1
|
Artamonov MY, Sokov EL, Kornilova LE, Minenko IA. The Intraosseous Environment: Physiological Parameters, Regulatory Mechanisms, and Emerging Insights in Bone Biology. Int J Mol Sci 2025; 26:3876. [PMID: 40332558 PMCID: PMC12027868 DOI: 10.3390/ijms26083876] [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: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/12/2025] [Indexed: 05/08/2025] Open
Abstract
The intraosseous environment is a dynamic and intricate system integral to bone health, encompassing vascular, cellular, and biochemical interactions that drive key processes such as hematopoiesis, bone remodeling, and systemic mineral regulation. This review examines the structural composition of the bone matrix, the diverse cellular landscape, and the interconnected vascular and nervous networks, highlighting their roles in preserving bone function and responding to pathological changes. Recent studies reveal regulatory mechanisms involving oxygen tension, ionic balance, signaling molecules, and mechanotransduction pathways that shape bone metabolism and its adaptation to mechanical forces. Insights into the bone microenvironment's metabolic shifts in cancer and its interaction with inflammation underscore its pivotal role in disease progression and therapeutic innovation. Additionally, advances in imaging techniques and biomaterials fuel progress in bone regeneration and understanding its microenvironment. Exploring the intricate physiochemical dynamics and regulatory networks within the intraosseous system unlocks potential clinical applications in bone diseases, tissue engineering, and systemic metabolic disorders. This comprehensive review bridges fundamental science with translational research, offering insights into the complex yet essential intraosseous environment.
Collapse
Affiliation(s)
- Mikhail Yu. Artamonov
- Department of Physical Medicine and Rehabilitation, Penn Medicine Princeton Health, Plainsboro, NJ 08536, USA
| | - Evgeniy L. Sokov
- Department of Algology, Peoples’ Friendship University, Moscow 117198, Russia
| | | | - Inessa A. Minenko
- Department of Sports Medicine, Sechenov Medical University, Moscow 119991, Russia;
| |
Collapse
|
2
|
Orita K, Fujibayashi S, Okuzu Y, Yamaguchi S, Goto K, Otsuki B, Kawai T, Shimizu T, Hayashi M, Ikeda N, Takaoka Y, Honda S, Ikezaki T, Matsuda S. Biocompatibility and antibacterial activity of strontium and silver ion-releasing titanium with high silver treatment concentration. J Biomater Appl 2025:8853282251329276. [PMID: 40112347 DOI: 10.1177/08853282251329276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
To overcome problems associated with surgical site infection and implant loosening, we developed a titanium (Ti)-based material employing a modified alkaline heat treatment that releases strontium (Sr) and silver (Ag) ions (CaSrAg-Ti). In this study, to determine the optimal Ag treatment concentration, we prepared four different materials-commercially pure Ti (cp-Ti) as a negative control, CaSr1mMAg-Ti, CaSr10mMAg-Ti, and CaSr50mMAg-Ti. Ion release test was performed by immersing the prepared disks in fetal bovine serum. With increased loading of Ag ions, the amount of released ions increased. Colony-forming unit count assay was performed using methicillin-susceptible Staphylococcus aureus and Escherichia coli. High antibacterial activity was observed in CaSr10mMAg-Ti and CaSr50mMAg-Ti groups. In vivo experiments were performed using the rat subcutaneous pocket infection model and evaluated by counting the attached bacteria, wound appearance, and histological evaluation. High antibacterial activity value (AAV >2) and anti-inflammatory effects were observed in the CaSr50mMAg-Ti group. However, CaSr10mMAg-Ti did not exhibit consistent antibacterial activity. For in vivo biocompatibility and bone-bonding ability evaluation, rods were implanted into the rat femur. No cytotoxicity was observed at 1 week, and good bone-bonding ability at 4 and 8 weeks was not significantly different from that of CaSr1mMAg-Ti. To evaluate in vivo bioactivity and cytotoxicity, MC3T3-E1 cells were cultured on disks. CaSr10mMAg-Ti and CaSr50mMAg-Ti significantly inhibited the proliferation and differentiation of MC3T3E1 cells, as well as the production of extracellular matrix in vivo, despite showing good biocompatibility in vivo. In conclusion, CaSr50mMAg-Ti, with increased Ag ion loading, exhibited high antibacterial activity in vivo while maintaining the bone-bonding ability and is a promising therapeutic biomaterial. Further research is needed to determine the optimal combination of therapeutic concentrations of Sr and Ag.
Collapse
Affiliation(s)
- Kazuki Orita
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shunsuke Fujibayashi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yaichiro Okuzu
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Seiji Yamaguchi
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai, Japan
| | - Koji Goto
- Department of Orthopaedic Surgery, Kindai University Hospital, Osaka-Sayama, Japan
| | - Bungo Otsuki
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshiyuki Kawai
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takayoshi Shimizu
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Makoto Hayashi
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Norimasa Ikeda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yusuke Takaoka
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shintaro Honda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tatsuhito Ikezaki
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
3
|
Li H, Wang W, Wang J. Mechanical Signal Transduction: A Key Role of Fluid Shear Forces in the Development of Osteoarthritis. J Inflamm Res 2024; 17:10199-10207. [PMID: 39649420 PMCID: PMC11624683 DOI: 10.2147/jir.s498914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/10/2024] [Indexed: 12/10/2024] Open
Abstract
Globally, osteoarthritis is a common and highly disabling disease that places a heavy burden on society and medical systems. The role of biomechanical factors in the development of osteoarthritis has gradually received more attention. As a key biomechanical stimulus, fluid shear force is becoming the focus of research for its dual role in maintaining cartilage health and disease progression. This paper conducts an in-depth discussion on the mechanism of fluid shear force in osteoarthritis and its impact on the disease process, aiming to reveal how fluid shear stress affects the development of osteoarthritis by regulating the physiological function and signal transduction pathways of chondrocytes.
Collapse
Affiliation(s)
- Haitao Li
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, 55000, People’s Republic of China
| | - Wei Wang
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, 55000, People’s Republic of China
| | - Jian Wang
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, 55000, People’s Republic of China
| |
Collapse
|
4
|
Zhu K, Cheng CH, Ma HL, Liu GX, Fan SG, Deng YQ, Jiang JJ, Feng J, Guo ZX. Identification and functional characterization of laminin receptor in the mud crab, Scylla paramamosain, in response to MCDV-1 challenge. FISH & SHELLFISH IMMUNOLOGY 2024; 150:109648. [PMID: 38777253 DOI: 10.1016/j.fsi.2024.109648] [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: 01/20/2024] [Revised: 05/05/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Laminin receptor (LR), which mediating cell adhesion to the extracellular matrix, plays a crucial role in cell signaling and regulatory functions. In the present study, a laminin receptor gene (SpLR) was cloned and characterized from the mud crab (Scylla paramamosain). The full length of SpLR contained an open reading frame (ORF) of 960 bp encoding 319 amino acids, a 5' untranslated region (UTR) of 66 bp and a 3' UTR of 49 bp. The predicted protein comprised two Ribosomal-S2 domains and a 40S-SA-C domain. The mRNA of SpLR was highly expressed in the gill, followed by the hepatopancreas. The expression of SpLR was up-regulated after mud crab dicistrovirus-1(MCDV-1) infection. Knocking down SpLR in vivo by RNA interference significantly down-regulated the expression of the immune genes SpJAK, SpSTAT, SpToll1, SpALF1 and SpALF5. This study shown that the expression level of SpToll1 and SpCAM in SpLR-interfered group significantly increased after MCDV-1 infection. Moreover, silencing of SpLR in vivo decreased the MCDV-1 replication and increased the survival rate of mud crabs after MCDV-1 infection. These findings collectively suggest a pivotal role for SpLR in the mud crab's response to MCDV-1 infection. By influencing the expression of critical innate immune factors and impacting viral replication dynamics, SpLR emerges as a key player in the intricate host-pathogen interaction, providing valuable insights into the molecular mechanisms underlying MCDV-1 pathogenesis in mud crabs.
