1
|
Tilton M, Weivoda M, Astudillo Potes M, Gingery A, Liu AY, Tchkonia T, Lu L, Kirkland JL. Stiffening symphony of aging: Biophysical changes in senescent osteocytes. Aging Cell 2024; 23:e14421. [PMID: 39582140 PMCID: PMC11634739 DOI: 10.1111/acel.14421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/23/2024] [Accepted: 11/13/2024] [Indexed: 11/26/2024] Open
Abstract
Senescent osteocytes are key contributors to age-related bone loss and fragility; however, the impact of mechanobiological changes in these cells remains poorly understood. This study provides a novel analysis of these changes in primary osteocytes following irradiation-induced senescence. By integrating subcellular mechanical measurements with gene expression analyses, we identified significant, time-dependent alterations in the mechanical properties of senescent bone cells. Increases in classical markers such as SA-β-Gal activity and p16Ink4a expression levels confirmed the senescence status post-irradiation. Our key findings include a time-dependent increase in cytoskeletal Young's modulus and altered viscoelastic properties of the plasma membrane, affecting the contractility of primary osteocytes. Additionally, we observed a significant increase in Sclerostin (Sost) expression 21 days post-irradiation. These biophysical changes may impair osteocyte mechanosensation and mechanotransduction, contributing to bone fragility. This is the first study to time-map senescence-associated mechanical changes in the osteocyte cytoskeleton. Our findings highlight the potential of biophysical markers as indicators of cellular senescence, providing more specificity than traditional, variable biomolecular markers. We believe these results may support biomechanical stimulation as a potential therapeutic strategy to rejuvenate aging osteocytes and enhance bone health.
Collapse
Affiliation(s)
- Maryam Tilton
- Walker Department of Mechanical EngineeringThe University of Texas at AustinAustinTexasUSA
| | - Megan Weivoda
- Department of HematologyMayo ClinicRochesterMinnesotaUSA
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesotaUSA
| | - Maria Astudillo Potes
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Department of Orthopedic SurgeryMayo ClinicRochesterMinnesotaUSA
| | - Anne Gingery
- Department of Biochemistry and Molecular BiologyMayo ClinicRochesterMinnesotaUSA
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Department of Orthopedic SurgeryMayo ClinicRochesterMinnesotaUSA
| | | | - Tamara Tchkonia
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - Lichun Lu
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Department of Orthopedic SurgeryMayo ClinicRochesterMinnesotaUSA
| | - James L. Kirkland
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
2
|
Gupta A, Saha S, Das A, Roy Chowdhury A. Evaluating the influence on osteocyte mechanobiology within the lacunar-canalicular system for varying lacunar equancy and perilacunar elasticity: A multiscale fluid-structure interaction analysis. J Mech Behav Biomed Mater 2024; 160:106767. [PMID: 39393133 DOI: 10.1016/j.jmbbm.2024.106767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/26/2024] [Accepted: 10/03/2024] [Indexed: 10/13/2024]
Abstract
The lacunar morphology and perilacunar tissue properties of osteocytes in bone can vary under different physiological and pathological conditions. How these alterations collectively change the overall micromechanics of osteocytes in the lacunar-canalicular system (LCS) of an osteon still requires special focus. Therefore, a Haversian canal and LCS-based osteon model was established to evaluate the changes in the hydrodynamic environment around osteocytes under physiological loading using fluid-structure interaction analysis, followed by a sub-modelled finite element analysis to assess the mechanical responses of osteocytes and their components. Osteocytes were modelled with detailed configurations, including cytoplasm, nucleus, and cytoskeleton, and parametric variations in lacunar equancy (L.Eq) and perilacunar elasticity (Pl.E) were considered within the osteon model. The study aimed to conduct a comparative study among osteon models with varying L. Eq and Pl. E to check the resulting differences in osteocyte mechanobiology. The results demonstrated that the average mechanical stimulation of each subcellular component of osteocytes increased with decreases in L. Eq and Pl. E, reflecting conditions typically seen in young, healthy bone as per previous literature. However, hydrodynamic responses, such as fluid flow and fluid shear stress on osteocytes, varied proportionally with the elasticity difference between the bone matrix and the perilacunar region during Pl. E variation. Additionally, the findings revealed that a minimal percentage of energy was used to transmit mechanical responses through microtubules from the cell membrane to the nucleus, and this energy percentage increased with higher L. Eq. The outcomes of the study could help to quantify how the osteocyte microenvironment and its mechanosensitivity within cortical bone changes with L. Eq and Pl. E alterations in different bone conditions, from young to aged and healthy to diseased.
Collapse
Affiliation(s)
- Abhisek Gupta
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, India
| | - Subrata Saha
- Department of Restorative Dentistry, University of Washington, Seattle, WA, USA
| | - Apurba Das
- Department of Mechanical Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, India
| | - Amit Roy Chowdhury
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, India.
| |
Collapse
|
3
|
Azari F, Hemmatian H, Banerjee A, van Lenthe GH. Subchondral Bone Osteocyte Lacunae Morphology in End-Stage Osteoarthritis of the Human Tibial Plateau. Calcif Tissue Int 2024; 115:78-84. [PMID: 38753025 DOI: 10.1007/s00223-024-01226-z] [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: 11/17/2023] [Accepted: 05/06/2024] [Indexed: 06/06/2024]
Abstract
Subchondral bone remodeling, mediated by osteocytes within the lacuno-canalicular network, plays a crucial role in osteoarthritis (OA) progression. Following cell death, lacunae preserve integrity, offering insights into bone remodeling mechanisms. Limited and controversial data on osteocyte lacuna morphology in OA result from small sample sizes and two-dimensional (2D) techniques that have been used thus far. This study aimed to quantify three-dimensional (3D) osteocyte lacunar characteristics at well-defined tibial plateau locations, known to be differently affected by OA. Specifically, 11 tibial plateaus were obtained from end-stage knee-OA patients with varus deformity. Each plateau provided one sample from the less affected lateral compartment and two samples from the medial compartment, at minimum and maximum bone volume fraction (BV/TV) locations. High-resolution desktop micro-computed tomography (micro-CT) at 0.7 μm voxel resolution imaged the 33 samples. Lacuna number density (Lc.N/BV) and lacuna volume density (Lc.TV/BV) were significantly lower (p < 0.02) in samples from the medial side with maximum BV/TV compared to lateral side samples. In the medial compartment at maximum local BV/TV, mean lacuna volume (Lc.V), total lacuna volume (Lc.TV), and Lc.TV/BV were significantly (p < 0.001) lower than in the region with minimum BV/TV. Lc.N/BV was also significantly lower (p < 0.02) at the maximum local BV/TV location compared to the region with minimum BV/TV. Our findings suggest that subchondral bone lacunae adapt to the changing loads in end-stage OA.
Collapse
Affiliation(s)
- Fahimeh Azari
- Biomechanics Section, Dept. Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Haniyeh Hemmatian
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg, VIC, Australia
| | - Anik Banerjee
- Biomechanics Section, Dept. Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - G Harry van Lenthe
- Biomechanics Section, Dept. Mechanical Engineering, KU Leuven, Leuven, Belgium.
| |
Collapse
|
4
|
Ravazzano L, Colaianni G, Tarakanova A, Xiao YB, Grano M, Libonati F. Multiscale and multidisciplinary analysis of aging processes in bone. NPJ AGING 2024; 10:28. [PMID: 38879533 PMCID: PMC11180112 DOI: 10.1038/s41514-024-00156-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/07/2024] [Indexed: 06/19/2024]
Abstract
The world population is increasingly aging, deeply affecting our society by challenging our healthcare systems and presenting an economic burden, thus turning the spotlight on aging-related diseases: exempli gratia, osteoporosis, a silent disease until you suddenly break a bone. The increase in bone fracture risk with age is generally associated with a loss of bone mass and an alteration in the skeletal architecture. However, such changes cannot fully explain increased fragility with age. To successfully tackle age-related bone diseases, it is paramount to comprehensively understand the fundamental mechanisms responsible for tissue degeneration. Aging mechanisms persist at multiple length scales within the complex hierarchical bone structure, raising the need for a multiscale and multidisciplinary approach to resolve them. This paper aims to provide an overarching analysis of aging processes in bone and to review the most prominent outcomes of bone aging. A systematic description of different length scales, highlighting the corresponding techniques adopted at each scale and motivating the need for combining diverse techniques, is provided to get a comprehensive description of the multi-physics phenomena involved.
Collapse
Affiliation(s)
- Linda Ravazzano
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Via Rubattino 81, Milano, 20134, Italy
| | - Graziana Colaianni
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Piazza Giulio Cesare 11, Bari, 70124, Italy
| | - Anna Tarakanova
- School of Mechanical, Aerospace, and Manufacturing Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, 06269, CT, USA
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Road, Unit 3247, CT, 06269, Storrs, USA
| | - Yu-Bai Xiao
- School of Mechanical, Aerospace, and Manufacturing Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, 06269, CT, USA
| | - Maria Grano
- Department of Precision and Regenerative Medicine and Ionian Area (DiMePRe-J), University of Bari Aldo Moro, Piazza Giulio Cesare 11, Bari, 70124, Italy
| | - Flavia Libonati
- Center for Nano Science and Technology@PoliMi, Istituto Italiano di Tecnologia, Via Rubattino 81, Milano, 20134, Italy.
- Department of Mechanical, Energy, Management and Transport Engineering - DIME, University of Genova, Via all'Opera Pia 15, Genova, 16145, Italy.
| |
Collapse
|
5
|
Migotsky N, Kumar S, Shuster JT, Coulombe JC, Senwar B, Gestos AA, Farber CR, Ferguson VL, Silva MJ. Multi-scale cortical bone traits vary in females and males from two mouse models of genetic diversity. JBMR Plus 2024; 8:ziae019. [PMID: 38634075 PMCID: PMC11021811 DOI: 10.1093/jbmrpl/ziae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 02/08/2024] [Indexed: 04/19/2024] Open
Abstract
Understanding the genetic basis of cortical bone traits can allow for the discovery of novel genes or biological pathways regulating bone health. Mice are the most widely used mammalian model for skeletal biology and allow for the quantification of traits that cannot easily be evaluated in humans, such as osteocyte lacunar morphology. The goal of our study was to investigate the effect of genetic diversity on multi-scale cortical bone traits of 3 long bones in skeletally-mature mice. We measured bone morphology, mechanical properties, material properties, lacunar morphology, and mineral composition of mouse bones from 2 populations of genetic diversity. Additionally, we compared how intrabone relationships varied in the 2 populations. Our first population of genetic diversity included 72 females and 72 males from the 8 inbred founder strains used to create the Diversity Outbred (DO) population. These 8 strains together span almost 90% of the genetic diversity found in mice (Mus musculus). Our second population of genetic diversity included 25 genetically unique, outbred females and 25 males from the DO population. We show that multi-scale cortical bone traits vary significantly with genetic background; heritability values range from 21% to 99% indicating genetic control of bone traits across length scales. We show for the first time that lacunar shape and number are highly heritable. Comparing the 2 populations of genetic diversity, we show that each DO mouse does not resemble a single inbred founder, but instead the outbred mice display hybrid phenotypes with the elimination of extreme values. Additionally, intrabone relationships (eg, ultimate force vs. cortical area) were mainly conserved in our 2 populations. Overall, this work supports future use of these genetically diverse populations to discover novel genes contributing to cortical bone traits, especially at the lacunar length scale.
