1
|
Majumder S, Gupta A, Das A, Barui A, Das M, Chowdhury AR. Comparing the bone regeneration potential between a trabecular bone and a porous scaffold through osteoblast migration and differentiation: A multiscale approach. INTERNATIONAL JOURNAL FOR NUMERICAL METHODS IN BIOMEDICAL ENGINEERING 2024; 40:e3821. [PMID: 38637289 DOI: 10.1002/cnm.3821] [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: 06/30/2023] [Revised: 02/19/2024] [Accepted: 03/19/2024] [Indexed: 04/20/2024]
Abstract
Both cell migration and osteogenic differentiation are critical for successful bone regeneration. Therefore, understanding the mechanobiological aspects that govern these two processes is essential in designing effective scaffolds that promote faster bone regeneration. Studying these two factors at different locations is necessary to manage bone regeneration in various sections of a scaffold. Hence, a multiscale computational model was used to observe the mechanical responses of osteoblasts placed in different positions of the trabecular bone and gyroid scaffold. Fluid shear stresses in scaffolds at cell seeded locations (representing osteogenic differentiation) and strain energy densities in cells at cell substrate interface (representing cell migration) were observed as mechanical response parameters in this study. Comparison of these responses, as two critical factors for bone regeneration, between the trabecular bone and gyroid scaffold at different locations, is the overall goal of the study. This study reveals that the gyroid scaffold exhibits higher osteogenic differentiation and cell migration potential compared to the trabecular bone. However, the responses in the gyroid only mimic the trabecular bone in two out of nine positions. These findings can guide us in predicting the ideal cell seeded sites within a scaffold for better bone regeneration and in replicating a replaced bone condition by altering the physical parameters of a scaffold.
Collapse
Affiliation(s)
- Santanu Majumder
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Howrah, India
| | - Abhisek Gupta
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Howrah, India
| | - Ankita Das
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Howrah, India
| | - Ananya Barui
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Howrah, India
| | - Mitun Das
- Bioceramics and Coating Division, CSIR-Central Glass & Ceramic Research Institute, Kolkata, India
| | - Amit Roy Chowdhury
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Howrah, India
| |
Collapse
|
2
|
Wu X, Gong H, Hu X. Fluid-solid coupling numerical simulation of the effects of different doses of verapamil on cancellous bone in type 2 diabetic rats. BMC Musculoskelet Disord 2024; 25:123. [PMID: 38336651 PMCID: PMC10854077 DOI: 10.1186/s12891-024-07235-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/27/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND The purpose of this study was to investigate the effects of four different doses of verapamil on the mechanical behaviors of solid and the characteristics of fluid flow in cancellous bone of distal femur of type 2 diabetes rats under dynamic external load. METHODS Based on the micro-CT images, the finite element models of cancellous bones and fluids at distal femurs of rats in control group, diabetes group, treatment groups VER 4, VER 12, VER 24, and VER 48 (verapamil doses of 4, 12, 24, and 48 mg/kg/day, respectively) were constructed. A sinusoidal time-varying displacement load with an amplitude of 0.8 μm and a period of 1s was applied to the upper surface of the solid region. Then, fluid-solid coupling numerical simulation method was used to analyze the magnitudes and distributions of von Mises stress, flow velocity, and fluid shear stress of cancellous bone models in each group. RESULTS The results for mean values of von Mises stress, flow velocity and FSS (t = 0.25s) were as follows: their values in control group were lower than those in diabetes group; the three parameters varied with the dose of verapamil; in the four treatment groups, the values of VER 48 group were the lowest, they were the closest to control group, and they were smaller than diabetes group. Among the four treatment groups, VER 48 group had the highest proportion of the nodes with FSS = 1-3 Pa on the surface of cancellous bone, and more areas in VER 48 group were subjected to fluid shear stress of 1-3 Pa for more than half of the time. CONCLUSION It could be seen that among the four treatment groups, osteoblasts on the cancellous bone surface in the highest dose group (VER 48 group) were more easily activated by mechanical loading, and the treatment effect was the best. This study might help in understanding the mechanism of verapamil's effect on the bone of type 2 diabetes mellitus, and provide theoretical guidance for the selection of verapamil dose in the clinical treatment of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Xiaodan Wu
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - He Gong
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.
| | - Xiaorong Hu
- Key Laboratory of Biomechanics and Mechanobiology, Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| |
Collapse
|
3
|
Zhao S, Gao Y, Yang A, Gao X, Leng H, Sun L, Huo B. Fluid-solid coupling numerical simulation of entire rat caudal vertebrae under dynamic loading. Comput Methods Biomech Biomed Engin 2024:1-10. [PMID: 38231258 DOI: 10.1080/10255842.2024.2304281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024]
Abstract
Trabeculae bone undergoes directional growth along the applied force under physiological loading. The growth of bone structure relies on the coordinated interplay among osteocytes, osteoblasts, and osteoclasts. Under normal circumstances, bone remodeling maintains a state of equilibrium. Excessive bone formation can lead to osteosclerosis, while excessive bone resorption can result in osteoporosis and osteonecrosis. The investigation of the structural characteristics of trabeculae and the mechanotransduction between bone cells plays a vital role in the treatment of bone-related diseases. In this study, a fluid-solid coupling model of the entire vertebral bone was established based on micro-CT images obtained from rat tail vertebrae subjected to tensile loading experiments. The flow characteristics of bone marrow and the mechanical response of osteocytes in different regions under physiological loading were investigated. The results revealed a U-shaped distribution of wall fluid shear stress (FSS) along the longitudinal axis in trabecular bone, with higher FSS regions exhibiting greater mechanical stimulation on osteocytes. These findings elucidate a positive correlation between the mechanical microenvironment among osteocytes, osteoblasts, and osteoclasts, providing potential strategies for the prevention and treatment of bone diseases.
Collapse
Affiliation(s)
- Sen Zhao
- Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, China
| | - Yan Gao
- Institute of Artificial Intelligence in Sports, Capital University of Physical Education and Sports, Beijing, China
| | - Ailing Yang
- Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, China
| | - Xianzhi Gao
- Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Lianwen Sun
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Bo Huo
- Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, China
- Institute of Artificial Intelligence in Sports, Capital University of Physical Education and Sports, Beijing, China
| |
Collapse
|
4
|
Bi M, Yang K, Yu T, Wu G, Li Q. Cell-based mechanisms and strategies of co-culture system both in vivo and vitro for bone tissue engineering. Biomed Pharmacother 2023; 169:115907. [PMID: 37984308 DOI: 10.1016/j.biopha.2023.115907] [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: 09/09/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023] Open
Abstract
The lack of a functional vascular supply has been identified as a major challenge limiting the clinical introduction of stem cell-based bone tissue engineering (BTE) for the repair of large-volume bone defects (LVBD). Various approaches have been explored to improve the vascular supply in tissue-engineered constructs, and the development of strategies that could effectively induce the establishment of a functional vascular supply has become a major goal of BTE research. One of the state-of-the-art methods is to incorporate both angiogenic and osteogenic cells in co-culture systems. This review clarifies the key concepts involved, summarises the cell types and models used to date, and systematically evaluates their performance. We also discuss the cell-to-cell communication between these two cell types and the strategies explored in BTE constructs with angiogenic and osteogenic cells to optimise their functions. In addition, we outline unresolved issues and remaining obstacles that need to be overcome for further development in this field and eventual successful repair of LVBD.
Collapse
Affiliation(s)
- Mengning Bi
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China; Department of Orthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology; National Clinical Research Center for Oral diseases; Shanghai Key Laboratory of Stomatology; Shanghai Research Institute of Stomatology Shanghai, China
| | - Kaiwen Yang
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China; Department of Oral Surgery, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; Shanghai Key Laboratory of Stomatology &Shanghai Research Institute of Stomatology; National Clinical Research Center of Stomatology, Shanghai, China
| | - Tao Yu
- Department of Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Gang Wu
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam UMC and Academic Center for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam (VU), Amsterdam Movement Science (AMS), Amsterdam, the Netherlands; Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam (UvA) and Vrije Universiteit Amsterdam (VU), Amsterdam, the Netherlands.
| | - Qiong Li
- Department of Prosthetic Dentistry, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China.
| |
Collapse
|
5
|
Pan C, Zhang Y, Yan J, Zhou Y, Wang S, Liu X, Zhang P, Yang H. Extreme environments and human health: From the immune microenvironments to immune cells. ENVIRONMENTAL RESEARCH 2023; 236:116800. [PMID: 37527745 DOI: 10.1016/j.envres.2023.116800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/20/2023] [Accepted: 07/29/2023] [Indexed: 08/03/2023]
Abstract
Exposure to extreme environments causes specific acute and chronic physiological responses in humans. The adaptation and the physiological processes under extreme environments predominantly affect multiple functional systems of the organism, in particular, the immune system. Dysfunction of the immune system affected by several extreme environments (including hyperbaric environment, hypoxia, blast shock, microgravity, hypergravity, radiation exposure, and magnetic environment) has been observed from clinical macroscopic symptoms to intracorporal immune microenvironments. Therefore, simulated extreme conditions are engineered for verifying the main influenced characteristics and factors in the immune microenvironments. This review summarizes the responses of immune microenvironments to these extreme environments during in vivo or in vitro exposure, and the approaches of engineering simulated extreme environments in recent decades. The related microenvironment engineering, signaling pathways, molecular mechanisms, clinical therapy, and prevention strategies are also discussed.
Collapse
Affiliation(s)
- Chengwei Pan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Yuzhi Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Jinxiao Yan
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Yidan Zhou
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Sijie Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Xiru Liu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Pan Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; School of Food Science and Engineering, Shaanxi University of Science & Technology, 710021, China.
| | - Hui Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Engineering Research Center of Chinese Ministry of Education for Biological Diagnosis, Treatment and Protection Technology and Equipment, China; Research Center of Special Environmental Biomechanics & Medical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
6
|
Josephson TO, Morgan EF. Harnessing mechanical cues in the cellular microenvironment for bone regeneration. Front Physiol 2023; 14:1232698. [PMID: 37877097 PMCID: PMC10591087 DOI: 10.3389/fphys.2023.1232698] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023] Open
Abstract
At the macroscale, bones experience a variety of compressive and tensile loads, and these loads cause deformations of the cortical and trabecular microstructure. These deformations produce a variety of stimuli in the cellular microenvironment that can influence the differentiation of marrow stromal cells (MSCs) and the activity of cells of the MSC lineage, including osteoblasts, osteocytes, and chondrocytes. Mechanotransduction, or conversion of mechanical stimuli to biochemical and biological signals, is thus part of a multiscale mechanobiological process that drives bone modeling, remodeling, fracture healing, and implant osseointegration. Despite strong evidence of the influence of a variety of mechanical cues, and multiple paradigms proposed to explain the influence of these cues on tissue growth and differentiation, even a working understanding of how skeletal cells respond to the complex combinations of stimuli in their microenvironments remains elusive. This review covers the current understanding of what types of microenvironmental mechanical cues MSCs respond to and what is known about how they respond in the presence of multiple such cues. We argue that in order to realize the vast potential for harnessing the cellular microenvironment for the enhancement of bone regeneration, additional investigations of how combinations of mechanical cues influence bone regeneration are needed.