Collapse
Affiliation(s)
- Ke Zhu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510300, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Chang-Hong Cheng
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510300, China
| | - Hong-Ling Ma
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510300, China
| | - Guang-Xin Liu
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510300, China
| | - Si-Gang Fan
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510300, China
| | - Yi-Qin Deng
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510300, China
| | - Jian-Jun Jiang
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510300, China
| | - Juan Feng
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510300, China
| | - Zhi-Xun Guo
- Key Laboratory of South China Sea Fishery Resources Exploitation & Utilization, Ministry of Agriculture, South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou, Guangdong, 510300, China.
| |
Collapse
|
5
|
Owen R, Wittkowske C, Lacroix D, Perrault CM, Reilly GC. β-glycerophosphate, not low magnitude fluid shear stress, increases osteocytogenesis in the osteoblast-to-osteocyte cell line IDG-SW3. Connect Tissue Res 2024; 65:313-329. [PMID: 38982804 PMCID: PMC11371265 DOI: 10.1080/03008207.2024.2375065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024]
Abstract
AIM As osteoblasts deposit a mineralized collagen network, a subpopulation of these cells differentiates into osteocytes. Biochemical and mechanical stimuli, particularly fluid shear stress (FSS), are thought to regulate this, but their relative influence remains unclear. Here, we assess both biochemical and mechanical stimuli on long-term bone formation and osteocytogenesis using the osteoblast-osteocyte cell line IDG-SW3. METHODS Due to the relative novelty and uncommon culture conditions of IDG-SW3 versus other osteoblast-lineage cell lines, effects of temperature and media formulation on matrix deposition and osteocytogenesis were initially characterized. Subsequently, the relative influence of biochemical (β-glycerophosphate (βGP) and ascorbic acid 2-phosphate (AA2P)) and mechanical stimulation on osteocytogenesis was compared, with intermittent application of low magnitude FSS generated by see-saw rocker. RESULTS βGP and AA2P supplementation were required for mineralization and osteocytogenesis, with 33°C cultures retaining a more osteoblastic phenotype and 37°C cultures undergoing significantly higher osteocytogenesis. βGP concentration positively correlated with calcium deposition, whilst AA2P stimulated alkaline phosphatase (ALP) activity and collagen deposition. We demonstrate that increasing βGP concentration also significantly enhances osteocytogenesis as quantified by the expression of green fluorescent protein linked to Dmp1. Intermittent FSS (~0.06 Pa) rocker had no effect on osteocytogenesis and matrix deposition. CONCLUSIONS This work demonstrates the suitability and ease with which IDG-SW3 can be utilized in osteocytogenesis studies. IDG-SW3 mineralization was only mediated through biochemical stimuli with no detectable effect of low magnitude FSS. Osteocytogenesis of IDG-SW3 primarily occurred in mineralized areas, further demonstrating the role mineralization of the bone extracellular matrix has in osteocyte differentiation.
Collapse
Affiliation(s)
- Robert Owen
- Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- School of Pharmacy, University of Nottingham Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Claudia Wittkowske
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Damien Lacroix
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Cecile M. Perrault
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
- Department of Mechanical Engineering, University of Sheffield, Sheffield, UK
| | - Gwendolen C. Reilly
- Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
- INSIGNEO Institute for In Silico Medicine, University of Sheffield, Sheffield, UK
| |
Collapse
|
6
|
Cao R, Tian H, Tian Y, Fu X. A Hierarchical Mechanotransduction System: From Macro to Micro. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302327. [PMID: 38145330 PMCID: PMC10953595 DOI: 10.1002/advs.202302327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/27/2023] [Indexed: 12/26/2023]
Abstract
Mechanotransduction is a strictly regulated process whereby mechanical stimuli, including mechanical forces and properties, are sensed and translated into biochemical signals. Increasing data demonstrate that mechanotransduction is crucial for regulating macroscopic and microscopic dynamics and functionalities. However, the actions and mechanisms of mechanotransduction across multiple hierarchies, from molecules, subcellular structures, cells, tissues/organs, to the whole-body level, have not been yet comprehensively documented. Herein, the biological roles and operational mechanisms of mechanotransduction from macro to micro are revisited, with a focus on the orchestrations across diverse hierarchies. The implications, applications, and challenges of mechanotransduction in human diseases are also summarized and discussed. Together, this knowledge from a hierarchical perspective has the potential to refresh insights into mechanotransduction regulation and disease pathogenesis and therapy, and ultimately revolutionize the prevention, diagnosis, and treatment of human diseases.
Collapse
Affiliation(s)
- Rong Cao
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Huimin Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Yan Tian
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Xianghui Fu
- Department of Endocrinology and MetabolismCenter for Diabetes Metabolism ResearchState Key Laboratory of Biotherapy and Cancer CenterWest China Medical SchoolWest China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| |
Collapse
|
7
|
Claude-Taupin A, Dupont N. To squeeze or not: Regulation of cell size by mechanical forces in development and human diseases. Biol Cell 2024; 116:e2200101. [PMID: 38059665 DOI: 10.1111/boc.202200101] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023]
Abstract
Physical constraints, such as compression, shear stress, stretching and tension play major roles during development and tissue homeostasis. Mechanics directly impact physiology, and their alteration is also recognized as having an active role in driving human diseases. Recently, growing evidence has accumulated on how mechanical forces are translated into a wide panel of biological responses, including metabolism and changes in cell morphology. The aim of this review is to summarize and discuss our knowledge on the impact of mechanical forces on cell size regulation. Other biological consequences of mechanical forces will not be covered by this review. Moreover, wherever possible, we also discuss mechanosensors and molecular and cellular signaling pathways upstream of cell size regulation. We finally highlight the relevance of mechanical forces acting on cell size in physiology and human diseases.