Collapse
Affiliation(s)
- Nicole Migotsky
- Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, United States
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States
| | - Surabhi Kumar
- Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, United States
| | - John T Shuster
- Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, United States
| | - Jennifer C Coulombe
- Department of Mechanical Engineering, University of Colorado, Boulder, CO 80309, United States
| | - Bhavya Senwar
- Department of Mechanical Engineering, University of Colorado, Boulder, CO 80309, United States
| | - Adrian A Gestos
- Materials Instrumentation and Multimodal Imaging Core, University of Colorado, Boulder, CO 80309, United States
| | - Charles R Farber
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA 22908, United States
| | - Virginia L Ferguson
- Department of Mechanical Engineering, University of Colorado, Boulder, CO 80309, United States
- Materials Instrumentation and Multimodal Imaging Core, University of Colorado, Boulder, CO 80309, United States
| | - Matthew J Silva
- Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110, United States
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, United States
| |
Collapse
|
6
|
Kim S, Jang S, Lee O. Single fiber curvature for muscle impairment assessment: Phase contrast imaging of stroke-induced animals. Microsc Res Tech 2024; 87:705-715. [PMID: 37983687 DOI: 10.1002/jemt.24459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 10/15/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023]
Abstract
There are technical challenges in imaging studies that can three-dimensionally (3D) analyze a single fiber (SF) to observe the functionality of the entire muscle after stroke. This study proposes a 3D assessment technique that only segments the SF of the right stroke-induced soleus muscle of a gerbil using synchrotron radiation x-ray microcomputed tomography (SR-μCT), which is capable of muscle structure analysis. Curvature damage in the SF of the left soleus muscle (impaired) progressed at 7-day intervals after the stroke in the control; particularly on the 7 days (1 week) and 14 days (2 weeks), as observed through visualization analysis. At 2 weeks, the SF volume was significantly reduced in the control impaired group (p = .033), and was significantly less than that in the non-impaired group (p = .009). We expect that animal post-stroke studies will improve the basic field of rehabilitation therapy by diagnosing the degree of SF curvature. RESEARCH HIGHLIGHTS: Muscle evaluation after ischemic stroke using synchrotron radiation x-ray microcomputed tomography (SR-μCT). Curvature is measured by segmenting a single fiber (SF) in the muscle. Structural changes in the SF of impaired gerbils at 7-day intervals were assessed.
Collapse
Affiliation(s)
- Subok Kim
- Department of Software Convergence, Graduate School, Soonchunhyang University, Asan, Republic of Korea
| | - Sanghun Jang
- Department of Physical Therapy, College of Health and Life Sciences, Korea National University of Transportation, Jeungpyeong-gun, Republic of Korea
| | - Onseok Lee
- Department of Software Convergence, Graduate School, Soonchunhyang University, Asan, Republic of Korea
- Department of Medical IT Engineering, College of Medical Sciences, Soonchunhyang University, Asan, Republic of Korea
| |
Collapse
|
7
|
Gehre C, Qiu W, Klaus Jäger P, Wang X, Marques FC, Nelson BJ, Müller R, Qin XH. Guiding bone cell network formation in 3D via photosensitized two-photon ablation. Acta Biomater 2024; 174:141-152. [PMID: 38061678 DOI: 10.1016/j.actbio.2023.11.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/18/2023]
Abstract
A long-standing challenge in skeletal tissue engineering is to reconstruct a three-dimensionally (3D) interconnected bone cell network in vitro that mimics the native bone microarchitecture. While conventional hydrogels are extensively used in studying bone cell behavior in vitro, current techniques lack the precision to manipulate the complex pericellular environment found in bone. The goal of this study is to guide single bone cells to form a 3D network in vitro via photosensitized two-photon ablation of microchannels in gelatin methacryloyl (GelMA) hydrogels. A water-soluble two-photon photosensitizer (P2CK) was added to soft GelMA hydrogels to enhance the ablation efficiency. Remarkably, adding 0.5 mM P2CK reduced the energy dosage threshold five-fold compared to untreated controls, enabling more cell-compatible ablation. By employing low-energy ablation (100 J/cm2) with a grid pattern of 1 µm wide and 30 µm deep microchannels, we induced dendritic outgrowth in human mesenchymal stem cells (hMSC). After 7 days, the cells successfully utilized the microchannels and formed a 3D network. Our findings reveal that cellular viability after low-energy ablation was comparable to unablated controls, whereas high-energy ablation (500 J/cm2) resulted in 42 % cell death. Low-energy grid ablation significantly promoted network formation and >40 µm long protrusion outgrowth. While the broad-spectrum matrix metalloproteinase inhibitor (GM6001) reduced cell spreading by inhibiting matrix degradation, cells invaded the microchannel grid with long protrusions. Collectively, these results emphasize the potential of photosensitized two-photon hydrogel ablation as a high-precision tool for laser-guided biofabrication of 3D cellular networks in vitro. STATEMENT OF SIGNIFICANCE: The inaccessible nature of osteocyte networks in bones renders fundamental research on skeletal biology a major challenge. This limit is partly due to the lack of high-resolution tools that can manipulate the pericellular environment in 3D cultures in vitro. To create bone-like cellular networks, we employ a two-photon laser in combination with a two-photon sensitizer to erode microchannels with low laser dosages into GelMA hydrogels. By providing a grid of microchannels, the cells self-organized into a 3D interconnected network within days. Laser-guided formation of 3D networks from single cells at micron-scale resolution is demonstrated for the first time. In future, we envisage in vitro generation of bone cell networks with user-dictated morphologies for both fundamental and translational bone research.
Collapse
Affiliation(s)
| | - Wanwan Qiu
- Institute for Biomechanics, ETH Zurich, Zürich, Switzerland
| | | | - Xiaopu Wang
- Institute of Robotics and Intelligent Systems, Zürich, Switzerland
| | | | - Bradley J Nelson
- Institute of Robotics and Intelligent Systems, Zürich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zürich, Switzerland
| | - Xiao-Hua Qin
- Institute for Biomechanics, ETH Zurich, Zürich, Switzerland.
| |
Collapse
|
8
|
Ahmed F, Minamizaki T, Aubin JE, Damayanti MA, Yoshiko Y. Large scale analysis of osteocyte lacunae in klotho hypomorphic mice using high-resolution micro-computed tomography. Ann Anat 2023; 250:152142. [PMID: 37572763 DOI: 10.1016/j.aanat.2023.152142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/14/2023] [Accepted: 07/26/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND Osteocytes are the most abundant cell type in adult bone, and the morphological characteristics of osteocytes and their lacunae appear to influence bone mass and fragility. Although conventional computed tomography (CT) has contributed greatly to advances in bone morphometry, capturing details of the entire hierarchical assembly, e.g., osteocyte lacuna parameters, has been limited by the analytical performance of CT (> 1 µm resolution). METHODS We used high-resolution (700 nm) micro-CT to evaluate and compare the osteocyte lacuna parameters over a large scale, i.e., in a maximum of about 45,700 lacunae (average), in tibial metaphyseal cortical bones of wild-type (WT) and αKlotho-hypomorphic (kl/kl) mice, the latter a model that exhibits osteopenia and aberrant osteocytes. RESULTS Of osteocyte lacuna parameters, lacunar surface per lacunar volume were significantly lower and lacuna diameter were significantly larger in kl/kl mice compared to WT mice. By analysis of individual osteocyte lacunae, we found that lacunar sphericity in kl/kl mice was higher than that in WT mice, and the diameters in the major and the minor axes were respectively lower and higher in kl/kl mice, especially at the proximal site of the region of interest. CONCLUSION We successfully assessed osteocyte lacuna parameters on the largest scale in mice reported to date and found that the shape of osteocyte lacunae of kl/kl mice are significantly different from those of WT mice. Although the mechanisms underlying the lacunar shape differences observed are not yet clear, changes in lacunar geometry are known to affect the transitions of strains to the osteocyte microenvironment and likely local osteocyte response(s). Thus, the fact that the differences are limited to the mesial region near the primary spongiosa suggests the likelihood of site-specific anomalies in mechanosensitive effects in kl/kl osteocytes with consequent site-specific effects bone metabolism and function.