Collapse
Affiliation(s)
- Timothy O. Josephson
- Biomedical Engineering, Boston University, Boston, MA, United States
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States
| | - Elise F. Morgan
- Biomedical Engineering, Boston University, Boston, MA, United States
- Center for Multiscale and Translational Mechanobiology, Boston University, Boston, MA, United States
- Mechanical Engineering, Boston University, Boston, MA, United States
| |
Collapse
|
7
|
Zhao S, Chen Z, Li T, Sun Q, Leng H, Huo B. Numerical simulations of fluid flow in trabecular-lacunar cavities under cyclic loading. Comput Biol Med 2023; 163:107144. [PMID: 37315384 DOI: 10.1016/j.compbiomed.2023.107144] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Under external loading, the fluid shear stress (FSS) in the porous structures of bones, such as trabecular or lacunar-canalicular cavity, can influence the biological response of bone cells. However, few studies have considered both cavities. The present study investigated the characteristics of fluid flow at different scales in cancellous bone in rat femurs, as well as the effects of osteoporosis and loading frequency. METHODS Sprague Dawley rats (3 months old) were divided into normal and osteoporotic groups. A multiscale 3D fluid-solid coupling finite element model considering trabecular system and lacunar-canalicular system was established. Cyclic displacement loadings with frequencies of 1, 2, and 4 Hz were applied. FINDINGS Results showed that the wall FSS around the adhesion complexes of osteocyte on the canaliculi was higher than that on the osteocyte body. Under the same loading conditions, the wall FSS of the osteoporotic group was smaller than that of the normal group. The fluid velocity and FSS in trabecular pores exhibited a linear relationship with loading frequency. Similarly, the FSS around osteocytes also showed the loading frequency-dependent phenomenon. INTERPRETATION The high cadence in movement can effectively increase the FSS level on osteocytes for osteoporotic bone, i.e., expand the space within the bone with physiological load. This study might help in understanding the process of bone remodeling under cyclic loading and provide the fundamental data for the development of strategies for osteoporosis treatment.
Collapse
Affiliation(s)
- Sen Zhao
- Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, 100081, PR China
| | - Zebin Chen
- Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, 100081, PR China
| | - Taiyang Li
- Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, 100081, PR China
| | - Qing Sun
- Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, 100081, PR China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, 100191, PR China
| | - Bo Huo
- Biomechanics Lab, Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing, 100081, PR China; Institute of Artificial Intelligence in Sports, Capital University of Physical Education and Sports, Beijing, 100091, PR China.
| |
Collapse
|
8
|
Yamada S, Ockermann PN, Schwarz T, Mustafa K, Hansmann J. Translation of biophysical environment in bone into dynamic cell culture under flow for bone tissue engineering. Comput Struct Biotechnol J 2023; 21:4395-4407. [PMID: 37711188 PMCID: PMC10498129 DOI: 10.1016/j.csbj.2023.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/14/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Bone is a dynamic environment where osteocytes, osteoblasts, and mesenchymal stem/progenitor cells perceive mechanical cues and regulate bone metabolism accordingly. In particular, interstitial fluid flow in bone and bone marrow serves as a primary biophysical stimulus, which regulates the growth and fate of the cellular components of bone. The processes of mechano-sensory and -transduction towards bone formation have been well studied mainly in vivo as well as in two-dimensional (2D) dynamic cell culture platforms, which elucidated mechanically induced osteogenesis starting with anabolic responses, such as production of nitrogen oxide and prostaglandins followed by the activation of canonical Wnt signaling, upon mechanosensation. The knowledge has been now translated into regenerative medicine, particularly into the field of bone tissue engineering, where multipotent stem cells are combined with three-dimensional (3D) scaffolding biomaterials to produce transplantable constructs for bone regeneration. In the presence of 3D scaffolds, the importance of suitable dynamic cell culture platforms increases further not only to improve mass transfer inside the scaffolds but to provide appropriate biophysical cues to guide cell fate. In principle, the concept of dynamic cell culture platforms is rooted to bone mechanobiology. Therefore, this review primarily focuses on biophysical environment in bone and its translation into dynamic cell culture platforms commonly used for 2D and 3D cell expansion, including their advancement, challenges, and future perspectives. Additionally, it provides the literature review of recent empirical studies using 2D and 3D flow-based dynamic cell culture systems for bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Center of Translational Oral Research-Tissue Engineering, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Philipp Niklas Ockermann
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
| | - Thomas Schwarz
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
| | - Kamal Mustafa
- Center of Translational Oral Research-Tissue Engineering, Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Norway
| | - Jan Hansmann
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Germany
- Chair of Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, Germany
- Department of Electrical Engineering, University of Applied Sciences Würzburg-Schweinfurt, Germany
| |
Collapse
|
9
|
Yamada S, Yassin MA, Torelli F, Hansmann J, Green JBA, Schwarz T, Mustafa K. Unique osteogenic profile of bone marrow stem cells stimulated in perfusion bioreactor is Rho-ROCK-mediated contractility dependent. Bioeng Transl Med 2023; 8:e10509. [PMID: 37206242 PMCID: PMC10189446 DOI: 10.1002/btm2.10509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/28/2023] [Accepted: 03/04/2023] [Indexed: 03/19/2023] Open
Abstract
The fate determination of bone marrow mesenchymal stem/stromal cells (BMSC) is tightly regulated by mechanical cues, including fluid shear stress. Knowledge of mechanobiology in 2D culture has allowed researchers in bone tissue engineering to develop 3D dynamic culture systems with the potential for clinical translation in which the fate and growth of BMSC are mechanically controlled. However, due to the complexity of 3D dynamic cell culture compared to the 2D counterpart, the mechanisms of cell regulation in the dynamic environment remain relatively undescribed. In the present study, we analyzed the cytoskeletal modulation and osteogenic profiles of BMSC under fluid stimuli in a 3D culture condition using a perfusion bioreactor. BMSC subjected to fluid shear stress (mean 1.56 mPa) showed increased actomyosin contractility, accompanied by the upregulation of mechanoreceptors, focal adhesions, and Rho GTPase-mediated signaling molecules. Osteogenic gene expression profiling revealed that fluid shear stress promoted the expression of osteogenic markers differently from chemically induced osteogenesis. Osteogenic marker mRNA expression, type 1 collagen formation, ALP activity, and mineralization were promoted in the dynamic condition, even in the absence of chemical supplementation. The inhibition of cell contractility under flow by Rhosin chloride, Y27632, MLCK inhibitor peptide-18, or Blebbistatin revealed that actomyosin contractility was required for maintaining the proliferative status and mechanically induced osteogenic differentiation in the dynamic culture. The study highlights the cytoskeletal response and unique osteogenic profile of BMSC in this type of dynamic cell culture, stepping toward the clinical translation of mechanically stimulated BMCS for bone regeneration.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| | - Mohammed A. Yassin
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| | - Francesco Torelli
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| | - Jan Hansmann
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISCWürzburgGermany
- Chair of Tissue Engineering and Regenerative MedicineUniversity Hospital WürzburgWürzburgGermany
- Department of Electrical EngineeringUniversity of Applied Sciences Würzburg‐SchweinfurtSchweinfurtGermany
| | - Jeremy B. A. Green
- Centre for Craniofacial & Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial SciencesKing's College LondonUK
| | - Thomas Schwarz
- Translational Center Regenerative TherapiesFraunhofer Institute for Silicate Research ISCWürzburgGermany
| | - Kamal Mustafa
- Center of Translational Oral Research (TOR)‐Tissue Engineering Group, Department of Clinical Dentistry, Faculty of MedicineUniversity of BergenNorway
| |
Collapse
|
10
|
Tian C, Liu H, Zhao C, Zhang C, Wang W. A Numerical Study on Mechanical Effects of Low-Intensity Pulsed Ultrasound on Trabecular Bone and Osteoblasts. J Biomech Eng 2023; 145:1156067. [PMID: 36629007 DOI: 10.1115/1.4056658] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
The lack of sufficient mechanical stimulation to the human bone, results in disuse osteoporosis. Low-intensity pulsed ultrasound (LIPUS) promotes fracture healing and the treatment of disuse osteoporosis, but its biomechanical mechanism remains unknown. Simulative research on the mechanical effects of LIPUS on disuse trabecular bone and osteoblasts have been performed. The von Mises stress of disuse trabecular bone and osteoblasts obviously increased under LIPUS irradiation. The average von Mises stress of osteoblasts were two orders of magnitude higher under the irradiation of simulant LIPUS than that without LIPUS irradiation, and the von Mises stress of osteoblasts was positively correlated with the amplitude of sound pressure excitation. The results showed that LIPUS irradiation could obviously improve the mechanical micro-environment of trabecular bone and osteoblasts to alleviate the lack of mechanical stimulation. The results of the research can reveal the biomechanical mechanism of LIPUS in the treatment of disuse osteoporosis to some extent and provide theoretical guidance for clinical treatment of disuse osteoporosis through physical methods.