Collapse
Affiliation(s)
- Aurore Claude-Taupin
- Institut Necker Enfants Malades (INEM), INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Paris, France
| | - Nicolas Dupont
- Institut Necker Enfants Malades (INEM), INSERM UMR-S1151, CNRS UMR-S8253, Université Paris Cité, Paris, France
| |
Collapse
|
8
|
Tang T, Landis W, Blouin S, Bertinetti L, Hartmann MA, Berzlanovich A, Weinkamer R, Wagermaier W, Fratzl P. Subcanalicular Nanochannel Volume Is Inversely Correlated With Calcium Content in Human Cortical Bone. J Bone Miner Res 2023; 38:313-325. [PMID: 36433915 DOI: 10.1002/jbmr.4753] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 11/10/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
The spatial distribution of mineralization density is an important signature of bone growth and remodeling processes, and its alterations are often related to disease. The extracellular matrix of some vertebrate mineralized tissues is known to be perfused by a lacunocanalicular network (LCN), a fluid-filled unmineralized structure that harbors osteocytes and their fine processes and transports extracellular fluid and its constituents. The current report provides evidence for structural and compositional heterogeneity at an even smaller, subcanalicular scale. The work reveals an extensive unmineralized three-dimensional (3D) network of nanochannels (~30 nm in diameter) penetrating the mineralized extracellular matrix of human femoral cortical bone and encompassing a greater volume fraction and surface area than these same parameters of the canaliculi comprising the LCN. The present study combines high-resolution focused ion beam-scanning electron microscopy (FIB-SEM) to investigate bone ultrastructure in 3D with quantitative backscattered electron imaging (qBEI) to estimate local bone mineral content. The presence of nanochannels has been found to impact qBEI measurements fundamentally, such that volume percentage (vol%) of nanochannels correlates inversely with weight percentage (wt%) of calcium. This mathematical relationship between nanochannel vol% and calcium wt% suggests that the nanochannels could potentially provide space for ion and small molecule transport throughout the bone matrix. Collectively, these data propose a reinterpretation of qBEI measurements, accounting for nanochannel presence in human bone tissue in addition to collagen and mineral. Further, the results yield insight into bone mineralization processes at the nanometer scale and present the possibility for a potential role of the nanochannel system in permitting ion and small molecule diffusion throughout the extracellular matrix. Such a possible function could thereby lead to the sequestration or occlusion of the ions and small molecules within the extracellular matrix. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Tengteng Tang
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - William Landis
- Department of Preventive and Restorative Dental Sciences, University of California at San Francisco, San Francisco, CA, USA
| | - Stéphane Blouin
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Med. Department Hanusch Hospital, Vienna, Austria
| | - Luca Bertinetti
- Center for Molecular Bioengineering, TU Dresden, Dresden, Germany
| | - Markus A Hartmann
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Med. Department Hanusch Hospital, Vienna, Austria
| | | | - Richard Weinkamer
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Wolfgang Wagermaier
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| | - Peter Fratzl
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, Potsdam, Germany
| |
Collapse
|
9
|
Ma Q, Miri Z, Haugen HJ, Moghanian A, Loca D. Significance of mechanical loading in bone fracture healing, bone regeneration, and vascularization. J Tissue Eng 2023; 14:20417314231172573. [PMID: 37251734 PMCID: PMC10214107 DOI: 10.1177/20417314231172573] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/13/2023] [Indexed: 05/31/2023] Open
Abstract
In 1892, J.L. Wolff proposed that bone could respond to mechanical and biophysical stimuli as a dynamic organ. This theory presents a unique opportunity for investigations on bone and its potential to aid in tissue repair. Routine activities such as exercise or machinery application can exert mechanical loads on bone. Previous research has demonstrated that mechanical loading can affect the differentiation and development of mesenchymal tissue. However, the extent to which mechanical stimulation can help repair or generate bone tissue and the related mechanisms remain unclear. Four key cell types in bone tissue, including osteoblasts, osteoclasts, bone lining cells, and osteocytes, play critical roles in responding to mechanical stimuli, while other cell lineages such as myocytes, platelets, fibroblasts, endothelial cells, and chondrocytes also exhibit mechanosensitivity. Mechanical loading can regulate the biological functions of bone tissue through the mechanosensor of bone cells intraosseously, making it a potential target for fracture healing and bone regeneration. This review aims to clarify these issues and explain bone remodeling, structure dynamics, and mechano-transduction processes in response to mechanical loading. Loading of different magnitudes, frequencies, and types, such as dynamic versus static loads, are analyzed to determine the effects of mechanical stimulation on bone tissue structure and cellular function. Finally, the importance of vascularization in nutrient supply for bone healing and regeneration was further discussed.
Collapse
Affiliation(s)
- Qianli Ma
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
- Department of Immunology, School of
Basic Medicine, Fourth Military Medical University, Xi’an, PR China
| | - Zahra Miri
- Department of Materials Engineering,
Isfahan University of Technology, Isfahan, Iran
| | - Håvard Jostein Haugen
- Department of Biomaterials, Institute
of Clinical Dentistry, University of Oslo, Norway
| | - Amirhossein Moghanian
- Department of Materials Engineering,
Imam Khomeini International University, Qazvin, Iran
| | - Dagnjia Loca
- Rudolfs Cimdins Riga Biomaterials
Innovations and Development Centre, Institute of General Chemical Engineering,
Faculty of Materials Science and Applied Chemistry, Riga Technical University, Riga,
Latvia
- Baltic Biomaterials Centre of
Excellence, Headquarters at Riga Technical University, Riga, Latvia
| |
Collapse
|
10
|
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: 0.5] [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
|
11
|
Hu Q, Bian Q, Rong D, Wang L, Song J, Huang HS, Zeng J, Mei J, Wang PY. JAK/STAT pathway: Extracellular signals, diseases, immunity, and therapeutic regimens. Front Bioeng Biotechnol 2023; 11:1110765. [PMID: 36911202 PMCID: PMC9995824 DOI: 10.3389/fbioe.2023.1110765] [Citation(s) in RCA: 117] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Janus kinase/signal transduction and transcription activation (JAK/STAT) pathways were originally thought to be intracellular signaling pathways that mediate cytokine signals in mammals. Existing studies show that the JAK/STAT pathway regulates the downstream signaling of numerous membrane proteins such as such as G-protein-associated receptors, integrins and so on. Mounting evidence shows that the JAK/STAT pathways play an important role in human disease pathology and pharmacological mechanism. The JAK/STAT pathways are related to aspects of all aspects of the immune system function, such as fighting infection, maintaining immune tolerance, strengthening barrier function, and cancer prevention, which are all important factors involved in immune response. In addition, the JAK/STAT pathways play an important role in extracellular mechanistic signaling and might be an important mediator of mechanistic signals that influence disease progression, immune environment. Therefore, it is important to understand the mechanism of the JAK/STAT pathways, which provides ideas for us to design more drugs targeting diseases based on the JAK/STAT pathway. In this review, we discuss the role of the JAK/STAT pathway in mechanistic signaling, disease progression, immune environment, and therapeutic targets.
Collapse
Affiliation(s)
- Qian Hu
- Department of Pharmacy, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Qihui Bian
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| | - Dingchao Rong
- Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Leiyun Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China.,Department of Pharmacy, Wuhan First Hospital, Wuhan, China
| | - Jianan Song
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| | - Hsuan-Shun Huang
- Department of Research, Center for Prevention and Therapy of Gynecological Cancers, Buddhist Tzu Chi General Hospital, Hualien, Taiwan
| | - Jun Zeng
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Mei
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China.,Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Peng-Yuan Wang
- Oujiang Laboratory, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Institute of Aging, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
12
|
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: 8] [Impact Index Per Article: 2.7] [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
|
13
|
Niu D, Zhang Y, Chen J, Li D, He C, Liu H. Mechanobiology Platform Realized Using Photomechanical Mxene Nanocomposites: Bilayer Photoactuator Design and In Vitro Mechanical Forces Stimulation. MATERIALS (BASEL, SWITZERLAND) 2022; 15:6869. [PMID: 36234210 PMCID: PMC9570783 DOI: 10.3390/ma15196869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/25/2022] [Accepted: 09/06/2022] [Indexed: 06/16/2023]
Abstract
Mechanotransduction is the process by which cells convert external forces and physical constraints into biochemical signals that control several aspects of cellular behavior. A number of approaches have been proposed to investigate the mechanisms of mechanotransduction; however, it remains a great challenge to develop a platform for dynamic multivariate mechanical stimulation of single cells and small colonies of cells. In this study, we combined polydimethylsiloxane (PDMS) and PDMS/Mxene nanoplatelets (MNPs) to construct a soft bilayer nanocomposite for extracellular mechanical stimulation. Fast backlash actuation of the bilayer as a result of near-infrared irradiation caused mechanical force stimulation of cells in a controllable manner. The excellent controllability of the light intensity and frequency allowed backlash bending acceleration and frequency to be manipulated. As gastric gland carcinoma cell line MKN-45 was the research subject, mechanical force loading conditions could trigger apoptosis of the cells in a stimulation duration time-dependent manner. Cell apoptotic rates were positively related to the duration time. In the case of 6 min mechanical force loading, apoptotic cell percentage rose to 34.46% from 5.5% of the control. This approach helps apply extracellular mechanical forces, even with predesigned loading cycles, and provides a solution to study cell mechanotransduction in complex force conditions. It is also a promising therapeutic technique for combining physical therapy and biomechanics.