Collapse
Affiliation(s)
- Faisal Ahmed
- Department of Calcified Tissue Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Tomoko Minamizaki
- Department of Calcified Tissue Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Jane E Aubin
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Merry Annisa Damayanti
- Department of Calcified Tissue Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan; Department of Dentomaxillofacial Radiology, Faculty of Dentistry, Padjadjaran University, Bandung, Indonesia
| | - Yuji Yoshiko
- Department of Calcified Tissue Biology, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan.
| |
Collapse
|
9
|
McGuire BD, Dees A, Hao L, Buckendahl P, Ogilvie AR, Sun H, Rezaee T, Barrett LO, Karim L, Dominguez-Bello MG, Bello NT, Shapses SA. A vitamin D deficient diet increases weight gain and compromises bone biomechanical properties without a reduction in BMD in adult female mice. J Steroid Biochem Mol Biol 2023; 231:106314. [PMID: 37088440 DOI: 10.1016/j.jsbmb.2023.106314] [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: 02/04/2023] [Revised: 03/27/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
Vitamin D contributes to the development and maintenance of bone. Evidence suggests vitamin D status can also alter energy balance and gut health. In young animals, vitamin D deficiency (VDD) negatively affects bone mineral density (BMD) and bone microarchitecture, and these effects may also occur due to chronic ethanol intake. However, evidence is limited in mature models, and addressing this was a goal of the current study. Seven-month-old female C57BL/6 mice (n = 40) were weight-matched and randomized to one of four ad libitum diets: control, alcohol (Alc), vitamin D deficient (0 IU/d), or Alc+VDD for 8 weeks. A purified (AIN-93) diet was provided with water or alcohol (10 %) ad libitum. Body weight and food intake were recorded weekly, and feces were collected at 0, 4, and 8 weeks. At the age of 9 months, intestinal permeability was assessed by oral gavage of fluorescein isothiocyanate-dextran. Thereafter, bone mineral density (BMD) was measured by dual-energy X-ray absorptiometry. The microarchitecture of the distal femur was assessed by micro-computed tomography and biomechanical properties were evaluated by cyclic reference point indentation. VDD did not affect BMD or most bone microarchitecture parameters, however, the polar moment of inertia (p < 0.05) was higher in the VDD groups compared to vitamin D sufficient groups. VDD mice also had lower whole bone water content (p < 0.05) and a greater average unloading slope (p < 0.01), and energy dissipated (p < 0.01), indicating the femur displayed a brittle phenotype. In addition, VDD caused a greater increase in energy intake (p < 0.05), weight gain (p < 0.05), and a trend for higher intestinal permeability (p = 0.08). The gut microbiota of the VDD group had a reduction in alpha diversity (p < 0.05) and a lower abundance of ASVs from Rikenellaceae, Clostridia_UCG-014, Oscillospiraceae, and Lachnospiraceae (p < 0.01). There was little to no effect of alcohol supplementation on outcomes. Overall, these findings suggest that vitamin D deficiency causes excess weight gain and reduces the biomechanical strength of the femur as indicated by the higher average unloading slope and energy dissipated without an effect on BMD in a mature murine model.
Collapse
Affiliation(s)
- Brandon D McGuire
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Azra Dees
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Lihong Hao
- Department of Animal Sciences, Rutgers University, New Brunswick, NJ, USA
| | | | - Anna R Ogilvie
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA
| | - Haipeng Sun
- Department of Microbiology and Biochemistry, New Brunswick, NJ, USA
| | - Taraneh Rezaee
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | - Leland O Barrett
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | - Lamya Karim
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA 02747, USA
| | - Maria Gloria Dominguez-Bello
- Department of Microbiology and Biochemistry, New Brunswick, NJ, USA; NJ Institute of Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA
| | - Nicholas T Bello
- Department of Animal Sciences, Rutgers University, New Brunswick, NJ, USA; NJ Institute of Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA
| | - Sue A Shapses
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, USA; NJ Institute of Food, Nutrition and Health, Rutgers University, New Brunswick, NJ, USA; Department of Medicine, Rutgers-Robert Wood Johnson Univ. Hospital, New Brunswick, NJ, USA.
| |
Collapse
|
10
|
Heveran CM, Boerckel JD. Osteocyte Remodeling of the Lacunar-Canalicular System: What's in a Name? Curr Osteoporos Rep 2023; 21:11-20. [PMID: 36512204 PMCID: PMC11223162 DOI: 10.1007/s11914-022-00766-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/18/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Osteocytes directly modify the bone surrounding the expansive lacunar-canalicular system (LCS) through both resorption and deposition. The existence of this phenomenon is now widely accepted, but is referred to as "osteocyte osteolysis," "LCS remodeling," and "perilacunar remodeling," among other names. The uncertainty in naming this physiological process reflects the many persistent questions about why and how osteocytes interact with local bone matrix. The goal of this review is to examine the purpose and nature of LCS remodeling and its impacts on multiscale bone quality. RECENT FINDINGS While LCS remodeling is clearly important for systemic calcium mobilization, this process may have additional potential drivers and may impact the ability of bone to resist fracture. There is abundant evidence that the osteocyte can resorb and replace bone mineral and does so outside of extreme challenges to mineral homeostasis. The impacts of the osteocyte on organic matrix are less certain, especially regarding whether osteocytes produce osteoid. Though multiple lines of evidence point towards osteocyte production of organic matrix, definitive work is needed. Recent high-resolution imaging studies demonstrate that LCS remodeling influences local material properties. The role of LCS remodeling in the maintenance and deterioration of bone matrix quality in aging and disease are active areas of research. In this review, we highlight current progress in understanding why and how the osteocyte removes and replaces bone tissue and the consequences of these activities to bone quality. We posit that answering these questions is essential for evaluating whether, how, when, and why LCS remodeling may be manipulated for therapeutic benefit in managing bone fragility.
Collapse
Affiliation(s)
- C M Heveran
- Department of Mechanical & Industrial Engineering, Montana State University, Bozeman, USA.
| | - J D Boerckel
- Department of Orthopaedic Surgery, Department of Bioengineering, University of Pennsylvania School of Medicine, Philadelphia, USA.
| |
Collapse
|
11
|
Fracture Healing in Elderly Mice and the Effect of an Additional Severe Blood Loss: A Radiographic and Biomechanical Murine Study. BIOENGINEERING (BASEL, SWITZERLAND) 2023; 10:bioengineering10010070. [PMID: 36671642 PMCID: PMC9855159 DOI: 10.3390/bioengineering10010070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 01/07/2023]
Abstract
Femoral fractures and severe bleeding frequently occur in old patients showing a delayed healing. As there are no studies investigating the combined effect of high age and severe blood loss on fracture healing, this was examined radiographically and biomechanically in this study. Therefore, young and old male mice were randomly assigned to three operation groups. In the fracture group (Fx), external fixator and osteotomy were applied to the femur. The combined trauma group (THFx) additionally received a pressure-controlled hemorrhage. Sham animals were only implanted with arterial catheter and external fixator. Sacrifice was performed after three weeks and bone healing was evaluated radiologically via µCT, as well as biomechanically using a three-point bending test. A decreased share of callus/total bone volume was observed in old mice with blood loss compared to old Fx. Hemorrhagic shock also reduced the trabecular number in old mice compared to Fx and young THFx. Moreover, a lower elastic limit in old Sham mice without fracture was revealed. Fracture combined with a high loss of blood further reduced the elastic limit in old mice compared to isolated Fx in old animals. In conclusion, this study showed that severe blood loss has a higher negative effect in old mice compared to young ones.
Collapse
|
12
|
Khass M, Rashid H, Burrows PD, Javed A, Schroeder HW. Loss of early B cell protein λ5 decreases bone mass and accelerates skeletal aging. Front Immunol 2022; 13:906649. [PMID: 36189270 PMCID: PMC9516392 DOI: 10.3389/fimmu.2022.906649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/16/2022] [Indexed: 11/13/2022] Open
Abstract
The early B cell protein λ5 is an essential component of the surrogate light chain and the preB cell receptor (preBCR), which is critical for optimal B cell development. To investigate the effect of λ5 and/or B cells on bone acquisition over time, we developed a panel of JH -/- , λ5-/-, JH -/- λ5-/-, and wild-type (WT) BALB/c mice and then studied postnatal bone development and aging in these mice at one, six, twelve, and twenty-two months of age. The trabecular bone volume over total volume (BV/TV) in JH -/- mice was similar to WT mice at all ages. In contrast, at six months of age and thereafter, λ5-/- and JH -/- λ5-/- mice demonstrated a severe decrease in trabecular bone mass. Surprisingly, bone mass in six-month-old λ5-/- and JH -/- λ5-/- mice was similar to or even lower than in aged (twenty-two-months) WT mice, suggesting accelerated skeletal aging. The postnatal development and the acquisition of cortical bone mass in JH -/- λ5-/- mice were generally comparable to WT. However, JH -/- λ5-/- mice showed a significant decrease in cortical BV/TV at six- and twelve months of age. To examine the contribution of λ5 and B cells to postnatal bone synthesis, we separately transplanted whole bone marrow cells from JH -/- λ5-/- and WT mice into irradiated JH -/- λ5-/- and WT recipients. WT recipients of JH -/- λ5-/- marrow cells failed to show acquisition of trabecular bone mass, whereas transplanting WT marrow cells into JH -/- λ5-/- recipients led to the recovery of trabecular bone mass. Transfer of WT marrow cells into JH -/- λ5-/- mice promoted synthesis of new cortical and trabecular bone. Our findings indicate that λ5 plays a major role in preserving bone mass during postnatal development and skeletal aging which is distinct from its role in B cell development. The absence of both λ5 and B cells in JH -/- λ5-/- mice leads to delayed acquisition of cortical bone during postnatal development. Dissecting the mechanism(s) by which λ5 regulates bone homeostasis may provide new avenues for the treatment of age-related loss of bone mass and osteoporosis.
Collapse
Affiliation(s)
- Mohamed Khass
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Harunur Rashid
- Department of Oral and Maxillofacial Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Peter D. Burrows
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Amjad Javed
- Department of Oral and Maxillofacial Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Harry W. Schroeder
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
13
|
Quantifying how altered lacunar morphology and perilacunar tissue properties influence local mechanical environment of osteocyte lacunae using finite element modeling. J Mech Behav Biomed Mater 2022; 135:105433. [DOI: 10.1016/j.jmbbm.2022.105433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/09/2022] [Accepted: 08/26/2022] [Indexed: 11/23/2022]
|
14
|
Allison H, O'Sullivan L, McNamara L. Temporal changes in cortical microporosity during estrogen deficiency associated with perilacunar resorption and osteocyte apoptosis: A pilot study. Bone Rep 2022; 16:101590. [PMID: 35663377 PMCID: PMC9156983 DOI: 10.1016/j.bonr.2022.101590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 02/07/2023] Open
Abstract
Osteocytes can actively regulate bone microporosity, through either perilacunar resorption or micropetrosis following apoptosis. Osteocyte apoptosis is more prevalent in estrogen deficiency and changes in the lacunar-canalicular network of osteocytes have been reported. Temporal changes in bone mineralisation and osteocytes cellular strains occur, which might be associated with osteocyte-driven microporosity changes, although time dependant changes in bone microporosity are not yet fully understood. In this pilot study we conducted micro-CT analysis, backscatter electron imaging and histological analysis of femoral cortical bone form an ovariectomized rat model of osteoporosis to investigate whether estrogen deficiency causes temporal changes in lacunar and vascular porosity. We also assessed MMP14 expression, lacunar occupancy and mineral infilling, as indicators of perilacunar resorption and micropetrosis. We report temporal changes in cortical microporosity in estrogen deficiency. Specifically, canalicular and vascular porosity initially increased (4 weeks post-OVX), coinciding with the period of rapid bone loss, whereas in the longer term (14 weeks post-OVX) lacunar and canalicular diameter decreased. Interestingly, these changes coincided with an increased prevalence of empty lacunae and osteocyte lacunae were observed to be more circular with a mineralised border around the lacunar space. In addition we report an increase in MMP14+ osteocytes, which also suggests active matrix degradation by these cells. Together these results provide an insight into the temporal changes in cortical microporosity during estrogen deficiency and suggest the likelihood of occurrence of both perilacunar resorption and osteocyte apoptosis leading to micropetrosis. We propose that microporosity changes arise due to processes driven by distinct populations of osteocytes, which are either actively resorbing their matrix or have undergone apoptosis and are infilling lacunae by micropetrosis.