Collapse
Affiliation(s)
- Congbiao Tian
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin 300384, China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin 300384, China
| | - Haiying Liu
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin 300384, China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin 300384, China
| | - Chaohui Zhao
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin 300384, China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin 300384, China
| | - Chunqiu Zhang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin 300384, China; National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin 300384, China
| | - Wei Wang
- Department of Mechanics, School of Mechanical Engineering, Tianjin University, Tianjin 300354, China
| |
Collapse
|
11
|
Murakami T, Ishida T, Tanaka S, Nakayama J, Tsurugizawa T, Takahashi Y, Kato F, Kawamata M. Inflammation and subsequent nociceptor sensitization in the bone marrow are involved in an animal model of osteoarthritis pain. Life Sci 2023; 324:121736. [PMID: 37121542 DOI: 10.1016/j.lfs.2023.121736] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/21/2023] [Accepted: 04/23/2023] [Indexed: 05/02/2023]
Abstract
AIMS This study aimed to determine whether pathological changes in the bone marrow cause Osteoarthritis (OA) pain based on magnetic resonance imaging (MRI), immunohistochemistry, and electrophysiology. MAIN METHODS Adjuvant-induced arthritis (AIA) was achieved by injecting 150 μL of complete Freund's adjuvant into the right knee joints of male Sprague-Dawley rats. AIA rats were compared with saline-injected rats. KEY FINDINGS AIA significantly induced mechanical hyperalgesia and spontaneous pain in the right hind paw 1-14 days after induction. Intratibial injection of 50 μL of 1 % lidocaine significantly suppressed AIA-induced mechanical hyperalgesia (p = 0.0001) and spontaneous pain (p = 0.0006) 3 days after induction. In T2-weighted MRI, AIA induced high-signal intensity within the proximal tibial metaphysis, and the mean T2 values in this area significantly increased on days 3 (p = 0.0043) and 14 (p = 0.0012) after induction. AIA induced intraosseous edema and significantly increased the number of intraosseous granulocytes on days 3 (p < 0.0001) and 14 (p < 0.0001) after induction. The electrophysiological study on days 3-7 after induction showed significantly increased spontaneous firing rates (p = 0.0166) and evoked responses to cutaneous stimuli (brush, p < 0.0001; pinching, p = 0.0359) in the right hind paw plantar surface and intratibial stimuli (p = 0.0002) in wide-dynamic-range neurons of the spinal dorsal horn. SIGNIFICANCE Intraosseous changes caused by OA induce hypersensitivity in the sensory afferents innervating bone marrow may be involved in OA pain. Novel bone marrow-targeted therapies could be beneficial for treating OA pain.
Collapse
Affiliation(s)
- Toru Murakami
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takashi Ishida
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan.
| | - Satoshi Tanaka
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Jun Nakayama
- Department of Molecular Pathology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Tomokazu Tsurugizawa
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | - Yukari Takahashi
- Center for Neuroscience of Pain and Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
| | - Fusao Kato
- Center for Neuroscience of Pain and Department of Neuroscience, The Jikei University School of Medicine, Tokyo, Japan
| | - Mikito Kawamata
- Department of Anesthesiology and Resuscitology, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| |
Collapse
|
12
|
Xiong Z, Rouquier L, Chappard C, Bachy M, Huang X, Potier E, Bensidhoum M, Hoc T. A New Microarchitecture-Based Parameter to Predict the Micromechanical Properties of Bone Allografts. MATERIALS (BASEL, SWITZERLAND) 2023; 16:ma16093349. [PMID: 37176232 PMCID: PMC10179528 DOI: 10.3390/ma16093349] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/05/2023] [Accepted: 04/23/2023] [Indexed: 05/15/2023]
Abstract
Scaffolds are an essential component of bone tissue engineering. They provide support and create a physiological environment for cells to proliferate and differentiate. Bone allografts extracted from human donors are promising scaffolds due to their mechanical and structural characteristics. Bone microarchitecture is well known to be an important determinant of macroscopic mechanical properties, but its role at the microscopic, i.e., the trabeculae level is still poorly understood. The present study investigated linear correlations between microarchitectural parameters obtained from X-ray computed tomography (micro-CT) images of bone allografts, such as bone volume fraction (BV/TV), degree of anisotropy (DA), or ellipsoid factor (EF), and micromechanical parameters derived from micro-finite element calculations, such as mean axial strain (εz) and strain energy density (We). DAEF, a new parameter based on a linear combination of the two microarchitectural parameters DA and EF, showed a strong linear correlation with the bone mechanical characteristics at the microscopic scale. Our results concluded that the spatial distribution and the plate-and-rod structure of trabecular bone are the main determinants of the mechanical properties of bone at the microscopic level. The DAEF parameter could, therefore, be used as a tool to predict the level of mechanical stimulation at the local scale, a key parameter to better understand and optimize the mechanism of osteogenesis in bone tissue engineering.
Collapse
Affiliation(s)
- Zhuang Xiong
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, 75010 Paris, France
| | - Léa Rouquier
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, 75010 Paris, France
| | | | - Manon Bachy
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, 75010 Paris, France
- Department of Pediatric Orthopedic Surgery, Armand Trousseau Hospital, Assistance Publique-Hôpitaux de Paris, Sorbonne University, 75012 Paris, France
| | - Xingrong Huang
- Laboratory of Complex Systems, Ecole Centrale de Pékin, Beihang University, Beijing 100191, China
| | - Esther Potier
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, 75010 Paris, France
| | - Morad Bensidhoum
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, 75010 Paris, France
| | - Thierry Hoc
- Université Paris Cité, CNRS, INSERM, ENVA, B3OA, 75010 Paris, France
- Mechanical Department, MSGMGC, Ecole Centrale de Lyon, 69134 Ecully, France
| |
Collapse
|
13
|
Wang J, Sun YX, Li J. The role of mechanosensor Piezo1 in bone homeostasis and mechanobiology. Dev Biol 2023; 493:80-88. [PMID: 36368521 DOI: 10.1016/j.ydbio.2022.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 10/15/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
Bones and articular cartilage are important load-bearing tissues. The fluid flow inside the bone cells and cell interaction with the extracellular matrix serve as the mechanical cues for bones and joints. Piezo1 is an ion channel found on the cell surface of many cell types, including osteocytes and chondrocytes. It is activated in response to mechanical stimulation, which subsequently mediates a variety of signaling pathways in osteoblasts, osteocytes, and chondrocytes. Piezo1 activation in osteoblastic cells positively regulates osteogenesis, while its activation in joints mediates cartilage degradation. This review focuses on the most recent research on Piezo1 in bone development and regeneration.
Collapse
Affiliation(s)
- Jiao Wang
- Department of Anesthesiology, The First Affiliated Hospital of China Medical University, NO.155 Nanjing North Street, Shenyang City, Liaoning Province, 110000, China.
| | - Yong-Xin Sun
- Department of Rehabilitation, The First Affiliated Hospital of China Medical University, NO.155 Nanjing North Street, Shenyang City, Liaoning Province, 110000, China.
| | - Jiliang Li
- Department of Biology, Indiana University Purdue University Indianapolis, 723 West Michigan Street, SL 306, Indianapolis, IN, 46202, USA.
| |
Collapse
|
14
|
Huang XH, Zheng LQ, Dai YX, Hu SN, Ning WC, Li SM, Fan YG, Lin ZL, Huang SH. Combined computational analysis and cytology show limited depth osteogenic effect on bone defects in negative pressure wound therapy. Front Bioeng Biotechnol 2023; 11:1056707. [PMID: 36873351 PMCID: PMC9978480 DOI: 10.3389/fbioe.2023.1056707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/09/2023] [Indexed: 02/18/2023] Open
Abstract
Background: The treatment of bone defects remains a clinical challenge. The effect of negative pressure wound therapy (NPWT) on osteogenesis in bone defects has been recognized; however, bone marrow fluid dynamics under negative pressure (NP) remain unknown. In this study, we aimed to examine the marrow fluid mechanics within trabeculae by computational fluid dynamics (CFD), and to verify osteogenic gene expression, osteogenic differentiation to investigate the osteogenic depth under NP. Methods: The human femoral head is scanned using micro-CT to segment the volume of interest (VOI) trabeculae. The VOI trabeculae CFD model simulating the bone marrow cavity is developed by combining the Hypermesh and ANSYS software. The effect of trabecular anisotropy is investigated, and bone regeneration effects are simulated under NP scales of -80, -120, -160, and -200 mmHg. The working distance (WD) is proposed to describe the suction depth of the NP. Finally, gene sequence analysis, cytological experiments including bone mesenchymal stem cells (BMSCs) proliferation and osteogenic differentiation are conducted after the BMSCs are cultured under the same NP scale. Results: The pressure, shear stress on trabeculae, and marrow fluid velocity decrease exponentially with an increase in WD. The hydromechanics of fluid at any WD inside the marrow cavity can be theoretically quantified. The NP scale significantly affects the fluid properties, especially those fluid close to the NP source; however, the effect of the NP scale become marginal as WD deepens. Anisotropy of trabecular structure coupled with the anisotropic hydrodynamic behavior of bone marrow; An NP of -120 mmHg demonstrates the majority of bone formation-related genes, as well as the most effective proliferation and osteogenic differentiation of BMSCs compared to the other NP scales. Conclusion: An NP of -120 mmHg may have the optimal activated ability to promote osteogenesis, but the effective WD may be limited to a certain depth. These findings help improve the understanding of fluid mechanisms behind NPWT in treating bone defects.
Collapse
Affiliation(s)
- Xiu-Hong Huang
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Li-Qin Zheng
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue-Xing Dai
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shao-Nan Hu
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Wan-Chen Ning
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Si-Min Li
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Yue-Guang Fan
- Department of Joint Surgery, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zi-Ling Lin
- Department of Orthopedic Trauma, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shao-Hong Huang
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
15
|
Wang F, Metzner F, Osterhoff G, Zheng L, Schleifenbaum S. The role of bone marrow on the mechanical properties of trabecular bone: a systematic review. Biomed Eng Online 2022; 21:80. [PMID: 36419171 PMCID: PMC9686043 DOI: 10.1186/s12938-022-01051-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/10/2022] [Indexed: 11/26/2022] Open
Abstract
Background Accurate evaluation of the mechanical properties of trabecular bone is important, in which the internal bone marrow plays an important role. The aim of this systematic review is to investigate the roles of bone marrow on the mechanical properties of trabecular bone to better support clinical work and laboratory research. Methods A systematic review of the literature published up to June 2022 regarding the role of bone marrow on the mechanical properties of trabecular bone was performed, using PubMed and Web of Science databases. The journal language was limited to English. A total of 431 articles were selected from PubMed (n = 186), Web of Science (n = 244) databases, and other sources (n = 1). Results After checking, 38 articles were finally included in this study. Among them, 27 articles discussed the subject regarding the hydraulic stiffening of trabecular bone due to the presence of bone marrow. Nine of them investigated the effects of bone marrow on compression tests with different settings, i.e., in vitro experiments under unconfined and confined conditions, and computer model simulations. Relatively few controlled studies reported the influence of bone marrow on the shear properties of trabecular bone. Conclusion Bone marrow plays a non-neglectable role in the mechanical properties of trabecular bone, its contribution varies depending on the different loading types and test settings. To obtain the mechanical properties of trabecular bone comprehensively and accurately, the solid matrix (trabeculae) and fluid-like component (bone marrow) should be considered in parallel rather than tested separately. Supplementary Information The online version contains supplementary material available at 10.1186/s12938-022-01051-1.