Collapse
Affiliation(s)
- Dong Niu
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Yanli Zhang
- Key Laboratory for Molecular Genetic Mechanisms and Intervention Research on High Altitude Disease of Tibet Autonomous Region, Medical College, Xizang Minzu University, Xianyang 712082, China
| | - Jinlan Chen
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Dachao Li
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Chunmeng He
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China
| | - Hongzhong Liu
- State Key Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an 710049, China
- The Joint Key Laboratory of Graphene, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
14
|
Mainardi VL, Rubert M, Sabato C, de Leeuw A, Arrigoni C, Dubini G, Candrian C, Müller R, Moretti M. Culture of 3D Bioprinted Bone Constructs Requires an Increased Fluid Dynamic Stimulation. Acta Biomater 2022; 153:374-385. [PMID: 36108964 DOI: 10.1016/j.actbio.2022.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 08/05/2022] [Accepted: 09/07/2022] [Indexed: 11/01/2022]
Abstract
In vitro flow-induced mechanical stimulation of developing bone tissue constructs has been shown to favor mineral deposition in scaffolds seeded with cells directly exposed to the fluid flow. However, the effect of fluid dynamic parameters, such as shear stress (SS), within 3D bioprinted constructs is still unclear. Thus, this study aimed at correlating the SS levels and the mineral deposition in 3D bioprinted constructs, evaluating the possible dampening effect of the hydrogel. Human mesenchymal stem cells (hMSCs) were embedded in 3D bioprinted porous structures made of alginate and gelatin. 3D bioprinted constructs were cultured in an osteogenic medium assessing the influence of different flow rates (0, 0.7 and 7 ml/min) on calcium and collagen deposition through histology, and bone volume (BV) through micro-computed tomography. Uniform distribution of calcium and collagen was observed in all groups. Nevertheless, BV significantly increased in perfused groups as compared to static control, ranging from 0.35±0.28 mm3, 11.90±8.74 mm3 and 25.81±5.02 mm3 at week 3 to 2.28±0.78 mm3, 22.55±2.45 mm3 and 46.05±5.95 mm3 at week 6 in static, 0.7 and 7 ml/min groups, respectively. SS values on construct fibers in the range 10-100 mPa in 7 ml/min samples were twice as high as those in 0.7 ml/min samples showing the same trend of BV. The obtained results suggest that it is necessary to enhance the flow-induced mechanical stimulation of cell-embedding hydrogels to increase the amount of mineral deposited by hMSCs, compared to what is generally reported for the development of in vitro bone constructs. STATEMENT OF SIGNIFICANCE: : Culture of 3D Bioprinted Bone Constructs Requires an Increased Fluid Dynamic Stimulation, In this study, we evaluated for the first time how the hydrogel structure dampens the effect of flow-induced mechanical stimulation during the culture of 3D bioprinted bone tissue constructs. By combining computational and experimental techniques we demonstrated that those shear stress thresholds generally considered for culturing cells seeded on scaffold surface, are no longer applicable when cells are embedded in 3D bioprinted constructs. Significantly, more bone volume was formed in constructs exposed to shear stress values generally considered as detrimental than in constructs exposed shear stress values generally considered as beneficial after 3 weeks and 6 weeks of dynamic culture using a perfusion bioreactor.
Collapse
Affiliation(s)
- V L Mainardi
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Bellinzona 6500, Switzerland; Laboratory of Biological Structures Mechanics (LaBS), Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan 20133, Italy; Laboratory for Bone Biomechanics, Institute for Biomechanics, ETH Zurich, Zurich 8093, Switzerland
| | - M Rubert
- Laboratory for Bone Biomechanics, Institute for Biomechanics, ETH Zurich, Zurich 8093, Switzerland
| | - C Sabato
- Laboratory of Biological Structures Mechanics (LaBS), Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan 20133, Italy; Laboratory for Bone Biomechanics, Institute for Biomechanics, ETH Zurich, Zurich 8093, Switzerland
| | - A de Leeuw
- Laboratory for Bone Biomechanics, Institute for Biomechanics, ETH Zurich, Zurich 8093, Switzerland
| | - C Arrigoni
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Bellinzona 6500, Switzerland
| | - G Dubini
- Laboratory of Biological Structures Mechanics (LaBS), Department of Chemistry, Materials and Chemical Engineering "Giulio Natta", Politecnico di Milano, Milan 20133, Italy
| | - C Candrian
- Servizio di Traumatologia e Ortopedia, Ente Ospedaliero Cantonale (EOC), Lugano 6900, Switzerland; Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6900, Switzerland
| | - R Müller
- Laboratory for Bone Biomechanics, Institute for Biomechanics, ETH Zurich, Zurich 8093, Switzerland.
| | - M Moretti
- Regenerative Medicine Technologies Lab, Ente Ospedaliero Cantonale (EOC), Bellinzona 6500, Switzerland; Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 6900, Switzerland; Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milan 20161, Italy.
| |
Collapse
|
15
|
Zhang K, Liu X, Tang Y, Liu Z, Yi Q, Wang L, Geng B, Xia Y. Fluid Shear Stress Promotes Osteoblast Proliferation and Suppresses Mitochondrial-Mediated Osteoblast Apoptosis Through the miR-214-3p-ATF4 Signaling Axis. Physiol Res 2022. [DOI: 10.33549/physiolres.934917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
MicroRNAs (miRNAs) play vital roles in bone metabolism and participate in the mechanically induced bone alterations. The underlying molecular mechanisms by which fluid shear stress (FSS) regulate the proliferative and apoptotic phenotypic changes of osteoblasts remain elusive. The study aimed to investigate the regulatory effects of FSS on osteoblast proliferative and apoptotic phenotypes and the roles of miR-214-3p-ATF4 (activating transcription factor 4) signaling axis in the mechanomodulation processes. FSS promoted the proliferative activity of osteoblasts and suppressed mitochondrial-mediated osteoblast apoptosis. FSS decreased miR-214-3p expression and increased ATF4 expression in MC3T3-E1 osteoblasts. MiR-214-3p inhibited osteoblast proliferative activity and promoted mitochondrial-mediated osteoblast apoptosis. Overexpression of miR-214-3p attenuated FSS-enhanced osteoblast proliferation and FSS-suppressed mitochondrial-mediated osteoblast apoptosis. We validated that ATF4 acted as a target gene of miR-214-3p. Moreover, miR-214 3p regulated osteoblast proliferation and apoptosis through targeting ATF4. Taken together, our study proved that FSS could suppress mitochondrial-mediated osteoblast apoptosis and promote osteoblast proliferation through the miR-214-3p-ATF4 signaling axis.
Collapse
Affiliation(s)
- K Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, Lanzhou Gansu, China, Orthopaedics Key Laboratory of Gansu Province, Lanzhou Gansu, China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Spasic M, Duffy MP, Jacobs CR. Fenoldopam Sensitizes Primary Cilia-Mediated Mechanosensing to Promote Osteogenic Intercellular Signaling and Whole Bone Adaptation. J Bone Miner Res 2022; 37:972-982. [PMID: 35230705 PMCID: PMC9098671 DOI: 10.1002/jbmr.4536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 02/01/2022] [Accepted: 02/12/2022] [Indexed: 11/05/2022]
Abstract
Bone cells actively respond to mechanical stimuli to direct bone formation, yet there is no current treatment strategy for conditions of low bone mass and osteoporosis designed to target the inherent mechanosensitivity of bone. Our group has previously identified the primary cilium as a critical mechanosensor within bone, and that pharmacologically targeting the primary cilium with fenoldopam can enhance osteocyte mechanosensitivity. Here, we demonstrate that potentiating osteocyte mechanosensing with fenoldopam in vitro promotes pro-osteogenic paracrine signaling to osteoblasts. Conversely, impairing primary cilia formation and the function of key ciliary mechanotransduction proteins attenuates this intercellular signaling cascade. We then utilize an in vivo model of load-induced bone formation to demonstrate that fenoldopam treatment sensitizes bones of both healthy and osteoporotic mice to mechanical stimulation. Furthermore, we show minimal adverse effects of this treatment and demonstrate that prolonged treatment biases trabecular bone adaptation. This work is the first to examine the efficacy of targeting primary cilia-mediated mechanosensing to enhance bone formation in osteoporotic animals. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Milos Spasic
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Michael P Duffy
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | | |
Collapse
|
17
|
Lipreri MV, Baldini N, Graziani G, Avnet S. Perfused Platforms to Mimic Bone Microenvironment at the Macro/Milli/Microscale: Pros and Cons. Front Cell Dev Biol 2022; 9:760667. [PMID: 35047495 PMCID: PMC8762164 DOI: 10.3389/fcell.2021.760667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/30/2021] [Indexed: 11/26/2022] Open
Abstract
As life expectancy increases, the population experiences progressive ageing. Ageing, in turn, is connected to an increase in bone-related diseases (i.e., osteoporosis and increased risk of fractures). Hence, the search for new approaches to study the occurrence of bone-related diseases and to develop new drugs for their prevention and treatment becomes more pressing. However, to date, a reliable in vitro model that can fully recapitulate the characteristics of bone tissue, either in physiological or altered conditions, is not available. Indeed, current methods for modelling normal and pathological bone are poor predictors of treatment outcomes in humans, as they fail to mimic the in vivo cellular microenvironment and tissue complexity. Bone, in fact, is a dynamic network including differently specialized cells and the extracellular matrix, constantly subjected to external and internal stimuli. To this regard, perfused vascularized models are a novel field of investigation that can offer a new technological approach to overcome the limitations of traditional cell culture methods. It allows the combination of perfusion, mechanical and biochemical stimuli, biological cues, biomaterials (mimicking the extracellular matrix of bone), and multiple cell types. This review will discuss macro, milli, and microscale perfused devices designed to model bone structure and microenvironment, focusing on the role of perfusion and encompassing different degrees of complexity. These devices are a very first, though promising, step for the development of 3D in vitro platforms for preclinical screening of novel anabolic or anti-catabolic therapeutic approaches to improve bone health.