Collapse
Key Words
- BMDD, Bone mineral density distribution
- BSEM
- BSEM, Backscattered scanning electron microscopy
- BV, Bone volume
- Dm, Diameter
- Estrogen deficiency
- Lacunar
- Lc, Lacunar
- MMP, Matrix metalloproteases
- Micro-CT, Micro computed tomography
- Microporosity
- OVX, Ovariectomized
- PLR, Perilacunar resorption
- Sp, spacing
- TRAP, Tartrate-resistant acid phosphatase
- TV, Total volume
- Tb, Trabecular
- Th, Thickness
- V Ca, Vascular canal
- Vascular canals
- micro-CT
- με, Microstrain (ε ×10−6)
Collapse
Affiliation(s)
- H. Allison
- Mechanobiology and Medical Devices Research Group (MMDRG), Centre for Biomechanics Research (BioMEC), Biomedical Engineering, College of Science and Engineering, National University of Ireland Galway, Ireland
| | - L.M. O'Sullivan
- Mechanobiology and Medical Devices Research Group (MMDRG), Centre for Biomechanics Research (BioMEC), Biomedical Engineering, College of Science and Engineering, National University of Ireland Galway, Ireland
| | - L.M. McNamara
- Mechanobiology and Medical Devices Research Group (MMDRG), Centre for Biomechanics Research (BioMEC), Biomedical Engineering, College of Science and Engineering, National University of Ireland Galway, Ireland
| |
Collapse
|
15
|
Cen H, Gong H, Liu H, Jia S, Wu X, Fan Y. A Comparative Study on the Multiscale Mechanical Responses of Human Femoral Neck Between the Young and the Elderly Using Finite Element Method. Front Bioeng Biotechnol 2022; 10:893337. [PMID: 35600894 PMCID: PMC9117745 DOI: 10.3389/fbioe.2022.893337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Femoral neck fracture (FNF) is the most serious bone disease in the elderly population. The multiscale mechanical response is a key to predicting the strength of the femoral neck, assessing the risk of FNF, and exploring the role of mechanosensation and mechanotransmission in bone remodeling, especially in the context of aging bone.Methods: Multiscale finite element (FE) models of the proximal femur for both young and elderly people were developed. The models included organ scale (proximal femur), tissue scale (cortical bone), tissue element scale (osteon), and cell scale [osteocyte lacuna-canalicular network (LCN) and extracellular matrix (ECM), OLCEM]. The mechanical responses of cortical bone and osteocytes in the mid-femoral neck and the differences in mechanical responses between these two scales were investigated.Results: The mechanical responses of cortical bone and osteocyte showed significant differences between the elderly and the young. The minimum principal strains and mean SEDs of cortical bone in the elderly were 2.067–4.708 times and 3.093–14.385 times of the values in the young, respectively; the minimum principal strains and mean SEDs of osteocyte in the elderly were 1.497–3.246 times and 3.044–12 times of the values in the young, respectively; the amplification factors of minimum principal strain in the inferior (Inf), anterior (Ant), and posterior (Post) quadrants in the young were 1.241–1.804 times of the values in the elderly, but the amplification factor of minimum principal strain in the superior (Sup) quadrant was 87.4% of the value in the elderly; the amplification factors of mean SED in the young were 1.124–9.637 times of the values in the elderly.Conclusion: The mass and bone mineral density (BMD) of cortical bone in the femoral neck is closely related to the mechanical response of osteocytes, which provides a new idea for improving cortical bone quality. Perhaps cortical bone quality could be improved by stimulating osteocytes. Quadrantal differences of bone quality in the mid-femoral neck should be considered to improve fracture risk prediction in the future.
Collapse
Affiliation(s)
- Haipeng Cen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - He Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- *Correspondence: He Gong,
| | - Haibo Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Shaowei Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xiaodan Wu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Yubo Fan
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- School of Engineering Medicine, Beihang University, Beijing, China
| |
Collapse
|
16
|
Cui J, Shibata Y, Zhu T, Zhou J, Zhang J. Osteocytes in bone aging: Advances, challenges, and future perspectives. Ageing Res Rev 2022; 77:101608. [PMID: 35283289 DOI: 10.1016/j.arr.2022.101608] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 02/08/2023]
Abstract
Osteocytes play a critical role in maintaining bone homeostasis and in regulating skeletal response to hormones and mechanical loading. Substantial evidence have demonstrated that osteocytes and their lacunae exhibit morphological changes in aged bone, indicating the underlying involvement of osteocytes in bone aging. Notably, recent studies have deciphered aged osteocytes to have characteristics such as impaired mechanosensitivity, accumulated cellular senescence, dysfunctional perilacunar/canalicular remodeling, and degenerated lacuna-canalicular network. However, detailed molecular mechanisms of osteocytes remain unclear. Nonetheless, osteocyte transcriptomes analyzed via advanced RNA sequencing (RNA-seq) techniques have identified several bone aging-related genes and signaling pathways, such as Wnt, Bmp/TGF, and Jak-STAT. Moreover, inflammation, immune dysfunction, energy shortage, and impaired hormone responses possibly affect osteocytes in age-related bone deterioration. In this review, we summarize the hallmarks of aging bone and osteocytes and discuss osteocytic mechanisms in age-related bone loss and impaired bone quality. Furthermore, we provide insights into the challenges faced and their possible solutions when investigating osteocyte transcriptomes. We also highlight that single-cell RNA-seq can decode transcriptomic messages in aged osteocytes; therefore, this technique can promote novel single cell-based investigations in osteocytes once a well-established standardized protocol specific for osteocytes is developed. Interestingly, improved understanding of osteocytic mechanisms have helped identify promising targets and effective therapies for aging-related osteoporosis and fragile fractures.
Collapse
|
17
|
Rux CJ, Vahidi G, Darabi A, Cox LM, Heveran CM. Perilacunar bone tissue exhibits sub-micrometer modulus gradation which depends on the recency of osteocyte bone formation in both young adult and early-old-age female C57Bl/6 mice. Bone 2022; 157:116327. [PMID: 35026452 PMCID: PMC8858864 DOI: 10.1016/j.bone.2022.116327] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/19/2021] [Accepted: 01/07/2022] [Indexed: 11/18/2022]
Abstract
Osteocytes resorb and replace bone local to the lacunar-canalicular system (LCS). However, whether osteocyte remodeling impacts bone quality adjacent to the LCS is not understood. Further, while aging is well-established to decrease osteocyte viability and truncate LCS geometry, it is unclear if aging also decreases perilacunar bone quality. To address these questions, we employed atomic force microscopy (AFM) to generate nanoscale-resolution modulus maps for cortical femur osteocyte lacunae from young (5-month) and early-old-age (22-month) female C57Bl/6 mice. AFM-mapped lacunae were also imaged with confocal laser scanning microscopy to determine which osteocytes recently deposited bone as determined by the presence of fluorochrome labels administered 2d and 8d before euthanasia. Modulus gradation with distance from the lacunar wall was compared for labeled (i.e., bone forming) and non-labeled lacunae in both young and aged mice. All mapped lacunae showed sub-microscale modulus gradation, with peak modulus values 200-400 nm from the lacunar wall. Perilacunar modulus gradations depended on the recency of osteocyte bone formation (i.e., the presence of labels). For both ages, 2d-labeled perilacunar bone had lower peak and bulk modulus compared to non-labeled perilacunar bone. Lacunar length reduced with age, but lacunar shape and size were not strong predictors of modulus gradation. Our findings demonstrate for the first time that osteocyte perilacunar remodeling impacts bone tissue modulus, one contributor to bone quality. Given the immense scale of the LCS, differences in perilacunar modulus resulting from osteocyte remodeling activity may affect the quality of a substantial amount of bone tissue.
Collapse
Affiliation(s)
- Caleb J Rux
- Department of Mechanical & Industrial Engineering, Montana State University, United States of America; UC Berkeley-UCSF Graduate Program in Bioengineering, United States of America
| | - Ghazal Vahidi
- Department of Mechanical & Industrial Engineering, Montana State University, United States of America
| | - Amir Darabi
- Department of Mechanical & Industrial Engineering, Montana State University, United States of America
| | - Lewis M Cox
- Department of Mechanical & Industrial Engineering, Montana State University, United States of America
| | - Chelsea M Heveran
- Department of Mechanical & Industrial Engineering, Montana State University, United States of America.
| |
Collapse
|
18
|
Santos LFG, Fernandes-Breitenbach F, Silva RAS, Santos DR, Peres-Ueno MJ, Ervolino E, Chaves-Neto AH, Dornelles RCM. The action of oxytocin on the bone of senescent female rats. Life Sci 2022; 297:120484. [PMID: 35301015 DOI: 10.1016/j.lfs.2022.120484] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/07/2022] [Accepted: 03/10/2022] [Indexed: 10/18/2022]
Abstract
AIMS This study verified the action of oxytocin (OT) as a preventive measure to control bone damage during aging in female rats. MAIN METHODS Wistar rats received saline (0.15 mol/L/IP; Vehicle Group), Atosiban/AT (300 μg/Kg/IP; At Group), OT (134 μg/Kg/IP; Ot Group), or AT+OT (OT injections 5 min after AT; At+Ot Group), at 19 and 20 months of age. A functional test was performed immediately before and 30 days after the injections to analyze the animals' gait. KEY FINDINGS Animals in the At group had higher alkaline phosphatase (ALP) activity, lower cortical and trabecular thickness, fewer trabeculae, higher expression of tartrate-resistant acid phosphatase (TRAP) and lower osteocalcin (OCN), higher cortical porosity, and lower moment of inertia and bone strength at the femoral neck. OT administration increased lipidic peroxidation and plasma superoxide dismutase (SOD), and provided, in the femoral neck, lower expression of TRAP and higher OCN, greater cortical and trabecular thickness, a greater number of trabeculae, bone mineral density (BMD), higher inertia bone strength, and lower cortical porosity. At + Ot group showed great similarity with the vehicle group, higher SOD, and BMD. An increase in stride length and no increase in base width of 21-month-old animals were observed after OT, unlike animal's vehicle or AT. SIGNIFICANCE Endogenous OT plays an important role in the regulation of bone remodeling during periestropause, and exogenous OT stands out as a potential preventive intervention in this period to improve bone quality with functional repercussions, possibly providing better gait activity.
