Cell aggregation occurred in both RCCS and suspension control (SC) conditions within 12 h but occurred more rapidly in the SC at all time intervals examined. RCCS-cultured myoblasts fused and differentiated into a 3D construct without serum deprivation or alterations. Syncitia were quantified at 3 and 6+ d in stained thin sections. A significantly greater number of syncitia was found at 6+ d in the RCCS cultures compared to the SC. The majority of syncitia were localized to the periphery of the cell constructs for all treatments. The expression of sarcomeric myosin heavy chain (MHC) was localized at or near the periphery of the 3D construct. The majority of MHC was associated with the large cells (syncitia) of the 6+-d aggregates. These results show, for the first time, that myoblasts form syncitia and express MHC in the presence of growth factors and without the use of exogenous supports or substrates. This model test system is useful for investigating initial cell binding, myoblast fusion and syncitia formation, and differentiation processes.
Collapse
Affiliation(s)
- Michele L Marquette
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA.
| | | | | |
Collapse
|