Collapse
Affiliation(s)
- Fangxing Wang
- grid.9647.c0000 0004 7669 9786ZESBO - Center for Research on Musculoskeletal Systems, Department of Orthopedic Surgery, Traumatology and Plastic Surgery, Leipzig University, Semmelweisstraße 14, 04103 Leipzig, Germany ,grid.9647.c0000 0004 7669 9786Department of Orthopedic Surgery, Traumatology and Plastic Surgery, Leipzig University, Liebigstraße 20 Haus 4, 04103 Leipzig, Germany
| | - Florian Metzner
- grid.9647.c0000 0004 7669 9786ZESBO - Center for Research on Musculoskeletal Systems, Department of Orthopedic Surgery, Traumatology and Plastic Surgery, Leipzig University, Semmelweisstraße 14, 04103 Leipzig, Germany ,grid.9647.c0000 0004 7669 9786Department of Orthopedic Surgery, Traumatology and Plastic Surgery, Leipzig University, Liebigstraße 20 Haus 4, 04103 Leipzig, Germany
| | - Georg Osterhoff
- grid.9647.c0000 0004 7669 9786Department of Orthopedic Surgery, Traumatology and Plastic Surgery, Leipzig University, Liebigstraße 20 Haus 4, 04103 Leipzig, Germany
| | - Leyu Zheng
- grid.9647.c0000 0004 7669 9786Department of Orthopedic Surgery, Traumatology and Plastic Surgery, Leipzig University, Liebigstraße 20 Haus 4, 04103 Leipzig, Germany
| | - Stefan Schleifenbaum
- grid.9647.c0000 0004 7669 9786ZESBO - Center for Research on Musculoskeletal Systems, Department of Orthopedic Surgery, Traumatology and Plastic Surgery, Leipzig University, Semmelweisstraße 14, 04103 Leipzig, Germany ,grid.9647.c0000 0004 7669 9786Department of Orthopedic Surgery, Traumatology and Plastic Surgery, Leipzig University, Liebigstraße 20 Haus 4, 04103 Leipzig, Germany
| |
Collapse
|
16
|
Changes in interstitial fluid flow, mass transport and the bone cell response in microgravity and normogravity. Bone Res 2022; 10:65. [PMID: 36411278 PMCID: PMC9678891 DOI: 10.1038/s41413-022-00234-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 11/22/2022] Open
Abstract
In recent years, our scientific interest in spaceflight has grown exponentially and resulted in a thriving area of research, with hundreds of astronauts spending months of their time in space. A recent shift toward pursuing territories farther afield, aiming at near-Earth asteroids, the Moon, and Mars combined with the anticipated availability of commercial flights to space in the near future, warrants continued understanding of the human physiological processes and response mechanisms when in this extreme environment. Acute skeletal loss, more severe than any bone loss seen on Earth, has significant implications for deep space exploration, and it remains elusive as to why there is such a magnitude of difference between bone loss on Earth and loss in microgravity. The removal of gravity eliminates a critical primary mechano-stimulus, and when combined with exposure to both galactic and solar cosmic radiation, healthy human tissue function can be negatively affected. An additional effect found in microgravity, and one with limited insight, involves changes in dynamic fluid flow. Fluids provide the most fundamental way to transport chemical and biochemical elements within our bodies and apply an essential mechano-stimulus to cells. Furthermore, the cell cytoplasm is not a simple liquid, and fluid transport phenomena together with viscoelastic deformation of the cytoskeleton play key roles in cell function. In microgravity, flow behavior changes drastically, and the impact on cells within the porous system of bone and the influence of an expanding level of adiposity are not well understood. This review explores the role of interstitial fluid motion and solute transport in porous bone under two different conditions: normogravity and microgravity.
Collapse
|
17
|
Gilchrist AE, Harley BA. Engineered Tissue Models to Replicate Dynamic Interactions within the Hematopoietic Stem Cell Niche. Adv Healthc Mater 2022; 11:e2102130. [PMID: 34936239 PMCID: PMC8986554 DOI: 10.1002/adhm.202102130] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/19/2021] [Indexed: 12/19/2022]
Abstract
Hematopoietic stem cells are the progenitors of the blood and immune system and represent the most widely used regenerative therapy. However, their rarity and limited donor base necessitate the design of ex vivo systems that support HSC expansion without the loss of long-term stem cell activity. This review describes recent advances in biomaterials systems to replicate features of the hematopoietic niche. Inspired by the native bone marrow, these instructive biomaterials provide stimuli and cues from cocultured niche-associated cells to support HSC encapsulation and expansion. Engineered systems increasingly enable study of the dynamic nature of the matrix and biomolecular environment as well as the role of cell-cell signaling (e.g., autocrine feedback vs paracrine signaling between dissimilar cells). The inherent coupling of material properties, biotransport of cell-secreted factors, and cell-mediated remodeling motivate dynamic biomaterial systems as well as characterization and modeling tools capable of evaluating a temporally evolving tissue microenvironment. Recent advances in HSC identification and tracking, model-based experimental design, and single-cell culture platforms facilitate the study of the effect of constellations of matrix, cell, and soluble factor signals on HSC fate. While inspired by the HSC niche, these tools are amenable to the broader stem cell engineering community.
Collapse
Affiliation(s)
- Aidan E. Gilchrist
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Brendan A.C. Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
18
|
Del Prado-Audelo ML, Caballero-Florán IH, Mendoza-Muñoz N, Giraldo-Gomez D, Sharifi-Rad J, Patra JK, González-Torres M, Florán B, Cortes H, Leyva-Gómez G. Current progress of self-healing polymers for medical applications in tissue engineering. IRANIAN POLYMER JOURNAL 2022. [DOI: 10.1007/s13726-021-00943-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
19
|
Pereira AR, Lipphaus A, Ergin M, Salehi S, Gehweiler D, Rudert M, Hansmann J, Herrmann M. Modeling of the Human Bone Environment: Mechanical Stimuli Guide Mesenchymal Stem Cell-Extracellular Matrix Interactions. MATERIALS (BASEL, SWITZERLAND) 2021; 14:4431. [PMID: 34442954 PMCID: PMC8398413 DOI: 10.3390/ma14164431] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 02/06/2023]
Abstract
In bone tissue engineering, the design of in vitro models able to recreate both the chemical composition, the structural architecture, and the overall mechanical environment of the native tissue is still often neglected. In this study, we apply a bioreactor system where human bone-marrow hMSCs are seeded in human femoral head-derived decellularized bone scaffolds and subjected to dynamic culture, i.e., shear stress induced by continuous cell culture medium perfusion at 1.7 mL/min flow rate and compressive stress by 10% uniaxial load at 1 Hz for 1 h per day. In silico modeling revealed that continuous medium flow generates a mean shear stress of 8.5 mPa sensed by hMSCs seeded on 3D bone scaffolds. Experimentally, both dynamic conditions improved cell repopulation within the scaffold and boosted ECM production compared with static controls. Early response of hMSCs to mechanical stimuli comprises evident cell shape changes and stronger integrin-mediated adhesion to the matrix. Stress-induced Col6 and SPP1 gene expression suggests an early hMSC commitment towards osteogenic lineage independent of Runx2 signaling. This study provides a foundation for exploring the early effects of external mechanical stimuli on hMSC behavior in a biologically meaningful in vitro environment, opening new opportunities to study bone development, remodeling, and pathologies.
Collapse
Affiliation(s)
- Ana Rita Pereira
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, 97070 Wuerzburg, Germany; (A.R.P.); (M.E.)
- Bernhard-Heine-Centrum for Locomotion Research, University of Wuerzburg, 97074 Wuerzburg, Germany
| | - Andreas Lipphaus
- Biomechanics Research Group, Ruhr-University Bochum, 44801 Bochum, Germany;
| | - Mert Ergin
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, 97070 Wuerzburg, Germany; (A.R.P.); (M.E.)
- Department of Biomaterials, Center of Energy Technology und Materials Science (TAO), University of Bayreuth, 95447 Bayreuth, Germany;
| | - Sahar Salehi
- Department of Biomaterials, Center of Energy Technology und Materials Science (TAO), University of Bayreuth, 95447 Bayreuth, Germany;
| | | | - Maximilian Rudert
- Department of Orthopedic Surgery, Koenig-Ludwig-Haus, University of Wuerzburg, 97074 Wuerzburg, Germany;
| | - Jan Hansmann
- Fraunhofer Institute for Silicate Research, Translational Center for Regenerative Therapies, 97082 Wuerzburg, Germany;
| | - Marietta Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, 97070 Wuerzburg, Germany; (A.R.P.); (M.E.)
- Bernhard-Heine-Centrum for Locomotion Research, University of Wuerzburg, 97074 Wuerzburg, Germany
| |
Collapse
|
20
|
Pant A, Paul E, Niebur GL, Vahdati A. Integration of mechanics and biology in computer simulation of bone remodeling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 164:33-45. [PMID: 33965425 DOI: 10.1016/j.pbiomolbio.2021.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/27/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022]
Abstract
Bone remodeling is a complex physiological process that spans across multiple spatial and temporal scales and is regulated by both mechanical and hormonal cues. An imbalance between bone resorption and bone formation in the process of bone remodeling may lead to various bone pathologies. One powerful and non-invasive approach to gain new insights into mechano-adaptive bone remodeling is computer modeling and simulation. Recent findings in bone physiology and advances in computer modeling have provided a unique opportunity to study the integration of mechanics and biology in bone remodeling. Our objective in this review is to critically appraise recent advances and developments and discuss future research opportunities in computational bone remodeling approaches that enable integration of mechanics and cellular and molecular pathways. Based on the critical appraisal of the relevant recent published literature, we conclude that multiscale in silico integration of personalized bone mechanics and mechanobiology combined with data science and analytics techniques offer the potential to deepen our knowledge of bone remodeling and provide ample opportunities for future research.
Collapse
Affiliation(s)
- Anup Pant
- Multi-disciplinary Mechanics and Modeling Laboratory, Department of Engineering, East Carolina University, Greenville, NC 27858, USA
| | - Elliot Paul
- Multi-disciplinary Mechanics and Modeling Laboratory, Department of Engineering, East Carolina University, Greenville, NC 27858, USA
| | - Glen L Niebur
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Ali Vahdati
- Multi-disciplinary Mechanics and Modeling Laboratory, Department of Engineering, East Carolina University, Greenville, NC 27858, USA.
| |
Collapse
|
21
|
Sun Y, Yuan Y, Wu W, Lei L, Zhang L. The effects of locomotion on bone marrow mesenchymal stem cell fate: insight into mechanical regulation and bone formation. Cell Biosci 2021; 11:88. [PMID: 34001272 PMCID: PMC8130302 DOI: 10.1186/s13578-021-00601-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 05/04/2021] [Indexed: 02/06/2023] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) refer to a heterogeneous population of cells with the capacity for self-renewal. BMSCs have multi-directional differentiation potential and can differentiate into chondrocytes, osteoblasts, and adipocytes under specific microenvironment or mechanical regulation. The activities of BMSCs are closely related to bone quality. Previous studies have shown that BMSCs and their lineage-differentiated progeny (for example, osteoblasts), and osteocytes are mechanosensitive in bone. Thus, a goal of this review is to discuss how these ubiquious signals arising from mechanical stimulation are perceived by BMSCs and then how the cells respond to them. Studies in recent years reported a significant effect of locomotion on the migration, proliferation and differentiation of BMSCs, thus, contributing to our bone mass. This regulation is realized by the various intersecting signaling pathways including RhoA/Rock, IFG, BMP and Wnt signalling. The mechanoresponse of BMSCs also provides guidance for maintaining bone health by taking appropriate exercises. This review will summarize the regulatory effects of locomotion/mechanical loading on BMSCs activities. Besides, a number of signalling pathways govern MSC fate towards osteogenic or adipocytic differentiation will be discussed. The understanding of mechanoresponse of BMSCs makes the foundation for translational medicine.