Collapse
Affiliation(s)
| | | | | | - Damáris Raíssa Santos
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas/SBFis/UNESP, Brazil
| | | | - Edilson Ervolino
- Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Antonio Hernandes Chaves-Neto
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas/SBFis/UNESP, Brazil; Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil
| | - Rita Cássia Menegati Dornelles
- Programa de Pós-Graduação Multicêntrico em Ciências Fisiológicas/SBFis/UNESP, Brazil; Department of Basic Sciences, School of Dentistry, São Paulo State University (UNESP), Araçatuba, São Paulo, Brazil.
| |
Collapse
|
19
|
Little-Letsinger SE, Rubin J, Diekman B, Rubin CT, McGrath C, Pagnotti GM, Klett EL, Styner M. Exercise to Mend Aged-tissue Crosstalk in Bone Targeting Osteoporosis & Osteoarthritis. Semin Cell Dev Biol 2022; 123:22-35. [PMID: 34489173 PMCID: PMC8840966 DOI: 10.1016/j.semcdb.2021.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 08/16/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022]
Abstract
Aging induces alterations in bone structure and strength through a multitude of processes, exacerbating common aging- related diseases like osteoporosis and osteoarthritis. Cellular hallmarks of aging are examined, as related to bone and the marrow microenvironment, and ways in which these might contribute to a variety of age-related perturbations in osteoblasts, osteocytes, marrow adipocytes, chondrocytes, osteoclasts, and their respective progenitors. Cellular senescence, stem cell exhaustion, mitochondrial dysfunction, epigenetic and intracellular communication changes are central pathways and recognized as associated and potentially causal in aging. We focus on these in musculoskeletal system and highlight knowledge gaps in the literature regarding cellular and tissue crosstalk in bone, cartilage, and the bone marrow niche. While senolytics have been utilized to target aging pathways, here we propose non-pharmacologic, exercise-based interventions as prospective "senolytics" against aging effects on the skeleton. Increased bone mass and delayed onset or progression of osteoporosis and osteoarthritis are some of the recognized benefits of regular exercise across the lifespan. Further investigation is needed to delineate how cellular indicators of aging manifest in bone and the marrow niche and how altered cellular and tissue crosstalk impact disease progression, as well as consideration of exercise as a therapeutic modality, as a means to enhance discovery of bone-targeted therapies.
Collapse
Affiliation(s)
- SE Little-Letsinger
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill
| | - J Rubin
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,North Carolina Diabetes Research Center (NCDRC), University of North Carolina at Chapel Hill,Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill
| | - B Diekman
- Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill,Joint Departments of Biomedical Engineering NC State & University of North Carolina at Chapel Hill
| | - CT Rubin
- Department of Biomedical Engineering, State University of New York at Stony Brook
| | - C McGrath
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill
| | - GM Pagnotti
- Dept of Endocrine, Neoplasia, and Hormonal Disorders, University Texas MD Anderson Cancer Center, Houston
| | - EL Klett
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,Department of Nutrition, School of Public Health, University of North Carolina at Chapel Hill
| | - M Styner
- Department of Medicine, Division of Endocrinology & Metabolism, University of North Carolina at Chapel Hill,North Carolina Diabetes Research Center (NCDRC), University of North Carolina at Chapel Hill,Department of Medicine, Thurston Arthritis Research Center (TARC), University of North Carolina at Chapel Hill
| |
Collapse
|
20
|
Josephson TO, Moore JP, Maghami E, Freeman TA, Najafi AR. Computational study of the mechanical influence of lacunae and perilacunar zones in cortical bone microcracking. J Mech Behav Biomed Mater 2021; 126:105029. [PMID: 34971951 DOI: 10.1016/j.jmbbm.2021.105029] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 11/05/2021] [Accepted: 12/02/2021] [Indexed: 02/01/2023]
Abstract
The mechanical behavior of cortical bone is influenced by microstructural components such as osteons, Haversian canals, and osteocyte lacunae that arise from biological remodeling processes. This study takes a computational approach to investigate the role of the perilacunar zones formed by the local remodeling processes of lacunar-dwelling osteocytes by utilizing phase-field finite element models based on histological imaging of human bone. The models simulated the microdamage accumulation that occurs in cortical bone under transverse compression in bone without lacunae, with lacunae, and with a perilacunar zone surrounding lacunae in order to investigate the role of these features. The results of the simulations found that while lacunae create stress concentration which initiate further damage, perilacunar regions can delay or prevent the emergence and growth of microcracks.
Collapse
Affiliation(s)
- Timothy O Josephson
- Department of Mechanical Engineering and Mechanics, Drexel University, 3141 Chestnut Street, Philadelphia, 19104, PA, USA
| | - Jason P Moore
- Department of Mechanical Engineering and Mechanics, Drexel University, 3141 Chestnut Street, Philadelphia, 19104, PA, USA
| | - Ebrahim Maghami
- Department of Mechanical Engineering and Mechanics, Drexel University, 3141 Chestnut Street, Philadelphia, 19104, PA, USA
| | - Theresa A Freeman
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College of Thomas Jefferson University, 1015 Walnut Street, Philadelphia, 19107, PA, USA
| | - Ahmad R Najafi
- Department of Mechanical Engineering and Mechanics, Drexel University, 3141 Chestnut Street, Philadelphia, 19104, PA, USA.
| |
Collapse
|
21
|
Cen H, Yao Y, Liu H, Jia S, Gong H. Multiscale mechanical responses of young and elderly human femurs: A finite element investigation. Bone 2021; 153:116125. [PMID: 34280582 DOI: 10.1016/j.bone.2021.116125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND Bone remodeling in the elderly is no longer balanced. As a result, the morphologies and mechanical properties of bone at different scales will change. These changes would affect the mechanical responses of bone, which might exacerbate the imbalance of bone remodeling and even cause age-related bone diseases. METHODS Considering those changes, multiscale finite element (FE) models of bone in the young and the elderly were developed that included macroscale (proximal femur), mesoscale (cortical bone), microscale (Haversian system) and sub-microscale (osteocyte-lacuna-canaliculus-extracellular matrix system, OLCES). The stress and strain distributions at different scales and transmissions among different scales were investigated. RESULTS The stresses of the elderly at macroscale, mesoscale and microscale were higher than those in the young by 23.7%, 62.5% and 8.0%, respectively, and the stresses of the elderly and the young at sub-microscale were almost the same. The strain of the elderly at macroscale, mesoscale, microscale and sub-microscale were higher than those in the young by 48.6%, 56.8%, 11.9% and 25.1%, respectively. The stress and strain transmission rates (ησand ηε) from mesoscale to microscale were decreased by 1.8%, and 2.5% than those from macroscale to mesoscale in the elderly, respectively; but increased by 13.8%, and 4.7% in the young, respectively. ηε from microscale to sub-microscale in the elderly was higher than that in the young by 21.3%. CONCLUSIONS Degeneration of cortical bone mechanical property in the elderly causes increases in stress and strain at macroscale and mesoscale. The reduction of lacunar number in the elderly is not conducive to the mechanical transmission from mesoscale to microscale. The differences in stress and strain at microscale between the young and the elderly are smaller than those at macroscale or mesoscale. The strain stimulus sensed by osteocyte in the elderly is not weakened compared with that in the young.
Collapse
Affiliation(s)
- Haipeng Cen
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Yan Yao
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Haibo Liu
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Shaowei Jia
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - He Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| |
Collapse
|
22
|
Hemmatian H, Conrad S, Furesi G, Mletzko K, Krug J, Faila AV, Kuhlmann JD, Rauner M, Busse B, Jähn-Rickert K. Reorganization of the osteocyte lacuno-canalicular network characteristics in tumor sites of an immunocompetent murine model of osteotropic cancers. Bone 2021; 152:116074. [PMID: 34174502 DOI: 10.1016/j.bone.2021.116074] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/04/2021] [Accepted: 06/18/2021] [Indexed: 11/16/2022]
Abstract
Mechanosensitive osteocytes are central regulators of bone resorption and formation. However, during the formation of bone metastases, which arise as consequences of breast and prostate cancer and skew homeostatic bone remodeling to favor osteolytic, osteosclerotic or mixed lesions, only a paucity of data exists on tumor-associated osteocyte interaction. Herein, we used a suite of high-resolution imaging and histological techniques to evaluate the effect of osteotropic cancer on cortical bone microarchitecture. Confocal imaging highlighted a direct contact between tumor cells residing in the bone marrow and osteocytes. High-resolution microcomputed tomography revealed a 10-12% larger osteocyte lacuna volume in the presence of tumor cells at day 21 after intratibial injection of EO771-Luc breast and RM1-Luc prostate cancer cells. The 3D representative of the spatial distribution of cortical bone microporosity showed i) a regional accumulation of vascular canals and large lacunae with low connectivity in osteosclerotic regions of interest and ii) an absence of vascular canals and large lacunae in osteolytic regions. These findings pinpoint the relationship between the presence of tumor cells in the bone marrow microenvironment and osteocyte lacunar characteristics and cortical bone blood vessel structure.
Collapse
Affiliation(s)
- Haniyeh Hemmatian
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 55a, 22529 Hamburg, Germany
| | - Stefanie Conrad
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Giulia Furesi
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Kathrin Mletzko
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 55a, 22529 Hamburg, Germany
| | - Johannes Krug
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 55a, 22529 Hamburg, Germany
| | - Antonio Virgilio Faila
- Microscopy Imaging Facility, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Dominik Kuhlmann
- Department of Gynecology and Obstetrics, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III and Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany.