Collapse
Affiliation(s)
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Biomedical Science and Technologies Lab, IRCSS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Gabriela Graziani
- Laboratory for NanoBiotechnology (NaBi), IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
18
|
Sforna L, Michelucci A, Morena F, Argentati C, Franciolini F, Vassalli M, Martino S, Catacuzzeno L. Piezo1 controls cell volume and migration by modulating swelling-activated chloride current through Ca 2+ influx. J Cell Physiol 2021; 237:1857-1870. [PMID: 34913176 DOI: 10.1002/jcp.30656] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/21/2022]
Abstract
Regulatory volume decrease (RVD), a homeostatic process responsible for the re-establishment of the original cell volume upon swelling, is critical in controlling several functions, including migration. RVD is mainly sustained by the swelling-activated Cl- current (ICl,swell ), which can be modulated by cytoplasmic Ca2+ . Cell swelling also activates mechanosensitive channels, including the ubiquitously expressed Ca2+ -permeable channel Piezo1. We hypothesized that, by controlling cytoplasmic Ca2+ and in turn ICl,swell , Piezo1 is involved in the fine regulation of RVD and cell migration. We compared RVD and ICl,swell in wild-type (WT) HEK293T cells, which express endogenous levels of Piezo1, and in cells overexpressing (OVER) or knockout (KO) for Piezo1. Compared to WT, RVD was markedly increased in OVER, while virtually absent in KO cells. Consistently, ICl,swell amplitude was highest in OVER and lowest in KO cells, with WT cells displaying an intermediate level, suggesting a Ca2+ -dependent modulation of the current by Piezo1 channels. Indeed, in the absence of external Ca2+ , ICl,swell in both WT and OVER cells, as well as the RVD probed in OVER cells, were significantly lower than in the presence of Ca2+ and no longer different compared to KO cells. However, the Piezo-mediated Ca2+ influx was ineffective in enhancing ICl,swell in the absence of releasable Ca2+ from intracellular stores. The different expression levels of Piezo1 affected also cell migration which was strongly enhanced in OVER, while reduced in KO cells, as compared to WT. Taken together, our data indicate that Piezo1 controls RVD and migration in HEK293T cells by modulating ICl,swell through Ca2+ influx.
Collapse
Affiliation(s)
- Luigi Sforna
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Antonio Michelucci
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio of Chieti, Chieti, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Chiara Argentati
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Fabio Franciolini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Massimo Vassalli
- James Watt School of Engineering, University of Glasgow, Center for the Cellular Microenvironment, School of Engineering, G12 8LT, Glasgow, UK
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy.,CEMIN, Center of Excellence on Nanostructured Innovative Materials, University of Perugia, Perugia, Italy
| | - Luigi Catacuzzeno
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| |
Collapse
|
19
|
The mechanosensory and mechanotransductive processes mediated by ion channels and the impact on bone metabolism: A systematic review. Arch Biochem Biophys 2021; 711:109020. [PMID: 34461086 DOI: 10.1016/j.abb.2021.109020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023]
Abstract
Mechanical environments were associated with alterations in bone metabolism. Ion channels present on bone cells are indispensable for bone metabolism and can be directly or indirectly activated by mechanical stimulation. This review aimed to discuss the literature reporting the mechanical regulatory effects of ion channels on bone cells and bone tissue. An electronic search was conducted in PubMed, Embase and Web of Science. Studies about mechanically induced alteration of bone cells and bone tissue by ion channels were included. Ion channels including TRP family channels, Ca2+ release-activated Ca2+ channels (CRACs), Piezo1/2 channels, purinergic receptors, NMDA receptors, voltage-sensitive calcium channels (VSCCs), TREK2 potassium channels, calcium- and voltage-dependent big conductance potassium (BKCa) channels, small conductance, calcium-activated potassium (SKCa) channels and epithelial sodium channels (ENaCs) present on bone cells and bone tissue participate in the mechanical regulation of bone development in addition to contributing to direct or indirect mechanotransduction such as altered membrane potential and ionic flux. Physiological (beneficial) mechanical stimulation could induce the anabolism of bone cells and bone tissue through ion channels, but abnormal (harmful) mechanical stimulation could also induce the catabolism of bone cells and bone tissue through ion channels. Functional expression of ion channels is vital for the mechanotransduction of bone cells. Mechanical activation (opening) of ion channels triggers ion influx and induces the activation of intracellular modulators that can influence bone metabolism. Therefore, mechanosensitive ion channels provide new insights into therapeutic targets for the treatment of bone-related diseases such as osteopenia and aseptic implant loosening.
Collapse
|
20
|
Nokhbatolfoghahaei H, Rad MR, Paknejad Z, Ardeshirylajimi A, Khojasteh A. Identification osteogenic signaling pathways following mechanical stimulation: A systematic review. Curr Stem Cell Res Ther 2021; 17:772-792. [PMID: 34615453 DOI: 10.2174/1574888x16666211006105915] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/17/2021] [Accepted: 07/26/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION It has been shown that mechanical forces can induce or promote osteogenic differentiation as well as remodeling of the new created bone tissues. To apply this characteristic in bone tissue engineering, it is important to know which mechanical stimuli through which signaling pathway has a more significant impact on osteogenesis. METHODS In this systematic study, an electronic search was conducted using PubMed and Google Scholar databases. This study has been prepared and organized according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. Included studies were first categorized according to the in vivo and in vitro studies. RESULTS Six types of mechanical stresses were used in these articles and the most commonly used mechanical force and cell source were tension and bone marrow-derived mesenchymal stem cells (BMMSCs), respectively. These forces were able to trigger twelve signaling pathways in which Wnt pathway was so prominent. CONCLUSION 1) Although specific signaling pathways are induced through specific mechanical forces, Wnt signaling pathways are predominantly activated by almost all types of force/stimulation, 2) All signaling pathways regulate expression of RUNX2, which is known as a master regulator of osteogenesis, 3) In Tension force, the mode of force administration, i.e, continuous or non-continuous tension is more important than the percentage of elongation.