Collapse
Affiliation(s)
- Yuanxiu Sun
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yu Yuan
- School of Sport and Health, Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Wei Wu
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Le Lei
- School of Kinesiology, Shanghai University of Sport, Shanghai, 200438, China
| | - Lingli Zhang
- School of Physical Education & Sports Science, South China Normal University, 55 Zhongshan Road West, Tianhe District, Guangzhou, 510631, Guangdong, China.
| |
Collapse
|
22
|
Designing Hydrogel-Based Bone-On-Chips for Personalized Medicine. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11104495] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The recent development of bone-on-chips (BOCs) holds the main advantage of requiring a low quantity of cells and material, compared to traditional In Vitro models. By incorporating hydrogels within BOCs, the culture system moved to a three dimensional culture environment for cells which is more representative of bone tissue matrix and function. The fundamental components of hydrogel-based BOCs, namely the cellular sources, the hydrogel and the culture chamber, have been tuned to mimic the hematopoietic niche in the bone aspirate marrow, cancer bone metastasis and osteo/chondrogenic differentiation. In this review, we examine the entire process of developing hydrogel-based BOCs to model In Vitro a patient specific situation. First, we provide bone biological understanding for BOCs design and then how hydrogel structural and mechanical properties can be tuned to meet those requirements. This is followed by a review on hydrogel-based BOCs, developed in the last 10 years, in terms of culture chamber design, hydrogel and cell source used. Finally, we provide guidelines for the definition of personalized pathological and physiological bone microenvironments. This review covers the information on bone, hydrogel and BOC that are required to develop personalized therapies for bone disease, by recreating clinically relevant scenarii in miniaturized devices.
Collapse
|
23
|
Hyaluronan Synthases' Expression and Activity Are Induced by Fluid Shear Stress in Bone Marrow-Derived Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms22063123. [PMID: 33803805 PMCID: PMC8003268 DOI: 10.3390/ijms22063123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/16/2021] [Accepted: 03/16/2021] [Indexed: 11/30/2022] Open
Abstract
During biomineralization, the cells generating the biominerals must be able to sense the external physical stimuli exerted by the growing mineralized tissue and change their intracellular protein composition according to these stimuli. In molluscan shell, the myosin-chitin synthases have been suggested to be the link for this communication between cells and the biomaterial. Hyaluronan synthases (HAS) belong to the same enzyme family as chitin synthases. Their product hyaluronan (HA) occurs in the bone and is supposed to have a regulatory function during bone regeneration. We hypothesize that HASes’ expression and activity are controlled by fluid-induced mechanotransduction as it is known for molluscan chitin synthases. In this study, bone marrow-derived human mesenchymal stem cells (hMSCs) were exposed to fluid shear stress of 10 Pa. The RNA transcriptome was analyzed by RNA sequencing (RNAseq). HA concentrations in the supernatants were measured by ELISA. The cellular structure of hMSCs and HAS2-overexpressing hMSCs was investigated after treatment with shear stress using confocal microscopy. Fluid shear stress upregulated the expression of genes that encode proteins belonging to the HA biosynthesis and bone mineralization pathways. The HAS activity appeared to be induced. Knowledge about the regulation mechanism governing HAS expression, trafficking, enzymatic activation and quality of the HA product in hMSCs is essential to understand the biological role of HA in the bone microenvironment.
Collapse
|
24
|
Yamada S, Yassin MA, Schwarz T, Hansmann J, Mustafa K. Induction of osteogenic differentiation of bone marrow stromal cells on 3D polyester-based scaffolds solely by subphysiological fluidic stimulation in a laminar flow bioreactor. J Tissue Eng 2021; 12:20417314211019375. [PMID: 34262684 PMCID: PMC8243246 DOI: 10.1177/20417314211019375] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 05/04/2021] [Indexed: 01/09/2023] Open
Abstract
The fatal determination of bone marrow mesenchymal stem/stromal cells (BMSC) is closely associated with mechano-environmental factors in addition to biochemical clues. The aim of this study was to induce osteogenesis in the absence of chemical stimuli using a custom-designed laminar flow bioreactor. BMSC were seeded onto synthetic microporous scaffolds and subjected to the subphysiological level of fluid flow for up to 21 days. During the perfusion, cell proliferation was significantly inhibited. There were also morphological changes, with F-actin polymerisation and upregulation of ROCK1. Notably, in BMSC subjected to flow, mRNA expression of osteogenic markers was significantly upregulated and RUNX2 was localised in the nuclei. Further, under perfusion, there was greater deposition of collagen type 1 and calcium onto the scaffolds. The results confirm that an appropriate level of fluid stimuli preconditions BMSC towards the osteoblastic lineage on 3D scaffolds in the absence of chemical stimulation, which highlights the utility of flow bioreactors in bone tissue engineering.
Collapse
Affiliation(s)
- Shuntaro Yamada
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| | - Mohammed Ahmed Yassin
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| | - Thomas Schwarz
- Fraunhofer Institute for Silicate
Research ISC, Translational Center Regenerative Therapies, Wurzburg, Bayern,
Germany
| | - Jan Hansmann
- Fraunhofer Institute for Silicate
Research ISC, Translational Center Regenerative Therapies, Wurzburg, Bayern,
Germany
- Chair of Tissue Engineering and
Regenerative Medicine, University Hospital Würzburg, Germany
- Department Electrical Engineering,
University of Applied Sciences Würzburg-Schweinfurt, Germany
| | - Kamal Mustafa
- Department of Clinical Dentistry,
Faculty of Medicine – Tissue engineering group, University of Bergen, Bergen,
Norway
| |
Collapse
|
25
|
Curtis KJ, Schiavi J, Mc Garrigle MJ, Kumar V, McNamara LM, Niebur GL. Mechanical stimuli and matrix properties modulate cancer spheroid growth in three-dimensional gelatin culture. J R Soc Interface 2020; 17:20200568. [PMID: 33323051 PMCID: PMC7811591 DOI: 10.1098/rsif.2020.0568] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022] Open
Abstract
Most patients who succumb to cancer have metastases to bone that contribute to their death. Cancer cells that metastasize to bone are regularly subjected to mechanical stimuli that may affect their proliferation, growth and protein expression. Understanding why some cancer cells thrive in this environment could provide insight into new approaches to prevent or treat metastasis to bone. We used 4T1 cells as a model of breast cancer cells, and implanted them in gelatin hydrogels with moduli of 1 or 2.7 kPa to mimic the properties of bone marrow. The constructs were subjected to either perfusion of media through the hydrogel or combined perfusion and cyclic mechanical compression for 1 h d-1 for 4 d. Controls were cultured in free-swelling conditions. The cells formed spheroids during the 4 d of culture, with larger spheroids in the statically cultured constructs than in perfusion or compressed constructs. In stiffer gelatin, smaller spheroids formed in compressed constructs than perfusion alone, while compression had no effect compared to perfusion in the softer gelatin. Immunostaining indicated that the spheroids expressed osteopontin, parathyroid hormone-related protein and fibronectin, which are all hallmarks of bone metastasis. The proliferative marker Ki67 was present in all spheroids on day 4. In the 1 kPa gelatin, Ki67 staining intensity was greater in the statically cultured, free-swelling constructs than in bioreactor culture, regardless of dynamic compression. By contrast, proliferation was higher in the compressed gelatins compared to perfusion alone in the 2.7 kPa constructs, although the spheroids were smaller, on average. This suggests the stiffer gelatin may restrict spheroid growth at the same time that it enhances mechanobiological signalling during compression. Taken together, 4T1 breast cancer cells are mechanically sensitive, and mechanical stimuli can alter their proliferation and protein expression within soft materials with mechanical properties similar to bone marrow. As such, both in vivo and in vitro models of cancer metastasis should consider the role of the mechanical environment in the bone.
Collapse
Affiliation(s)
- Kimberly J. Curtis
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, Harper Cancer Research Institute, University of Notre Dame, IN 46556, USA
| | - Jessica Schiavi
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland
| | - Myles J. Mc Garrigle
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland
| | - Vatsal Kumar
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland
| | - Laoise M. McNamara
- Mechanobiology and Medical Devices Research Group, Biomedical Engineering, College of Engineering and Informatics, National University of Ireland, Galway, Ireland
| | - Glen L. Niebur
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, Harper Cancer Research Institute, University of Notre Dame, IN 46556, USA
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, IN 46556, USA
| |
Collapse
|
26
|
Li T, Chen Z, Gao Y, Zhu L, Yang R, Leng H, Huo B. Fluid-solid coupling numerical simulation of trabecular bone under cyclic loading in different directions. J Biomech 2020; 109:109912. [PMID: 32807313 DOI: 10.1016/j.jbiomech.2020.109912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 06/19/2020] [Accepted: 06/24/2020] [Indexed: 11/16/2022]
Abstract
The structure of a bone tissue is capable of adapting to mechanical loading through the process of bone remodeling, which is regulated by osteoblasts and osteoclasts. Fluid flow within trabecular porosity under cyclic loading is one of the factors stimulating the biological response of osteoblasts and osteoclasts. However, the relation between loading directions and interstitial fluid flow was seldom studied. In the present study, a finite element model based on micro-computed tomographic reconstructions is built by using a mouse femur. Results from the fluid-solid coupling numerical simulation indicate that the loading in different directions generates a distinct distribution of von Mises stress in the bone matrix and a fluid shear stress (FSS) in the bone marrow. The loading along the physiological direction leads to a more uniform distribution of solid stress and produces an FSS level beneficial to the biological response of osteoblasts and osteoclasts compared with those along the non-physiological direction. There was a minimum threshold line of wall FSS with a specific solid stress at the bone surface, suggesting that the wall FSS is mainly induced by the solid strain. These results may offer fundamental data in understanding the mechanical environment around osteoblasts and osteoclasts and the cellular and molecular mechanisms of mechanical loading-induced bone remodeling.