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 55a, 22529 Hamburg, Germany; Interdisciplinary Competence Center for Interface Research (ICCIR), Hamburg, Germany; Forum Medical Technology Health Hamburg (FMTHH), Hamburg, Germany.
| | - Katharina Jähn-Rickert
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 55a, 22529 Hamburg, Germany; Mildred Scheel Cancer Career Center Hamburg, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg.
| |
Collapse
|
23
|
Hemmatian H, Bakker AD, Klein-Nulend J, van Lenthe GH. Alterations in osteocyte lacunar morphology affect local bone tissue strains. J Mech Behav Biomed Mater 2021; 123:104730. [PMID: 34438250 DOI: 10.1016/j.jmbbm.2021.104730] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/16/2021] [Accepted: 07/17/2021] [Indexed: 11/17/2022]
Abstract
Osteocytes are capable of remodeling their perilacunar bone matrix, which causes considerable variations in the shape and size of their lacunae. If these variations in lacunar morphology cause changes in the mechanical environment of the osteocytes, in particular local strains, they would subsequently affect bone mechanotransduction, since osteocytes are likely able to directly sense these strains. The purpose of this study is to quantify the effect of alterations in osteocyte lacunar morphology on peri-lacunar bone tissue strains. To this end, we related the actual lacunar shape in fibulae of six young-adult (5-month) and six old (23-month) mice, quantified by high-resolution micro-computed tomography, to microscopic strains, analyzed by micro-finite element modeling. We showed that peak effective strain increased by 12.6% in osteocyte cell bodies (OCYs), 9.6% in pericellular matrix (PCM), and 5.3% in extra cellular matrix (ECM) as the lacunae volume increased from 100-200 μm3 to 500-600 μm3. Lacunae with a larger deviation (>8°) in orientation from the longitudinal axis of the bone are exposed to 8% higher strains in OCYs, 6.5% in PCM, 4.2% in ECM than lacunae with a deviation in orientation below 8°. Moreover, increased lacuna sphericity from 0 to 0.5 to 0.7-1 led to 25%, 23%, and 13% decrease in maximum effective strains in OCYs, PCM, and ECM, respectively. We further showed that due to the presence of smaller and more round lacunae in old mice, local bone tissue strains are on average 5% lower in the vicinity of lacunae and their osteocytes of old mice compared to young. Understanding how changes in lacunar morphology affect the micromechanical environment of osteocytes presents a first step in unraveling their potential role in impaired bone mechanoresponsiveness with e.g. aging.
Collapse
Affiliation(s)
- Haniyeh Hemmatian
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium; Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands; Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - 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
| | - 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
| | - G Harry van Lenthe
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium.
| |
Collapse
|
24
|
Tits A, Plougonven E, Blouin S, Hartmann MA, Kaux JF, Drion P, Fernandez J, van Lenthe GH, Ruffoni D. Local anisotropy in mineralized fibrocartilage and subchondral bone beneath the tendon-bone interface. Sci Rep 2021; 11:16534. [PMID: 34400706 PMCID: PMC8367976 DOI: 10.1038/s41598-021-95917-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/02/2021] [Indexed: 12/19/2022] Open
Abstract
The enthesis allows the insertion of tendon into bone thanks to several remarkable strategies. This complex and clinically relevant location often features a thin layer of fibrocartilage sandwiched between tendon and bone to cope with a highly heterogeneous mechanical environment. The main purpose of this study was to investigate whether mineralized fibrocartilage and bone close to the enthesis show distinctive three-dimensional microstructural features, possibly to enable load transfer from tendon to bone. As a model, the Achilles tendon-calcaneus bone system of adult rats was investigated with histology, backscattered electron imaging and micro-computed tomography. The microstructural porosity of bone and mineralized fibrocartilage in different locations including enthesis fibrocartilage, periosteal fibrocartilage and bone away from the enthesis was characterized. We showed that calcaneus bone presents a dedicated protrusion of low porosity where the tendon inserts. A spatially resolved analysis of the trabecular network suggests that such protrusion may promote force flow from the tendon to the plantar ligament, while partially relieving the trabecular bone from such a task. Focusing on the tuberosity, highly specific microstructural aspects were highlighted. Firstly, the interface between mineralized and unmineralized fibrocartilage showed the highest roughness at the tuberosity, possibly to increase failure resistance of a region carrying large stresses. Secondly, fibrochondrocyte lacunae inside mineralized fibrocartilage, in analogy with osteocyte lacunae in bone, had a predominant alignment at the enthesis and a rather random organization away from it. Finally, the network of subchondral channels inside the tuberosity was highly anisotropic when compared to contiguous regions. This dual anisotropy of subchondral channels and cell lacunae at the insertion may reflect the alignment of the underlying collagen network. Our findings suggest that the microstructure of fibrocartilage may be linked with the loading environment. Future studies should characterize those microstructural aspects in aged and or diseased conditions to elucidate the poorly understood role of bone and fibrocartilage in enthesis-related pathologies.
Collapse
Affiliation(s)
- Alexandra Tits
- Mechanics of Biological and Bioinspired Materials Laboratory, Department of Aerospace and Mechanical Engineering, University of Liège, Quartier Polytech 1, Allée de la Découverte 9, 4000, Liège, Belgium
| | - Erwan Plougonven
- Chemical Engineering Department, University of Liège, Liège, Belgium
| | - Stéphane Blouin
- Ludwig Boltzmann Institute of Osteology, Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria
| | - Markus A Hartmann
- Ludwig Boltzmann Institute of Osteology, Hanusch Hospital of OEGK and AUVA Trauma Centre Meidling, 1st Medical Department Hanusch Hospital, Vienna, Austria
| | - Jean-François Kaux
- Department of Physical Medicine and Sports Traumatology, University of Liège and University Hospital of Liège, Liège, Belgium
| | - Pierre Drion
- Experimental Surgery Unit, GIGA and Credec, University of Liege, Liege, Belgium
| | - Justin Fernandez
- Auckland Bioengineering Institute and Department of Engineering Science, University of Auckland, Auckland, New Zealand
| | | | - Davide Ruffoni
- Mechanics of Biological and Bioinspired Materials Laboratory, Department of Aerospace and Mechanical Engineering, University of Liège, Quartier Polytech 1, Allée de la Découverte 9, 4000, Liège, Belgium.
| |
Collapse
|
25
|
Uniyal P, Sihota P, Tikoo K, Kumar N. Anatomical variation in intracortical canal network microarchitecture and its influence on bone fracture risk. J Mech Behav Biomed Mater 2021; 123:104770. [PMID: 34392038 DOI: 10.1016/j.jmbbm.2021.104770] [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: 02/03/2021] [Revised: 07/05/2021] [Accepted: 08/07/2021] [Indexed: 11/30/2022]
Abstract
Intracortical canals are a major contributor to cortical bone porosity and influence its mechanical response. Canal networks act as stress concentrators and the magnitude of which depends on the size and spatial distribution of canals. In the present study, we investigated site-dependent variation in intracortical canal network morphological indices and their effect on the mechanical response of bone. For this, mid-diaphysis of rat tibia bones were scanned using high-resolution micro-CT and morphological indices were measured for four main anatomical sites-anterior, posterior, medial and lateral. Further, a micro-finite element (μFE) model was developed to quantify the stress concentration regions in different cortices. The fracture risk was assessed using an effective strain approach. Results show that canal porosity, canal orientation and canal length are site-dependent whereas canal diameter and canal number density are independent of the site. The lateral cortex has significantly higher porosity compared to the posterior cortex (p < 0.05). The orientation of canals is found significantly different between endosteal and periosteal regions for anterior and medial quadrants. Canals are inclined at higher angles with bone axis in the endosteal region as compare to the periosteal region. The μ-FE results show that the regions with higher effective strain are concentrated around the canals. Further, failed element volume per unit bone volume is found highest for medial cortex whereas lowest for posterior cortex. The higher failed volume is associated with more radial canals in the medial cortex as compare to other cortices. The linear regression analysis shows that the volume of overstrained elements strongly depends on canal orientation (R2 = 0.73, p < 0.0001) and canal porosity (R2 = 0.61, p < 0.0001). The findings from this study suggest that along with vascular canal porosity, canal orientation and canal diameter can further improve the bone fracture risk assessment.
Collapse
Affiliation(s)
- Piyush Uniyal
- Department for Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Praveer Sihota
- Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, India
| | - Kulbhushan Tikoo
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Mohali, India
| | - Navin Kumar
- Department for Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, India; Department of Mechanical Engineering, Indian Institute of Technology Ropar, Rupnagar, India.
| |
Collapse
|
26
|
Jähn-Rickert K, Zimmermann EA. Potential Role of Perilacunar Remodeling in the Progression of Osteoporosis and Implications on Age-Related Decline in Fracture Resistance of Bone. Curr Osteoporos Rep 2021; 19:391-402. [PMID: 34117624 DOI: 10.1007/s11914-021-00686-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/22/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW We took an interdisciplinary view to examine the potential contribution of perilacunar/canalicular remodeling to declines in bone fracture resistance related to age or progression of osteoporosis. RECENT FINDINGS Perilacunar remodeling is most prominent as a result of lactation; recent advances further elucidate the molecular players involved and their effect on bone material properties. Of these, vitamin D and calcitonin could be active during aging or osteoporosis. Menopause-related hormonal changes or osteoporosis therapies affect bone material properties and mechanical behavior. However, investigations of lacunar size or osteocyte TRAP activity with age or osteoporosis do not provide clear evidence for or against perilacunar remodeling. While the occurrence and potential role of perilacunar remodeling in aging and osteoporosis progression are largely under-investigated, widespread changes in bone matrix composition in OVX models and following osteoporosis therapies imply osteocytic maintenance of bone matrix. Perilacunar remodeling-induced changes in bone porosity, bone matrix composition, and bone adaptation could have significant implications for bone fracture resistance.
Collapse
Affiliation(s)
- Katharina Jähn-Rickert
- Heisenberg Research Group, Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Lottestr. 55a, 22529, Hamburg, Germany.