Collapse
Affiliation(s)
- Hanieh Nokhbatolfoghahaei
- Dental Research Center, Research Institute of Dental Sciences, 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
| | - Zahrasadat Paknejad
- Medical nanotechnology and tissue engineering research Center, Shahid Beheshti University of Medical Sciences, Tehran. Iran
| | - Abdolreza Ardeshirylajimi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran. Iran
| | - Arash Khojasteh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran. Iran
| |
Collapse
|
21
|
Crous A, Abrahamse H. The Signalling Effects of Photobiomodulation on Osteoblast Proliferation, Maturation and Differentiation: A Review. Stem Cell Rev Rep 2021; 17:1570-1589. [PMID: 33686595 DOI: 10.1007/s12015-021-10142-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2021] [Indexed: 02/06/2023]
Abstract
Proliferation of osteoblasts is essential for maturation and mineralization of bone matrix. Ossification, the natural phase of bone-forming and hardening is a carefully regulated phase where deregulation of this process may result in insufficient or excessive bone mineralization or ectopic calcification. Osteoblasts can also be differentiated into osteocytes, populating short interconnecting passages within the bone matrix. Over the past few decades, we have seen a significant improvement in awareness and techniques using photobiomodulation (PBM) to stimulate cell function. One of the applications of PBM is the promotion of osteoblast proliferation and maturation. PBM research results on osteoblasts showed increased mitochondrial ATP production, increased osteoblast activity and proliferation, increased and pro-osteoblast expression in the presence of red and NIR radiation. Osteocyte differentiation was also accomplished using blue and green light, showing that different light parameters have various signalling effects. The current review addresses osteoblast function and control, a new understanding of PBM on osteoblasts and its therapeutic impact using various parameters to optimize osteoblast function that may be clinically important. Graphical Abstract.
Collapse
Affiliation(s)
- Anine Crous
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, PO Box 17011, Johannesburg, 2028, South Africa.
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, PO Box 17011, Johannesburg, 2028, South Africa
| |
Collapse
|
22
|
Dole NS, Yoon J, Monteiro DA, Yang J, Mazur CM, Kaya S, Belair CD, Alliston T. Mechanosensitive miR-100 coordinates TGFβ and Wnt signaling in osteocytes during fluid shear stress. FASEB J 2021; 35:e21883. [PMID: 34569659 PMCID: PMC9153140 DOI: 10.1096/fj.202100930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 08/12/2021] [Indexed: 11/12/2022]
Abstract
Organism scale mechanical forces elicit cellular scale changes through coordinated regulation of multiple signaling pathways. The mechanisms by which cells integrate signaling to generate a unified biological response remains a major question in mechanobiology. For example, the mechanosensitive response of bone and other tissues requires coordinated signaling by the transforming growth factor beta (TGFβ) and Wnt pathways through mechanisms that are not well‐defined. Here we report a new microRNA‐dependent mechanism that mediates mechanosensitive crosstalk between TGFβ and Wnt signaling in osteocytes exposed to fluid shear stress (FSS). From 60 mechanosensitive microRNA (miRs) identified by small‐RNAseq, miR100 expression is suppressed by in vivo hindlimb loading in the murine tibia and by cellular scale FSS in OCY454 cells. Though FSS activates both TGFβ and Wnt signaling in osteocytes, only TGFβ represses miR‐100 expression. miR‐100, in turn, antagonizes Wnt signaling by targeting and inhibiting expression of Frizzled receptors (FZD5/FZD8). Accordingly, miR‐100 inhibition blunts FSS‐ and TGFβ‐inducible Wnt signaling. Therefore, our results identify FSS‐responsive miRNAs in osteocytes, including one that integrates the mechanosensitive function of two essential signaling pathways in the osteoanabolic response of bone to mechanical load.
Collapse
Affiliation(s)
- Neha S Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Jihee Yoon
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - David A Monteiro
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Jason Yang
- Department of Molecular & Cell Biology, University of California Berkeley, Berkeley, California, USA
| | - Courtney M Mazur
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Cassandra D Belair
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA.,Department of Urology, University of California, San Francisco, San Francisco, California, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
23
|
Claude-Taupin A, Codogno P, Dupont N. Links between autophagy and tissue mechanics. J Cell Sci 2021; 134:271984. [PMID: 34472605 DOI: 10.1242/jcs.258589] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Physical constraints, such as compression, shear stress, stretching and tension, play major roles during development, tissue homeostasis, immune responses and pathologies. Cells and organelles also face mechanical forces during migration and extravasation, and investigations into how mechanical forces are translated into a wide panel of biological responses, including changes in cell morphology, membrane transport, metabolism, energy production and gene expression, is a flourishing field. Recent studies demonstrate the role of macroautophagy in the integration of physical constraints. The aim of this Review is to summarize and discuss our knowledge of the role of macroautophagy in controlling a large panel of cell responses, from morphological and metabolic changes, to inflammation and senescence, for the integration of mechanical forces. Moreover, wherever possible, we also discuss the cell surface molecules and structures that sense mechanical forces upstream of macroautophagy.
Collapse
Affiliation(s)
- Aurore Claude-Taupin
- Institut Necker-Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, Université de Paris, 75015 Paris, France
| | - Patrice Codogno
- Institut Necker-Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, Université de Paris, 75015 Paris, France
| | - Nicolas Dupont
- Institut Necker-Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, Université de Paris, 75015 Paris, France
| |
Collapse
|
24
|
The role of physical cues in the development of stem cell-derived organoids. EUROPEAN BIOPHYSICS JOURNAL: EBJ 2021; 51:105-117. [PMID: 34120215 PMCID: PMC8964551 DOI: 10.1007/s00249-021-01551-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023]
Abstract
Organoids are a novel three-dimensional stem cells’ culture system that allows the in vitro recapitulation of organs/tissues structure complexity. Pluripotent and adult stem cells are included in a peculiar microenvironment consisting of a supporting structure (an extracellular matrix (ECM)-like component) and a cocktail of soluble bioactive molecules that, together, mimic the stem cell niche organization. It is noteworthy that the balance of all microenvironmental components is the most critical step for obtaining the successful development of an accurate organoid instead of an organoid with heterogeneous morphology, size, and cellular composition. Within this system, mechanical forces exerted on stem cells are collected by cellular proteins and transduced via mechanosensing—mechanotransduction mechanisms in biochemical signaling that dictate the stem cell specification process toward the formation of organoids. This review discusses the role of the environment in organoids formation and focuses on the effect of physical components on the developmental system. The work starts with a biological description of organoids and continues with the relevance of physical forces in the organoid environment formation. In this context, the methods used to generate organoids and some relevant published reports are discussed as examples showing the key role of mechanosensing–mechanotransduction mechanisms in stem cell-derived organoids.
Collapse
|
25
|
Gauthier R, Jeannin C, Attik N, Trunfio-Sfarghiu AM, Gritsch K, Grosgogeat B. Tissue Engineering for Periodontal Ligament Regeneration: Biomechanical Specifications. J Biomech Eng 2021; 143:030801. [PMID: 33067629 DOI: 10.1115/1.4048810] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Indexed: 11/08/2022]
Abstract
The periodontal biomechanical environment is very difficult to investigate. By the complex geometry and composition of the periodontal ligament (PDL), its mechanical behavior is very dependent on the type of loading (compressive versus tensile loading; static versus cyclic loading; uniaxial versus multiaxial) and the location around the root (cervical, middle, or apical). These different aspects of the PDL make it difficult to develop a functional biomaterial to treat periodontal attachment due to periodontal diseases. This review aims to describe the structural and biomechanical properties of the PDL. Particular importance is placed in the close interrelationship that exists between structure and biomechanics: the PDL structural organization is specific to its biomechanical environment, and its biomechanical properties are specific to its structural arrangement. This balance between structure and biomechanics can be explained by a mechanosensitive periodontal cellular activity. These specifications have to be considered in the further tissue engineering strategies for the development of an efficient biomaterial for periodontal tissues regeneration.