Collapse
Affiliation(s)
- Taiyang Li
- Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing 100081, PR China
| | - Zebin Chen
- Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing 100081, PR China
| | - Yan Gao
- Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing 100081, PR China
| | - Lingsu Zhu
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Ruili Yang
- Laboratory of Biomimetic Nanomaterials, Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, PR China
| | - Huijie Leng
- Department of Orthopaedics, Peking University Third Hospital, Beijing 100191, PR China
| | - Bo Huo
- Department of Mechanics, School of Aerospace Engineering, Beijing Institute of Technology, Beijing 100081, PR China.
| |
Collapse
|
27
|
Chen Z, Zilberberg J, Lee W. Pumpless microfluidic device with open top cell culture under oscillatory shear stress. Biomed Microdevices 2020; 22:58. [PMID: 32833129 DOI: 10.1007/s10544-020-00515-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Here we developed a 96-well plate-based pumpless microfluidic device to mimic bidirectional oscillatory shear stress experienced by osteoblasts at the endosteal niche located at the interface between bone and bone marrow. The culture device was designed to be high-throughput with 32 open top culture chambers for convenient cell seeding and staining. Mathematical modeling was used to simulate the control of oscillatory shear stress with the peak stress in the range of 0.3 to 50 mPa. Osteoblasts, cultured under oscillatory shear stress, were found to be highly viable and significantly aligned along the direction of flow. The modeling and experimental results demonstrate for the first time that cells can be cultured under controllable oscillatory shear stress in the open top culture chamber and pumpless configurations.
Collapse
Affiliation(s)
- Zhehuan Chen
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, 1 Castle Point on Hudson, Hoboken, NJ, 07030, USA
| | - Jenny Zilberberg
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, 07110, USA
| | - Woo Lee
- Department of Chemical Engineering and Materials Science, Stevens Institute of Technology, 1 Castle Point on Hudson, Hoboken, NJ, 07030, USA.
| |
Collapse
|
28
|
Nokhbatolfoghahaei H, Bohlouli M, Adavi K, Paknejad Z, Rezai Rad M, Khani MM, Salehi-Nik N, Khojasteh A. Computational modeling of media flow through perfusion-based bioreactors for bone tissue engineering. Proc Inst Mech Eng H 2020; 234:1397-1408. [PMID: 32692276 DOI: 10.1177/0954411920944039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Bioreactor system has been used in bone tissue engineering in order to simulate dynamic nature of bone tissue environments. Perfusion bioreactors have been reported as the most efficient types of shear-loading bioreactor. Also, combination of forces, such as rotation plus perfusion, has been reported to enhance cell growth and osteogenic differentiation. Mathematical modeling using sophisticated infrastructure processes could be helpful and streamline the development of functional grafts by estimating and defining an effective range of bioreactor settings for better augmentation of tissue engineering. This study is aimed to conduct computational modeling for newly designed bioreactors in order to alleviate the time and material consuming for evaluating bioreactor parameters and effect of fluid flow hydrodynamics (various amounts of shear stress) on osteogenesis. Also, biological assessments were performed in order to validate similar parameters under implementing the perfusion or rotating and perfusion fluid motions in bioreactors' prototype. Finite element method was used to investigate the effect of hydrodynamic of fluid flow inside the bioreactors. The equations used in the simulation to calculate the velocity values and consequently the shear stress values include Navier-Stokes and Brinkman equations. It has been shown that rotational fluid motion in rotating and perfusion bioreactor produces more velocity and shear stress compared with perfusion bioreactor. Moreover, implementing the perfusion together with rotational force in rotating and perfusion bioreactors has been shown to have more cell proliferation and higher activity of alkaline phosphatase enzyme as well as formation of extra cellular matrix sheet, as an indicator of bone-like tissue formation.
Collapse
Affiliation(s)
- Hanieh Nokhbatolfoghahaei
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahboubeh Bohlouli
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Student Research Committee, Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kazem Adavi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Zahrasadat Paknejad
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Rezai Rad
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mehdi Khani
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nasim Salehi-Nik
- Department of Biomechanical Engineering, Faulty of Engineering Technology, University of Twente, Enschede, The Netherlands
| | - Arash Khojasteh
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
29
|
Schipani R, Scheurer S, Florentin R, Critchley SE, Kelly DJ. Reinforcing interpenetrating network hydrogels with 3D printed polymer networks to engineer cartilage mimetic composites. Biofabrication 2020; 12:035011. [DOI: 10.1088/1758-5090/ab8708] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
30
|
Wang L, Wang C, Wu S, Fan Y, Li X. Influence of the mechanical properties of biomaterials on degradability, cell behaviors and signaling pathways: current progress and challenges. Biomater Sci 2020; 8:2714-2733. [DOI: 10.1039/d0bm00269k] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
We have clarified the influence of the mechanical properties of biomaterials on degradability and cell response, and also mechanical design targets and approaches.
Collapse
Affiliation(s)
- Lu Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beihang University
- Beijing 100083
- China
| | - Cunyang Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beihang University
- Beijing 100083
- China
| | - Shuai Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beihang University
- Beijing 100083
- China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beihang University
- Beijing 100083
- China
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beihang University
- Beijing 100083
- China
| |
Collapse
|
31
|
Jin J, Bakker AD, Wu G, Klein-Nulend J, Jaspers RT. Physicochemical Niche Conditions and Mechanosensing by Osteocytes and Myocytes. Curr Osteoporos Rep 2019; 17:235-249. [PMID: 31428977 PMCID: PMC6817749 DOI: 10.1007/s11914-019-00522-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Bone and muscle mass increase in response to mechanical loading and biochemical cues. Bone-forming osteoblasts differentiate into early osteocytes which ultimately mature into late osteocytes encapsulated in stiff calcified matrix. Increased muscle mass originates from muscle stem cells (MuSCs) enclosed between their plasma membrane and basal lamina. Stem cell fate and function are strongly determined by physical and chemical properties of their microenvironment, i.e., the cell niche. RECENT FINDINGS The cellular niche is a three-dimensional structure consisting of extracellular matrix components, signaling molecules, and/or other cells. Via mechanical interaction with their niche, osteocytes and MuSCs are subjected to mechanical loads causing deformations of membrane, cytoskeleton, and/or nucleus, which elicit biochemical responses and secretion of signaling molecules into the niche. The latter may modulate metabolism, morphology, and mechanosensitivity of the secreting cells, or signal to neighboring cells and cells at a distance. Little is known about how mechanical loading of bone and muscle tissue affects osteocytes and MuSCs within their niches. This review provides an overview of physicochemical niche conditions of (early) osteocytes and MuSCs and how these are sensed and determine cell fate and function. Moreover, we discuss how state-of-the-art imaging techniques may enhance our understanding of these conditions and mechanisms.
Collapse
Affiliation(s)
- Jianfeng Jin
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Astrid D Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Gang Wu
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, The Netherlands
| | - Richard T Jaspers
- Laboratory for Myology, Faculty of Behavioral and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands.
| |
Collapse
|
32
|
Curtis KJ, Oberman AG, Niebur GL. Effects of mechanobiological signaling in bone marrow on skeletal health. Ann N Y Acad Sci 2019; 1460:11-24. [PMID: 31508828 DOI: 10.1111/nyas.14232] [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: 05/31/2019] [Revised: 08/09/2019] [Accepted: 08/15/2019] [Indexed: 01/27/2023]
Abstract
Bone marrow is a cellular tissue that forms within the pore space and hollow diaphysis of bones. As a tissue, its primary function is to support the hematopoietic progenitor cells that maintain the populations of both erythroid and myeloid lineage cells in the bone marrow, making it an essential element of normal mammalian physiology. However, bone's primary function is load bearing, and deformations induced by external forces are transmitted to the encapsulated marrow. Understanding the effects of these mechanical inputs on marrow function and adaptation requires knowledge of the material behavior of the marrow at multiple scales, the loads that are applied, and the mechanobiology of the cells. This paper reviews the current state of knowledge of each of these factors. Characterization of the marrow mechanical environment and its role in skeletal health and other marrow functions remains incomplete, but research on the topic is increasing, driven by interest in skeletal adaptation and the mechanobiology of cancer metastasis.
Collapse
Affiliation(s)
- Kimberly J Curtis
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana.,Advanced Diagnostics and Therapeutics Initiative, University of Notre Dame, Notre Dame, Indiana
| | - Alyssa G Oberman
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana
| | - Glen L Niebur
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana.,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana.,Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| |
Collapse
|
33
|
Yang G, Li F, Lu J, Niu Y, Dai Y, Zuo L, Tian G, Wang F. The dysplastic trochlear sulcus due to the insufficient patellar stress in growing rats. BMC Musculoskelet Disord 2019; 20:411. [PMID: 31488123 PMCID: PMC6729031 DOI: 10.1186/s12891-019-2802-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 08/29/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Developmental factors were assumed to be the key factors that influenced the morphology of femoral trochlea. This study investigated the effects of insufficient patellar stress after birth on the morphological development of the femoral trochlea. Effects of insufficient patellar stress on femoral trochlea were investigated using surgical induced patellectomy and patellar dislocation in growing rat model. METHODS In this study, two experimental groups and one sham group (SG) were established. Thirty-six Wistar rats (female, 28 days of age) were randomly assigned to three groups. The patellectomy group (PG), rats underwent the patellectomy in this group. The dislocation group (DG), rats underwent the surgery-induced patellar dislocation. Histological staining (Safranin-O and fast green), Micro-computed tomographic (Micro-CT) analysis in two experimental endpoints (3, 12 weeks postoperatively) were selected to evaluate morphological changes of the femoral trochlea. RESULTS Articular cartilage on the trochlear sulcus was remodeled at 3 weeks after the surgery, and degenerated at 12 weeks through the histological staining. The femoral trochlear angle (FTA) did not show a significant difference at 3 week between the experimental groups and the sham group (PG vs SG P = 0.38, DG vs SG p = 0.05), but the FTA was significantly increased in experimental groups at 12 weeks(PG vs SG P = 0.001, DG vs SG p = 0.005). The Bone volume density (BV/TV), Trabecular number (Tb.N) under the trochlea groove were significantly reduced at 3 weeks postoperatively in the experimental groups (PG vs SG p = 0.001, DG vs SG p = 0.002). No significant difference was found in BV/TV and Tb. N among the three groups at 12 weeks postoperatively. CONCLUSION Surgical induced patellectomy and patellar dislocation leads to the dysplastic trochlear sulcus in growing rats. Besides the bone morphology of trochlear sulcus, the articular cartilage and subchondral trabecula under the trochlear sulcus were remodeled early stage after the surgery.