- Mildred Scheel Cancer Career Center Hamburg, University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Elizabeth A Zimmermann
- Faculty of Dentistry, McGill University, Strathcona Anatomy and Dentistry Building, 3640 Rue University, Montreal, Canada.
| |
Collapse
|
27
|
Vahidi G, Rux C, Sherk VD, Heveran CM. Lacunar-canalicular bone remodeling: Impacts on bone quality and tools for assessment. Bone 2021; 143:115663. [PMID: 32987198 PMCID: PMC7769905 DOI: 10.1016/j.bone.2020.115663] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 01/06/2023]
Abstract
Osteocytes can resorb as well as replace bone adjacent to the expansive lacunar-canalicular system (LCS). Suppressed LCS remodeling decreases bone fracture toughness, but it is unclear how altered LCS remodeling impacts bone quality. The first goal of this review is to assess how LCS remodeling impacts LCS morphology as well as the composition and mechanical properties of surrounding bone tissue. The second goal is to compare tools available for the assessment of bone quality at length-scales that are physiologically-relevant to LCS remodeling. We find that changes to LCS morphology occur in response to a variety of physiological conditions and diseases and can be classified in two general phenotypes. In the 'aging phenotype', seen in aging and in some disuse models, the LCS is truncated and osteocytes apoptosis is increased. In the 'osteocytic osteolysis' phenotype, which is adaptive in some physiological settings and possibly maladaptive in others, the LCS enlarges and osteocytes generally maintain viability. Bone composition and mechanical properties vary near the osteocyte and change with at least some conditions that alter LCS morphology. However, few studies have evaluated bone composition and mechanical properties close to the LCS and so the impacts of LCS remodeling phenotypes on bone tissue quality are still undetermined. We summarize the current understanding of how LCS remodeling impacts LCS morphology, tissue-scale bone composition and mechanical properties, and whole-bone material properties. Tools are compared for assessing tissue-scale bone properties, as well as the resolution, advantages, and limitations of these techniques.
Collapse
Affiliation(s)
- G Vahidi
- Department of Mechanical & Industrial Engineering, Montana State University, United States of America
| | - C Rux
- Department of Mechanical & Industrial Engineering, Montana State University, United States of America
| | - V D Sherk
- Department of Orthopedics, University of Colorado Anschutz School of Medicine, United States of America
| | - C M Heveran
- Department of Mechanical & Industrial Engineering, Montana State University, United States of America.
| |
Collapse
|
28
|
The mechanoresponse of bone is closely related to the osteocyte lacunocanalicular network architecture. Proc Natl Acad Sci U S A 2020; 117:32251-32259. [PMID: 33288694 PMCID: PMC7768754 DOI: 10.1073/pnas.2011504117] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The explanation of how bone senses and adapts to mechanical stimulation still relies on hypotheses. The fluid flow hypothesis claims that a load-induced fluid flow through the lacunocanalicular network can be sensed by osteocytes, which reside within the network structure. We show that considering the network architecture results in a better prediction of bone remodeling than mechanical strain alone. This was done by calculating the fluid flow through the lacunocanalicular network in bone volumes covering the complete cross-sections of mouse tibiae, which underwent controlled in vivo loading. The established relationship between mechanosensitivity and network architecture in individual animals implies possibilities for patient-specific therapies. A new connectomics approach to analyze lacunocanalicular network properties is necessary to understand skeletal mechanobiology. Organisms rely on mechanosensing mechanisms to adapt to changes in their mechanical environment. Fluid-filled network structures not only ensure efficient transport but can also be employed for mechanosensation. The lacunocanalicular network (LCN) is a fluid-filled network structure, which pervades our bones and accommodates a cell network of osteocytes. For the mechanism of mechanosensation, it was hypothesized that load-induced fluid flow results in forces that can be sensed by the cells. We use a controlled in vivo loading experiment on murine tibiae to test this hypothesis, whereby the mechanoresponse was quantified experimentally by in vivo micro-computed tomography (µCT) in terms of formed and resorbed bone volume. By imaging the LCN using confocal microscopy in bone volumes covering the entire cross-section of mouse tibiae and by calculating the fluid flow in the three-dimensional (3D) network, we could perform a direct comparison between predictions based on fluid flow velocity and the experimentally measured mechanoresponse. While local strain distributions estimated by finite-element analysis incorrectly predicts preferred bone formation on the periosteal surface, we demonstrate that additional consideration of the LCN architecture not only corrects this erroneous bias in the prediction but also explains observed differences in the mechanosensitivity between the three investigated mice. We also identified the presence of vascular channels as an important mechanism to locally reduce fluid flow. Flow velocities increased for a convergent network structure where all of the flow is channeled into fewer canaliculi. We conclude that, besides mechanical loading, LCN architecture should be considered as a key determinant of bone adaptation.
Collapse
|
29
|
Schemenz V, Gjardy A, Chamasemani FF, Roschger A, Roschger P, Zaslansky P, Helfen L, Burghammer M, Fratzl P, Weinkamer R, Brunner R, Willie BM, Wagermaier W. Heterogeneity of the osteocyte lacuno-canalicular network architecture and material characteristics across different tissue types in healing bone. J Struct Biol 2020; 212:107616. [PMID: 32920138 DOI: 10.1016/j.jsb.2020.107616] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 02/08/2023]
Abstract
Various tissue types, including fibrous connective tissue, bone marrow, cartilage, woven and lamellar bone, coexist in healing bone. Similar to most bone tissue type, healing bone contains a lacuno-canalicular network (LCN) housing osteocytes. These cells are known to orchestrate bone remodeling in healthy bone by sensing mechanical strains and translating them into biochemical signals. The structure of the LCN is hypothesized to influence mineralization processes. Hence, the aim of the present study was to visualize and match spatial variations in the LCN topology with mineral characteristics, within and at the interfaces of the different tissue types that comprise healing bone. We applied a correlative multi-method approach to visualize the LCN architecture and quantify mineral particle size and orientation within healing femoral bone in a mouse osteotomy model (26 weeks old C57BL/6 mice). This approach revealed structural differences across several length scales during endochondral ossification within the following regions: calcified cartilage, bony callus, cortical bone and a transition zone between the cortical and callus region analyzed 21 days after the osteotomy. In this transition zone, we observed a continuous convergence of mineral characteristics and osteocyte lacunae shape as well as discontinuities in the lacunae volume and LCN connectivity. The bony callus exhibits a 34% higher lacunae number density and 40% larger lacunar volume compared to cortical bone. The presented correlations between LCN architecture and mineral characteristics improves our understanding of how bone develops during healing and may indicate a contribution of osteocytes to bone (re)modeling.
Collapse
Affiliation(s)
- Victoria Schemenz
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - André Gjardy
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Am Mühlenberg 1, 14476 Potsdam, Germany
| | | | - Andreas Roschger
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Am Mühlenberg 1, 14476 Potsdam, Germany; Paris-Lodron-University of Salzburg, Department of Chemistry and Physics of Materials, Salzburg, Austria
| | - Paul Roschger
- Ludwig Boltzmann Institute of Osteology at Hanusch Hospital of ÖGK and AUVA Trauma Centre Meidling, 1st Medical Department, Hanusch Hospital, Vienna, Austria
| | - Paul Zaslansky
- Department for Restorative and Preventive Dentistry, Charité-Universitaetsmedizin Berlin, Berlin 14197, Germany
| | - Lukas Helfen
- Institute for Photon Science and Synchrotron Radiation, Karlsruhe Institute of Technology, D-76021 Karlsruhe, Germany; Institut Laue-Langevin, CS 20156, 38042 Grenoble Cedex 9, France
| | | | - Peter Fratzl
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Richard Weinkamer
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Am Mühlenberg 1, 14476 Potsdam, Germany
| | - Roland Brunner
- Materials Center Leoben Forschung GmbH, 8700 Leoben, Austria
| | - Bettina M Willie
- Research Centre, Shriners Hospitals for Children-Canada, Department of Pediatric Surgery, McGill University, 1003 Decarie Blvd, Montreal, Quebec H4A 0A9, Canada
| | - Wolfgang Wagermaier
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Am Mühlenberg 1, 14476 Potsdam, Germany.
| |
Collapse
|
30
|
Dallas SL, Moore DS. Using confocal imaging approaches to understand the structure and function of osteocytes and the lacunocanalicular network. Bone 2020; 138:115463. [PMID: 32512167 PMCID: PMC7423610 DOI: 10.1016/j.bone.2020.115463] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 05/29/2020] [Indexed: 02/07/2023]
Abstract
Although overlooked in the past, osteocytes have come to the forefront of skeletal biology and are now recognized as a key cell type that integrates hormonal, mechanical and other signals to control bone mass through regulation of both osteoblast and osteoclast activity. With the surge of recent interest in osteocytes as bone regulatory cells and the discovery that they also function as endocrine regulators of phosphate homeostasis, there has been renewed interest in understanding the structure and function of these unique and relatively inaccessible cells. Osteocytes are embedded within the mineralized bone matrix and are housed within a complex lacunocanalicular system which connects them with the circulation and with other organ systems. This has presented unique challenges for imaging these cells. This review summarizes recent advances in confocal imaging approaches for visualizing osteocytes and their lacunocanalicular networks in both living and fixed bone specimens and discusses how computational approaches can be combined with live and fixed cell imaging techniques to generate quantitative outputs and predictive models. The integration of advanced imaging with computational approaches promises to lead to a more in depth understanding of the structure and function of osteocyte networks and the lacunocanalicular system in the healthy and aging state as well as in pathological conditions in bone.
Collapse
Affiliation(s)
- Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri Kansas City, Kansas City, MO 64108, United States of America.
| | - David S Moore
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri Kansas City, Kansas City, MO 64108, United States of America
| |
Collapse
|
31
|
Oxytocin and bone quality in the femoral neck of rats in periestropause. Sci Rep 2020; 10:7937. [PMID: 32404873 PMCID: PMC7220952 DOI: 10.1038/s41598-020-64683-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 04/17/2020] [Indexed: 11/08/2022] Open
Abstract
The objective of this study is to identify whether oxytocin (OT) contributes to the reduction of osteopenia in the femoral neck of rats in periestropause. Animals in irregular estrous cycles received two NaCl injections (0.15 mol/L) or OT (134 μg/kg) over a 12-h interval, and after thirty-five days without treatments, the biological sample collection was performed. The oxytocin group (Ot) demonstrated the highest enzymatic activity of alkaline phosphatase (p = 0.0138), lowest enzymatic activity of tartrate-resistant acid phosphatase (p = 0.0045), higher percentage of compact bone (p = 0.0359), cortical expression of runt-related transcription factor 2 (p = 0.0101), osterix (p = 0.0101), bone morphogenetic protein-2/4 (p = 0.0101) and periostin (p = 0.0455). Furthermore, the mineral-to-matrix ratio (ν1PO4/Proline) was higher and type-B carbonate substitution (CO3/ν1PO4) was lower (p = 0.0008 and 0.0303) in Ot group. The Ot showed higher areal bone mineral density (p = 0.0050), cortical bone area (p = 0.0416), polar moment of inertia, maximum, minimum (p = 0.0480, 0.0480, 0.0035), bone volume fraction (p = 0.0166), connectivity density (p < 0.0001), maximal load (p = 0.0003) and bone stiffness (p = 0.0145). In Ot percentage of cortical pores (p = 0.0102) and trabecular number (p = 0.0088) was lower. The results evidence action of OT in the reduction of osteopenia, suggesting that it is a promising anabolic strategy for the prevention of primary osteoporosis during the periestropause period.