Collapse
Affiliation(s)
- R Gauthier
- Univ Lyon - Claude Bernard Lyon 1, UMR CNRS 5615, Laboratoire des Multimatériaux et Interfaces, Villeurbanne F-69622, France; Univ Lyon, Université Claude Bernard Lyon 1, Faculté d'Odontologie, Lyon 69008, France
| | - Christophe Jeannin
- Univ Lyon - Claude Bernard Lyon 1, UMR CNRS 5615, Laboratoire des Multimatériaux et Interfaces, Villeurbanne F-69622, France; Univ Lyon, Université Claude Bernard Lyon 1, Faculté d'Odontologie, Lyon 69008, France; Hospices Civils de Lyon, Service d'Odontologie, Lyon 69007, France
| | - N Attik
- Univ Lyon - Claude Bernard Lyon 1, UMR CNRS 5615, Laboratoire des Multimatériaux et Interfaces, Villeurbanne F-69622, France; Univ Lyon, Université Claude Bernard Lyon 1, Faculté d'Odontologie, Lyon 69008, France
| | | | - K Gritsch
- Univ Lyon - Claude Bernard Lyon 1, UMR CNRS 5615, Laboratoire des Multimatériaux et Interfaces, Villeurbanne F-69622, France; Univ Lyon, Université Claude Bernard Lyon 1, Faculté d'Odontologie, Lyon 69008, France; Hospices Civils de Lyon, Service d'Odontologie, Lyon 69007, France
| | - B Grosgogeat
- Univ Lyon - Claude Bernard Lyon 1, UMR CNRS 5615, Laboratoire des Multimatériaux et Interfaces, Villeurbanne F-69622, France; Univ Lyon, Université Claude Bernard Lyon 1, Faculté d'Odontologie, Lyon 69008, France; Hospices Civils de Lyon, Service d'Odontologie, Lyon 69007, France
| |
Collapse
|
26
|
Saghati S, Nasrabadi HT, Khoshfetrat AB, Moharamzadeh K, Hassani A, Mohammadi SM, Rahbarghazi R, Fathi Karkan S. Tissue Engineering Strategies to Increase Osteochondral Regeneration of Stem Cells; a Close Look at Different Modalities. Stem Cell Rev Rep 2021; 17:1294-1311. [PMID: 33547591 DOI: 10.1007/s12015-021-10130-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2021] [Indexed: 02/06/2023]
Abstract
The homeostasis of osteochondral tissue is tightly controlled by articular cartilage chondrocytes and underlying subchondral bone osteoblasts via different internal and external clues. As a correlate, the osteochondral region is frequently exposed to physical forces and mechanical pressure. On this basis, distinct sets of substrates and physicochemical properties of the surrounding matrix affect the regeneration capacity of chondrocytes and osteoblasts. Stem cells are touted as an alternative cell source for the alleviation of osteochondral diseases. These cells appropriately respond to the physicochemical properties of different biomaterials. This review aimed to address some of the essential factors which participate in the chondrogenic and osteogenic capacity of stem cells. Elements consisted of biomechanical forces, electrical fields, and biochemical and physical properties of the extracellular matrix are the major determinant of stem cell differentiation capacity. It is suggested that an additional certain mechanism related to signal-transduction pathways could also mediate the chondro-osteogenic differentiation of stem cells. The discovery of these clues can enable us to modulate the regeneration capacity of stem cells in osteochondral injuries and lead to the improvement of more operative approaches using tissue engineering modalities.
Collapse
Affiliation(s)
- Sepideh Saghati
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Tayefi Nasrabadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ali Baradar Khoshfetrat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Keyvan Moharamzadeh
- Hamdan Bin Mohammed College of Dental Medicine (HBMCDM), Mohammed Bin Rashid University of Medicine and Health Sciences (MBRU), Dubai, United Arab Emirates
| | - Ayla Hassani
- Chemical Engineering Faculty, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Seyedeh Momeneh Mohammadi
- Department of Anatomical Sciences, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Sonia Fathi Karkan
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
27
|
Purinergic Signaling Mediates PTH and Fluid Flow-Induced Osteoblast Proliferation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6674570. [PMID: 33575337 PMCID: PMC7864748 DOI: 10.1155/2021/6674570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/09/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023]
Abstract
Both parathyroid hormone (PTH) and mechanical signals are able to regulate bone growth and regeneration. They also can work synergistically to regulate osteoblast proliferation, but little is known about the mechanisms how PTH and mechanical signals interact with each other during this process. In this study, we investigated responses of MC3T3-E1 osteoblasts to PTH and oscillatory fluid flow. We found that osteoblasts are more sensitive to mechanical signals in the presence of PTH according to ERK1/2 phosphorylation, ATP release, CREB phosphorylation, and cell proliferation. PTH may also reduce the osteoblast refractory period after desensitization due to mechanical signals. We further found that the synergistic responses of osteoblasts to fluid flow or ATP with PTH had similar patterns, suggesting that synergy between fluid flow and PTH may be through the ATP pathway. After we inhibited ATP effects using apyrase in osteoblasts, their synergistic responses to mechanical stimulation and PTH were also inhibited. Additionally, knocking down P2Y2 purinergic receptors can significantly attenuate osteoblast synergistic responses to mechanical stimulation and PTH in terms of ERK1/2 phosphorylation, CREB phosphorylation, and cell proliferation. Thus, our results suggest that PTH enhances mechanosensitivity of osteoblasts via a mechanism involving ATP and P2Y2 purinergic receptors.
Collapse
|
28
|
Nishida T, Kubota S. Roles of CCN2 as a mechano-sensing regulator of chondrocyte differentiation. JAPANESE DENTAL SCIENCE REVIEW 2020; 56:119-126. [PMID: 33088364 PMCID: PMC7560579 DOI: 10.1016/j.jdsr.2020.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/07/2020] [Accepted: 07/20/2020] [Indexed: 12/17/2022] Open
Abstract
Cellular communication network factor 2 (CCN2) is a cysteine-rich secreted matricellular protein that regulates various cellular functions including cell differentiation. CCN2 is highly expressed under several types of mechanical stress, such as stretch, compression, and shear stress, in mesenchymal cells including chondrocytes, osteoblasts, and fibroblasts. In particular, CCN2 not only promotes cell proliferation and differentiation of various cells but also regulates the stability of mRNA of TRPV4, a mechanosensitive ion channel in chondrocytes. Of note, CCN2 behaves like a biomarker to sense suitable mechanical stress, because CCN2 expression is down-regulated when chondrocytes are subjected to excessive mechanical stress. These findings suggest that CCN2 is a mechano-sensing regulator. CCN2 expression is regulated by the activation of various mechano-sensing signaling pathways, e.g., mechanosensitive ion channels, integrin-focal adhesion-actin dynamics, Rho GTPase family members, Hippo-YAP signaling, and G protein-coupled receptors. This review summarizes the characterization of mechanoreceptors involved in CCN2 gene regulation and discusses the role of CCN2 as a mechano-sensing regulator of mesenchymal cell differentiation, with particular focus on chondrocytes.
Collapse
Affiliation(s)
- Takashi Nishida
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8525, Japan.,Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8525, Japan
| | - Satoshi Kubota
- Department of Biochemistry and Molecular Dentistry, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama 700-8525, Japan
| |
Collapse
|
29
|
The Analysis of Gap Junctional Intercellular Communication Among Osteocytes in Chick Calvariae by Fluorescence Recovery After Photobleaching. Methods Mol Biol 2020. [PMID: 32949375 DOI: 10.1007/7651_2020_322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
This chapter describes the use of fluorescence recovery after photobleaching (FRAP) for analyzing gap junctional intercellular communication (GJIC) among osteocytes in chick calvariae by confocal laser scanning microscope.