Collapse
Affiliation(s)
- Guangmin Yang
- Department of joint surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Faquan Li
- Department of joint surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Jiangfeng Lu
- Department of joint surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Yingzhen Niu
- Department of joint surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Yike Dai
- Department of joint surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Lixiong Zuo
- Department of joint surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Gengshuang Tian
- Department of joint surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China
| | - Fei Wang
- Department of joint surgery, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, Hebei, China.
| |
Collapse
|
34
|
Shear Stress in Bone Marrow has a Dose Dependent Effect on cFos Gene Expression in In Situ Culture. Cell Mol Bioeng 2019; 12:559-568. [PMID: 33281987 DOI: 10.1007/s12195-019-00594-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 08/20/2019] [Indexed: 12/26/2022] Open
Abstract
Introduction Mechanical stimulation of bone is necessary to maintain its mass and architecture. Osteocytes within the mineralized matrix are sensors of mechanical deformation of the hard tissue, and communicate with cells in the marrow to regulate bone remodeling. However, marrow cells are also subjected to mechanical stress during whole bone loading, and may contribute to mechanically regulated bone physiology. Previous results from our laboratory suggest that mechanotransduction in marrow cells is sufficient to cause bone formation in the absence of osteocyte signaling. In this study, we investigated whether bone formation and altered marrow cell gene expression response to stimulation was dependent on the shear stress imparted on the marrow by our loading regime. Methods Porcine trabecular bone explants were cultured in an in situ bioreactor for 5 or 28 days with stimulation twice daily. Gene expression and bone formation were quantified and compared to unstimulated controls. Correlation was used to assess the dependence on shear stress imparted by the loading regime calculated using computational fluid dynamics models. Results Vibratory stimulation resulted in a higher trabecular bone formation rate (p = 0.01) and a greater increase in bone volume fraction (p = 0.02) in comparison to control explants. Marrow cell expression of cFos increased with the calculated marrow shear stress in a dose-dependent manner (p = 0.002). Conclusions The results suggest that the shear stress due to interactions between marrow cells induces a mechanobiological response. Identification of marrow cell mechanotransduction pathways is essential to understand healthy and pathological bone adaptation and remodeling.
Collapse
|
35
|
Talebian S, Mehrali M, Taebnia N, Pennisi CP, Kadumudi FB, Foroughi J, Hasany M, Nikkhah M, Akbari M, Orive G, Dolatshahi‐Pirouz A. Self-Healing Hydrogels: The Next Paradigm Shift in Tissue Engineering? ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1801664. [PMID: 31453048 PMCID: PMC6702654 DOI: 10.1002/advs.201801664] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 03/04/2019] [Indexed: 05/18/2023]
Abstract
Given their durability and long-term stability, self-healable hydrogels have, in the past few years, emerged as promising replacements for the many brittle hydrogels currently being used in preclinical or clinical trials. To this end, the incompatibility between hydrogel toughness and rapid self-healing remains unaddressed, and therefore most of the self-healable hydrogels still face serious challenges within the dynamic and mechanically demanding environment of human organs/tissues. Furthermore, depending on the target tissue, the self-healing hydrogels must comply with a wide range of properties including electrical, biological, and mechanical. Notably, the incorporation of nanomaterials into double-network hydrogels is showing great promise as a feasible way to generate self-healable hydrogels with the above-mentioned attributes. Here, the recent progress in the development of multifunctional and self-healable hydrogels for various tissue engineering applications is discussed in detail. Their potential applications within the rapidly expanding areas of bioelectronic hydrogels, cyborganics, and soft robotics are further highlighted.
Collapse
Affiliation(s)
- Sepehr Talebian
- Intelligent Polymer Research InstituteARC Centre of Excellence for Electromaterials ScienceAIIM FacilityUniversity of WollongongNSW2522Australia
- Illawarra Health and Medical Research InstituteUniversity of WollongongWollongongNSW2522Australia
| | - Mehdi Mehrali
- DTU NanotechCenter for Intestinal Absorption and Transport of BiopharmaceuticalsTechnical University of DenmarkLyngby2800KgsDenmark
| | - Nayere Taebnia
- DTU NanotechCenter for Intestinal Absorption and Transport of BiopharmaceuticalsTechnical University of DenmarkLyngby2800KgsDenmark
| | - Cristian Pablo Pennisi
- Laboratory for Stem Cell ResearchDepartment of Health Science and TechnologyAalborg UniversityFredrik Bajers vej 3B9220AalborgDenmark
| | - Firoz Babu Kadumudi
- DTU NanotechCenter for Intestinal Absorption and Transport of BiopharmaceuticalsTechnical University of DenmarkLyngby2800KgsDenmark
| | - Javad Foroughi
- Intelligent Polymer Research InstituteARC Centre of Excellence for Electromaterials ScienceAIIM FacilityUniversity of WollongongNSW2522Australia
- Illawarra Health and Medical Research InstituteUniversity of WollongongWollongongNSW2522Australia
| | - Masoud Hasany
- DTU NanotechCenter for Intestinal Absorption and Transport of BiopharmaceuticalsTechnical University of DenmarkLyngby2800KgsDenmark
| | - Mehdi Nikkhah
- School of Biological Health and Systems Engineering (SBHSE)Arizona State UniversityTempeAZ85287USA
| | - Mohsen Akbari
- Laboratory for Innovations in MicroEngineering (LiME)Department of Mechanical EngineeringUniversity of VictoriaVictoriaBCV8P 5C2Canada
- Center for Biomedical ResearchUniversity of Victoria3800VictoriaCanada
- Center for Advanced Materials and Related TechnologiesUniversity of Victoria3800VictoriaCanada
| | - Gorka Orive
- NanoBioCel GroupLaboratory of PharmaceuticsSchool of PharmacyUniversity of the Basque Country UPV/EHUPaseo de la Universidad 701006Vitoria‐GasteizSpain
- Biomedical Research Networking Centre in BioengineeringBiomaterials, and Nanomedicine (CIBER‐BBN)Vitoria‐Gasteiz28029Spain
- University Institute for Regenerative Medicine and Oral Implantology – UIRMI (UPV/EHU‐Fundación Eduardo Anitua)Vitoria01007Spain
- BTI Biotechnology InstituteVitoria01007Spain
| | - Alireza Dolatshahi‐Pirouz
- DTU NanotechCenter for Intestinal Absorption and Transport of BiopharmaceuticalsTechnical University of DenmarkLyngby2800KgsDenmark
- Department of Dentistry‐Regenerative BiomaterialsRadboud University Medical CenterPhilips van Leydenlaan 25Nijmegen6525EXThe Netherlands
| |
Collapse
|
36
|
Falcinelli C, Di Martino A, Gizzi A, Vairo G, Denaro V. Mechanical behavior of metastatic femurs through patient-specific computational models accounting for bone-metastasis interaction. J Mech Behav Biomed Mater 2019; 93:9-22. [PMID: 30738327 DOI: 10.1016/j.jmbbm.2019.01.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/23/2019] [Accepted: 01/27/2019] [Indexed: 12/21/2022]
Abstract
This paper proposes a computational model based on a finite-element formulation for describing the mechanical behavior of femurs affected by metastatic lesions. A novel geometric/constitutive description is introduced by modelling healthy bone and metastases via a linearly poroelastic constitutive approach. A Gaussian-shaped graded transition of material properties between healthy and metastatic tissues is prescribed, in order to account for the bone-metastasis interaction. Loading-induced failure processes are simulated by implementing a progressive damage procedure, formulated via a quasi-static displacement-driven incremental approach, and considering both a stress- and a strain-based failure criterion. By addressing a real clinical case, left and right patient-specific femur models are geometrically reconstructed via an ad-hoc imaging procedure and embedding multiple distributions of metastatic lesions along femurs. Significant differences in fracture loads, fracture mechanisms, and damage patterns, are highlighted by comparing the proposed constitutive description with a purely elastic formulation, where the metastasis is treated as a pseudo-healthy tissue or as a void region. Proposed constitutive description allows to capture stress/strain localization mechanisms within the metastatic tissue, revealing the model capability in describing possible strain-induced mechano-biological stimuli driving onset and evolution of the lesion. The proposed approach opens towards the definition of effective computational strategies for supporting clinical decision and treatments regarding metastatic femurs, contributing also to overcome some limitations of actual standards and procedures.
Collapse
Affiliation(s)
- Cristina Falcinelli
- Department of Engineering, Campus Bio-Medico University of Rome, Italy; Department of Civil Engineering & Computer Science, University of Rome "Tor Vergata", Italy
| | - Alberto Di Martino
- Department of Orthopaedics and Trauma Surgery, Campus Bio-Medico University of Rome, Italy; Sideny Kimmel Medical College, Thomas Jefferson University (SKMC), Philadelphia, USA
| | - Alessio Gizzi
- Department of Engineering, Campus Bio-Medico University of Rome, Italy
| | - Giuseppe Vairo
- Department of Civil Engineering & Computer Science, University of Rome "Tor Vergata", Italy.
| | - Vincenzo Denaro
- Department of Orthopaedics and Trauma Surgery, Campus Bio-Medico University of Rome, Italy
| |
Collapse
|
37
|
Wang L, Wu S, Cao G, Fan Y, Dunne N, Li X. Biomechanical studies on biomaterial degradation and co-cultured cells: mechanisms, potential applications, challenges and prospects. J Mater Chem B 2019; 7:7439-7459. [DOI: 10.1039/c9tb01539f] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This review provides a comprehensive overview of biomechanical studies on biomaterial degradation and co-cultured cells as well as valuable biomechanical ideas on how to design or optimize cell biomaterial co-culture system.
Collapse
Affiliation(s)
- Lu Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beihang University
- Beijing 100083
- China
| | - Shuai Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beihang University
- Beijing 100083
- China
| | - Guangxiu Cao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beihang University
- Beijing 100083
- China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beihang University
- Beijing 100083
- China
| | - Nicholas Dunne
- Centre for Medical Engineering Research
- School of Mechanical and Manufacturing Engineering
- Dublin City University
- Dublin 9
- Ireland
| | - Xiaoming Li
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education
- School of Biological Science and Medical Engineering
- Beihang University
- Beijing 100083
- China
| |
Collapse
|
38
|
Paul GR, Malhotra A, Müller R. Mechanical Stimuli in the Local In Vivo Environment in Bone: Computational Approaches Linking Organ-Scale Loads to Cellular Signals. Curr Osteoporos Rep 2018; 16:395-403. [PMID: 29915967 PMCID: PMC6579731 DOI: 10.1007/s11914-018-0448-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Connecting organ-scale loads to cellular signals in their local in vivo environment is a current challenge in the field of bone (re)modelling. Understanding this critical missing link would greatly improve our ability to anticipate mechanotransduction during different modes of stimuli and the resultant cellular responses. This review characterises computational approaches that could enable coupling links across the multiple scales of bone. RECENT FINDINGS Current approaches using strain and fluid shear stress concepts have begun to link organ-scale loads to cellular signals; however, these approaches fail to capture localised micro-structural heterogeneities. Furthermore, models that incorporate downstream communication from osteocytes to osteoclasts, bone-lining cells and osteoblasts, will help improve the understanding of (re)modelling activities. Incorporating this potentially key information in the local in vivo environment will aid in developing multiscale models of mechanotransduction that can predict or help describe resultant biological events related to bone (re)modelling. Progress towards multiscale determination of the cell mechanical environment from organ-scale loads remains elusive. Construction of organ-, tissue- and cell-scale computational models that include localised environmental variation, strain amplification and intercellular communication mechanisms will ultimately help couple the hierarchal levels of bone.