Collapse
|
30
|
Phosphodiesterase 10A Is a Mediator of Osteogenic Differentiation and Mechanotransduction in Bone Marrow-Derived Mesenchymal Stromal Cells. Stem Cells Int 2020; 2020:7865484. [PMID: 32587621 PMCID: PMC7294361 DOI: 10.1155/2020/7865484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/13/2020] [Accepted: 05/21/2020] [Indexed: 11/17/2022] Open
Abstract
Bone marrow-derived mesenchymal stromal cells (hMSCs) are capable of differentiating into the osteogenic lineage, and for osteogenic differentiation, mechanical loading is a relevant stimulus. Mechanotransduction leads to the formation of second messengers such as cAMP, cGMP, or Ca2+ influx resulting in the activation of transcription factors mediating gene regulation. The second messengers cAMP and cGMP are degraded by phosphodiesterase isoenzymes (PDE), but the role of these enzymes during osteogenic differentiation or mechanotransduction remains unclear. Here, we focused on the isoenzyme phosphodiesterase 10A (PDE10A) and its role during osteogenic commitment and mechanotransduction. We observed a time-dependent decrease of PDE10A expression in hMSC undergoing differentiation towards the osteogenic lineage. PDE10A inhibition by papaverine diminished osteogenic differentiation. While applying mechanical strain via cyclic stretching of hMSCs led to an upregulation of PDE10A gene expression, inhibition of PDE10A using the drug papaverine repressed expression of mechanoresponsive genes. We conclude that PDE10A is a modulator of osteogenic differentiation as well as mechanotransduction in hMSCs. Our data further suggests that the relative increase of cAMP, rather than the absolute cAMP level, is a key driver of the observed effects.
Collapse
|
31
|
Herrmann M, Engelke K, Ebert R, Müller-Deubert S, Rudert M, Ziouti F, Jundt F, Felsenberg D, Jakob F. Interactions between Muscle and Bone-Where Physics Meets Biology. Biomolecules 2020; 10:biom10030432. [PMID: 32164381 PMCID: PMC7175139 DOI: 10.3390/biom10030432] [Citation(s) in RCA: 103] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/27/2020] [Accepted: 03/05/2020] [Indexed: 02/06/2023] Open
Abstract
Muscle and bone interact via physical forces and secreted osteokines and myokines. Physical forces are generated through gravity, locomotion, exercise, and external devices. Cells sense mechanical strain via adhesion molecules and translate it into biochemical responses, modulating the basic mechanisms of cellular biology such as lineage commitment, tissue formation, and maturation. This may result in the initiation of bone formation, muscle hypertrophy, and the enhanced production of extracellular matrix constituents, adhesion molecules, and cytoskeletal elements. Bone and muscle mass, resistance to strain, and the stiffness of matrix, cells, and tissues are enhanced, influencing fracture resistance and muscle power. This propagates a dynamic and continuous reciprocity of physicochemical interaction. Secreted growth and differentiation factors are important effectors of mutual interaction. The acute effects of exercise induce the secretion of exosomes with cargo molecules that are capable of mediating the endocrine effects between muscle, bone, and the organism. Long-term changes induce adaptations of the respective tissue secretome that maintain adequate homeostatic conditions. Lessons from unloading, microgravity, and disuse teach us that gratuitous tissue is removed or reorganized while immobility and inflammation trigger muscle and bone marrow fatty infiltration and propagate degenerative diseases such as sarcopenia and osteoporosis. Ongoing research will certainly find new therapeutic targets for prevention and treatment.
Collapse
Affiliation(s)
- Marietta Herrmann
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, IZKF Research Group Tissue regeneration in musculoskeletal diseases, University Hospital Würzburg, University of Wuerzburg, 97070 Würzburg, Germany;
| | - Klaus Engelke
- Department of Medicine 3, FAU University Erlangen-Nürnberg and Universitätsklinikum Erlangen, 91054 Erlangen, Germany;
| | - Regina Ebert
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, University of Würzburg, IGZ, 97076 Würzburg, Germany; (R.E.)
| | - Sigrid Müller-Deubert
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, University of Würzburg, IGZ, 97076 Würzburg, Germany; (R.E.)
| | - Maximilian Rudert
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, University of Würzburg, 97074 Würzburg, Germany;
| | - Fani Ziouti
- Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany; (F.Z.); (F.J.)
| | - Franziska Jundt
- Department of Internal Medicine II, University Hospital Würzburg, 97080 Würzburg, Germany; (F.Z.); (F.J.)
| | - Dieter Felsenberg
- Privatpraxis für Muskel- und Knochenkrankheiten, 12163 Berlin Germany;
| | - Franz Jakob
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, University of Würzburg, IGZ, 97076 Würzburg, Germany; (R.E.)
- Orthopedic Department, Bernhard-Heine-Center for Locomotion Research, University of Würzburg, 97074 Würzburg, Germany;
- Correspondence:
| |
Collapse
|
32
|
Bordoni B. The Shape and Function of Solid Fascias Depend on the Presence of Liquid Fascias. Cureus 2020; 12:e6939. [PMID: 32190491 PMCID: PMC7067346 DOI: 10.7759/cureus.6939] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 02/10/2020] [Indexed: 12/21/2022] Open
Abstract
Scientific research is not a showcase of his own talent or own resources, it is a chance to improve common knowledge on certain topics for the collective well-being. A researcher should use multidisciplinarity to observe a phenomenon in its entirety and not only its alignment of thought, federations, committees, and knowledge; to get to understand it is necessary to exploit more tools and more disciplines. The article discusses the importance of the fluids (or liquid fascia) in maintaining the shape and function of the human body, as, currently, many texts forget how much body fluids are fundamental for understanding structural dynamics (bones and muscles, fibrils, and cells). By revisiting the current literature, the text wishes to highlight how the liquid fascia determines body adaptation in the presence of mechanical stress. Without fluids, there would be no body shape that we know.
Collapse
Affiliation(s)
- Bruno Bordoni
- Physical Medicine and Rehabilitation, Foundation Don Carlo Gnocchi, Milan, ITA
| |
Collapse
|
33
|
Bartolozzi A, Viti F, De Stefano S, Sbrana F, Petecchia L, Gavazzo P, Vassalli M. Development of label-free biophysical markers in osteogenic maturation. J Mech Behav Biomed Mater 2019; 103:103581. [PMID: 32090910 DOI: 10.1016/j.jmbbm.2019.103581] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 12/23/2022]
Abstract
The spatial and temporal changes of morphological and mechanical properties of living cells reflect complex functionally-associated processes. Monitoring these modifications could provide a direct information on the cellular functional state. Here we present an integrated biophysical approach to the quantification of the morphological and mechanical phenotype of single cells along a maturation pathway. Specifically, quantitative phase microscopy and single cell biomechanical testing were applied to the characterization of the maturation of human foetal osteoblasts, demonstrating the ability to identify effective label-free biomarkers along this fundamental biological process.
Collapse
Affiliation(s)
- Alice Bartolozzi
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy; Dipartimento di Ingegneria dell'Informazione, Università di Firenze, Florence, Italy
| | - Federica Viti
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy.
| | - Silvia De Stefano
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| | - Francesca Sbrana
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy; Schaefer South-East Europe Srl, Rovigo, Italy
| | - Loredana Petecchia
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| | - Paola Gavazzo
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| | - Massimo Vassalli
- Institute of Biophysics, National Research Council of Italy, Genoa, Italy
| |
Collapse
|
34
|
Ferrari A, Capitanio M, Vassalli M, Martinac B. Science by the sea: how nanoengineering met mechanobiology in Camogli. Biophys Rev 2019; 11:659-661. [PMID: 31529357 PMCID: PMC6815305 DOI: 10.1007/s12551-019-00598-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 09/04/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Aldo Ferrari
- EMPA, Swiss Federal Laboratories for Materials Science and Technology, 8600, Dübendorf, Switzerland.
| | - Marco Capitanio
- LENS-European Laboratory for Non-linear Spectroscopy, Via Nello Carrara 1, 50019, Sesto Fiorentino, Italy
- Department of Physics and Astronomy, University of Florence, Via Sansone 1, 50019, Sesto Fiorentino, Italy
| | - Massimo Vassalli
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via De Marini 6, Genoa, Italy
| | - Boris Martinac
- The Victor Chang Cardiac Research Institute, Lowy Packer Building, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, University of New South Wales, Darlinghurst, NSW, 2010, Australia
| |
Collapse
|