Collapse
Affiliation(s)
- Graeme R Paul
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zürich, Switzerland
| | - Angad Malhotra
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zürich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zürich, Switzerland.
| |
Collapse
|
39
|
Curtis KJ, Coughlin TR, Mason DE, Boerckel JD, Niebur GL. Bone marrow mechanotransduction in porcine explants alters kinase activation and enhances trabecular bone formation in the absence of osteocyte signaling. Bone 2018; 107:78-87. [PMID: 29154967 DOI: 10.1016/j.bone.2017.11.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 01/12/2023]
Abstract
Bone is a dynamic tissue that can adapt its architecture in response to mechanical signals under the control of osteocytes, which sense mechanical deformation of the mineralized bone. However, cells in the marrow are also mechanosensitive and may contribute to load-induced bone adaptation, as marrow is subjected to mechanical stress during bone deformation. We investigated the contribution of mechanotransduction in marrow cells to trabecular bone formation by applying low magnitude mechanical stimulation (LMMS) to porcine vertebral trabecular bone explants in an in situ bioreactor. The bone formation rate was higher in stimulated explants compared to unloaded controls which represent a disuse condition (CNT). However, sclerostin protein expression in osteocytes was not different between groups, nor was expression of osteocytic mechanoregulatory genes SOST, IGF-1, CTGF, and Cyr61, suggesting the mechanoregulatory program of osteocytes was unaffected by the loading regime. In contrast, c-Fos, a gene indicative of mechanical stimulation, was upregulated in the marrow cells of mechanically stimulated explants, while the level of activated c-Jun decreased by 25%. The activator protein 1 (AP-1) transcription factor is a heterodimer of c-Fos and c-Jun, which led us to investigate the expression of the downstream target gene cyclin-D1, a gene associated with cell cycle progression and osteogenesis. Cyclin-D1 gene expression in the stimulated marrow was approximately double that of the controls. The level of phosphorylated PYK2, a purported inhibitor of osteoblast differentiation, also decreased in marrow cells from stimulated explants. Taken together, mechanotransduction in marrow cells induced trabecular bone formation independent of osteocyte signaling. Identifying the specific cells and signaling pathways involved, and verifying them with inhibition of specific signaling molecules, could lead to potential therapeutic targets for diseases characterized by bone loss.
Collapse
Affiliation(s)
- Kimberly J Curtis
- Tissue Mechanics Laboratory, University of Notre Dame, IN, 46556, USA; Bioengineering Graduate Program, University of Notre Dame, IN, 46556, USA
| | - Thomas R Coughlin
- Tissue Mechanics Laboratory, University of Notre Dame, IN, 46556, USA; Bioengineering Graduate Program, University of Notre Dame, IN, 46556, USA
| | - Devon E Mason
- Bioengineering Graduate Program, University of Notre Dame, IN, 46556, USA
| | - Joel D Boerckel
- Bioengineering Graduate Program, University of Notre Dame, IN, 46556, USA
| | - Glen L Niebur
- Tissue Mechanics Laboratory, University of Notre Dame, IN, 46556, USA; Bioengineering Graduate Program, University of Notre Dame, IN, 46556, USA.
| |
Collapse
|
40
|
Scheuren A, Wehrle E, Flohr F, Müller R. Bone mechanobiology in mice: toward single-cell in vivo mechanomics. Biomech Model Mechanobiol 2017; 16:2017-2034. [PMID: 28735414 DOI: 10.1007/s10237-017-0935-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 07/11/2017] [Indexed: 01/27/2023]
Abstract
Mechanically driven bone (re)modeling is a multiscale process mediated through complex interactions between multiple cell types and their microenvironments. However, the underlying mechanisms of how cells respond to mechanical signals are still unclear and are at the focus of the field of bone mechanobiology. Traditionally, this complex process has been addressed by reducing the system to single scales and cell types. It is only recently that more integrative approaches have been established to study bone mechanobiology across multiple scales in which mechanical load at the organ level is related to molecular responses at the cellular level. The availability of mouse loading models and imaging techniques with improved spatial and temporal resolution has made it possible to track dynamic bone (re)modeling at the tissue and cellular level in vivo. Coupled with advanced computational models, the (re)modeling activities at the tissue scale can be associated with the mechanical microenvironment. However, methods are lacking to link the molecular responses of different cell types to their local mechanical microenvironment and bone (re)modeling activities occurring at the tissue scale. With recent improvements in "omics" technologies and single-cell molecular biology, it is now possible to sequence the complete genome and transcriptome of single cells. These technologies offer unique opportunities to comprehensively investigate the cellular transcriptional profiles within their specific microenvironment. By combining single-cell "omics" technologies with well-established tissue-scale models of bone mechanobiology, we propose a mechanomics approach to locally analyze the transcriptome of single cells with respect to their local 3D mechanical in vivo environment.
Collapse
Affiliation(s)
- Ariane Scheuren
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Felicitas Flohr
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Leopold-Ruzicka-Weg 4, 8093, Zurich, Switzerland.
| |
Collapse
|
41
|
Rödling L, Schwedhelm I, Kraus S, Bieback K, Hansmann J, Lee-Thedieck C. 3D models of the hematopoietic stem cell niche under steady-state and active conditions. Sci Rep 2017; 7:4625. [PMID: 28676663 PMCID: PMC5496931 DOI: 10.1038/s41598-017-04808-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/22/2017] [Indexed: 12/11/2022] Open
Abstract
Hematopoietic stem cells (HSCs) in the bone marrow are able to differentiate into all types of blood cells and supply the organism each day with billions of fresh cells. They are applied to cure hematological diseases such as leukemia. The clinical need for HSCs is high and there is a demand for being able to control and multiply HSCs in vitro. The hematopoietic system is highly proliferative and thus sensitive to anti-proliferative drugs such as chemotherapeutics. For many of these drugs suppression of the hematopoietic system is the dose-limiting toxicity. Therefore, biomimetic 3D models of the HSC niche that allow to control HSC behavior in vitro and to test drugs in a human setting are relevant for the clinics and pharmacology. Here, we describe a perfused 3D bone marrow analog that allows mimicking the HSC niche under steady-state and activated conditions that favor either HSC maintenance or differentiation, respectively, and allows for drug testing.
Collapse
Affiliation(s)
- Lisa Rödling
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Ivo Schwedhelm
- Institute for Tissue Engineering and Regenerative Medicine, University of Würzburg, 97070, Würzburg, Germany
| | - Saskia Kraus
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology Mannheim, Medical Faculty Mannheim, Heidelberg University; German Red Cross Blood Donor Service Baden-Württemberg-Hessen, 68167, Mannheim, Germany
| | - Jan Hansmann
- Institute for Tissue Engineering and Regenerative Medicine, University of Würzburg, 97070, Würzburg, Germany
| | - Cornelia Lee-Thedieck
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany.
| |
Collapse
|
42
|
Kreipke TC, Niebur GL. Anisotropic Permeability of Trabecular Bone and its Relationship to Fabric and Architecture: A Computational Study. Ann Biomed Eng 2017; 45:1543-1554. [DOI: 10.1007/s10439-017-1805-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/28/2017] [Indexed: 11/30/2022]
|
43
|
Mechanical stimuli of trabecular bone in osteoporosis: A numerical simulation by finite element analysis of microarchitecture. J Mech Behav Biomed Mater 2017; 66:19-27. [DOI: 10.1016/j.jmbbm.2016.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/15/2016] [Accepted: 10/13/2016] [Indexed: 01/08/2023]
|
44
|
Yang H, Embry RE, Main RP. Effects of Loading Duration and Short Rest Insertion on Cancellous and Cortical Bone Adaptation in the Mouse Tibia. PLoS One 2017; 12:e0169519. [PMID: 28076363 PMCID: PMC5226737 DOI: 10.1371/journal.pone.0169519] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 12/19/2016] [Indexed: 11/19/2022] Open
Abstract
The skeleton's osteogenic response to mechanical loading can be affected by loading duration and rest insertion during a series of loading events. Prior animal loading studies have shown that the cortical bone response saturates quickly and short rest insertions between load cycles can enhance cortical bone formation. However, it remains unknown how loading duration and short rest insertion affect load-induced osteogenesis in the mouse tibial compressive loading model, and particularly in cancellous bone. To address this issue, we applied cyclic loading (-9 N peak load; 4 Hz) to the tibiae of three groups of 16 week-old female C57BL/6 mice for two weeks, with a different number of continuous load cycles applied daily to each group (36, 216 and 1200). A fourth group was loaded under 216 daily load cycles with a 10 s rest insertion after every fourth cycle. We found that as few as 36 load cycles per day were able to induce osteogenic responses in both cancellous and cortical bone. Furthermore, while cortical bone area and thickness continued to increase through 1200 cycles, the incremental increase in the osteogenic response decreased as load number increased, indicating a reduced benefit of the increasing number of load cycles. In the proximal metaphyseal cancellous bone, trabecular thickness increased with load up to 216 cycles. We also found that insertion of a 10 s rest between load cycles did not improve the osteogenic response of the cortical or cancellous tissues compared to continuous loading in this model given the age and sex of the mice and the loading parameters used here. These results suggest that relatively few load cycles (e.g. 36) are sufficient to induce osteogenic responses in both cortical and cancellous bone in the mouse tibial loading model. Mechanistic studies using the mouse tibial loading model to examine bone formation and skeletal mechanobiology could be accomplished with relatively few load cycles.
Collapse
Affiliation(s)
- Haisheng Yang
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Rachel E. Embry
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, United States of America
| | - Russell P. Main
- Musculoskeletal Biology and Mechanics Lab, Department of Basic Medical Sciences, Purdue University, West Lafayette, Indiana, United States of America
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
45
|
Altered architecture and cell populations affect bone marrow mechanobiology in the osteoporotic human femur. Biomech Model Mechanobiol 2016; 16:841-850. [DOI: 10.1007/s10237-016-0856-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 11/11/2016] [Indexed: 10/20/2022]
|