Collapse
|
32
|
Kegelman CD, Coulombe JC, Jordan KM, Horan DJ, Qin L, Robling AG, Ferguson VL, Bellido TM, Boerckel JD. YAP and TAZ Mediate Osteocyte Perilacunar/Canalicular Remodeling. J Bone Miner Res 2020; 35:196-210. [PMID: 31610061 PMCID: PMC7066596 DOI: 10.1002/jbmr.3876] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 08/13/2019] [Accepted: 09/07/2019] [Indexed: 12/31/2022]
Abstract
Bone fragility fractures are caused by low bone mass or impaired bone quality. Osteoblast/osteoclast coordination determines bone mass, but the factors that control bone quality are poorly understood. Osteocytes regulate osteoblast and osteoclast activity on bone surfaces but can also directly reorganize the bone matrix to improve bone quality through perilacunar/canalicular remodeling; however, the molecular mechanisms remain unclear. We previously found that deleting the transcriptional regulators Yes-associated protein (YAP) and transcriptional co-activator with PDZ-motif (TAZ) from osteoblast-lineage cells caused lethality in mice due to skeletal fragility. Here, we tested the hypothesis that YAP and TAZ regulate osteocyte-mediated bone remodeling by conditional ablation of both YAP and TAZ from mouse osteocytes using 8 kb-DMP1-Cre. Osteocyte-conditional YAP/TAZ deletion reduced bone mass and dysregulated matrix collagen content and organization, which together decreased bone mechanical properties. Further, YAP/TAZ deletion impaired osteocyte perilacunar/canalicular remodeling by reducing canalicular network density, length, and branching, as well as perilacunar flourochrome-labeled mineral deposition. Consistent with recent studies identifying TGF-β as a key inducer of osteocyte expression of matrix-remodeling enzymes, YAP/TAZ deletion in vivo decreased osteocyte expression of matrix proteases MMP13, MMP14, and CTSK. In vitro, pharmacologic inhibition of YAP/TAZ transcriptional activity in osteocyte-like cells abrogated TGF-β-induced matrix protease gene expression. Together, these data show that YAP and TAZ control bone matrix accrual, organization, and mechanical properties by regulating osteocyte-mediated bone remodeling. Elucidating the signaling pathways that control perilacunar/canalicular remodeling may enable future therapeutic targeting of bone quality to reverse skeletal fragility. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Christopher D Kegelman
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer C Coulombe
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Kelsey M Jordan
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel J Horan
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ling Qin
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander G Robling
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Virginia L Ferguson
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, USA
| | - Teresita M Bellido
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA.,Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Effects of ionizing radiation on woven bone: influence on the osteocyte lacunar network, collagen maturation, and microarchitecture. Clin Oral Investig 2019; 24:2763-2771. [PMID: 31732880 DOI: 10.1007/s00784-019-03138-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/24/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Evaluate the effects of ionizing radiation on microarchitecture, the osteocyte lacunar network, and collagen maturity in a bone repair site. MATERIALS AND METHODS Bone defects were created on tibias of 20 New Zealand rabbits. After 2 weeks, the animals were randomly divided into (n = 10) NoIr (nonirradiated group) and Ir (irradiated group). In the Ir, the animals received single-dose irradiation of 30 Gy on the tibia and were euthanized after 2 weeks. Bone microarchitecture parameters were analyzed by using micro-CT, and the osteocyte lacunar network, bone matrix, and collagen maturation by histomorphometric analysis. The data were analyzed using unpaired Student's t test (α = 0.05). RESULTS Trabecular thickness in Ir was lower than that in NoIr (P = 0.028). No difference was found for bone volume fraction and bone area. Lacunae filled with osteocytes were more numerous (P < 0.0001) in NoIr (2.6 ± 0.6) than in Ir (1.97 ± 0.53). Empty lacunae were more prevalent (P < 0.003) in Ir (0.14 ± 0.10) than in NoIr (0.1 ± 0.1). The mean osteocyte lacunae size was higher (P < 0.01) in Ir (15.4 ± 4.41) than in NoIr (12.7 ± 3.7). Picrosirius red analysis showed more (P < 0.05) mature collagen in NoIr (29.0 ± 5.3) than in Ir (23.4 ± 4.5). Immature collagen quantification revealed no difference between groups. CONCLUSIONS Ionizing radiation compromised bone formation and an impairment in bone repair in irradiated woven bone was observed. CLINICAL RELEVANCE Before radiotherapy, patients usually need surgical intervention, which may be better performed, if clinicians understand the repair process in irradiated bone, using novel approaches for treating these individuals.
Collapse
|
34
|
Heveran CM, Schurman CA, Acevedo C, Livingston EW, Howe D, Schaible EG, Hunt HB, Rauff A, Donnelly E, Carpenter RD, Levi M, Lau AG, Bateman TA, Alliston T, King KB, Ferguson VL. Chronic kidney disease and aging differentially diminish bone material and microarchitecture in C57Bl/6 mice. Bone 2019; 127:91-103. [PMID: 31055118 PMCID: PMC6760860 DOI: 10.1016/j.bone.2019.04.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 03/15/2019] [Accepted: 04/26/2019] [Indexed: 12/31/2022]
Abstract
Chronic kidney disease (CKD) is a common disease of aging and increases fracture risk over advanced age alone. Aging and CKD differently impair bone turnover and mineralization. We thus hypothesize that the loss of bone quality would be greatest with the combination of advanced age and CKD. We evaluated bone from young adult (6 mo.), middle-age (18 mo.), and old (24 mo.) male C57Bl/6 mice three months following either 5/6th nephrectomy, to induce CKD, or Sham procedures. CKD exacerbated losses of cortical and trabecular microarchitecture associated with aging. Aging and CKD each resulted in thinner, more porous cortices and fewer and thinner trabeculae. Bone material quality was also reduced with CKD, and these changes to bone material were distinct from those due to age. Aging reduced whole-bone flexural strength and modulus, micrometer-scale nanoindentation modulus, and nanometer-scale tissue and collagen strain (small-angle x-ray scattering [SAXS]. By contrast, CKD reduced work to fracture and variation in bone tissue modulus and composition (Raman spectroscopy), and increased percent collagen strain. The increased collagen strain burden was associated with loss of toughness in CKD. In addition, osteocyte lacunae became smaller, sparser, and more disordered with age for Sham mice, yet these age-related changes were not clearly observed in CKD. However, for CKD, larger lacunae positively correlated with increased serum phosphate levels, suggesting that osteocytes play a role in systemic mineral homeostasis. This work demonstrates that CKD reduces bone quality, including microarchitecture and bone material properties, and that loss of bone quality with age is compounded by CKD. These findings may help reconcile why bone mass does not consistently predict fracture in the CKD population, as well as why older individuals with CKD are at high risk of fragility.
Collapse
Affiliation(s)
- Chelsea M Heveran
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, United States of America
| | - Charles A Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, United States of America
| | - Claire Acevedo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, UT, United States of America
| | - Eric W Livingston
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, United States of America
| | - Danielle Howe
- Department of Biomedical Engineering, The College of New Jersey, Ewing, NJ, United States of America
| | - Eric G Schaible
- Advanced Light Source, Lawrence Berkeley National Laboratory, Berkeley, CA, United States of America
| | - Heather B Hunt
- Department of Materials Science & Engineering, Cornell University, Ithaca, NY, United States of America
| | - Adam Rauff
- Department of Bioengineering, University of Colorado, Denver, CO, United States of America
| | - Eve Donnelly
- Department of Materials Science & Engineering, Cornell University, Ithaca, NY, United States of America
| | - R Dana Carpenter
- Department of Mechanical Engineering, University of Colorado, Denver, CO, United States of America
| | - Moshe Levi
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington D.C., United States of America
| | - Anthony G Lau
- Department of Biomedical Engineering, The College of New Jersey, Ewing, NJ, United States of America
| | - Ted A Bateman
- Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, United States of America
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, United States of America
| | - Karen B King
- Department of Orthopaedics, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Virginia L Ferguson
- Department of Mechanical Engineering, University of Colorado, Boulder, CO, United States of America.
| |
Collapse
|
35
|
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: 1.3] [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
|
36
|
Wittig NK, Laugesen M, Birkbak ME, Bach-Gansmo FL, Pacureanu A, Bruns S, Wendelboe MH, Brüel A, Sørensen HO, Thomsen JS, Birkedal H. Canalicular Junctions in the Osteocyte Lacuno-Canalicular Network of Cortical Bone. ACS NANO 2019; 13:6421-6430. [PMID: 31095362 DOI: 10.1021/acsnano.8b08478] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The osteocyte lacuno-canalicular network (LCN) is essential for bone remodeling because osteocytes regulate cell recruitment. This has been proposed to occur through liquid-flow-induced shear forces in the canaliculi. Models of the LCN have thus far assumed that it contains canaliculi connecting the osteocyte lacunae. However, here, we reveal that enlarged spaces occur at places where several canaliculi cross; we name these spaces canalicular junctions. We characterize them in detail within mice cortical bone using synchrotron nanotomography at two length scales, with 50 and 130 nm voxel size, and show that canalicular junctions occur at a density similar to that of osteocyte lacunae and that canalicular junctions tend to cluster. Through confocal laser scanning microscopy, we show that canalicular junctions are widespread as we have observed them in cortical bone from several species, even though the number density of the canalicular junctions was not universal. Fluid flow simulations of a simple model system with and without a canalicular junction clearly show that liquid mass transport and flow velocities are altered by the presence of canalicular junctions. We suggest that these canalicular junctions may play an important role in osteocyte communication and possibly also in canalicular fluid flow. Therefore, we believe that they constitute an important component in the bone osteocyte network.
Collapse
Affiliation(s)
| | | | | | | | | | - Stefan Bruns
- Department of Chemistry, University of Copenhagen , 2100 Copenhagen Ø , Denmark
| | | | | | | | | | | |
Collapse
|
37
|
Tiede-Lewis LM, Dallas SL. Changes in the osteocyte lacunocanalicular network with aging. Bone 2019; 122:101-113. [PMID: 30743014 PMCID: PMC6638547 DOI: 10.1016/j.bone.2019.01.025] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 01/28/2019] [Indexed: 12/22/2022]
Abstract
Osteoporosis is an aging-related disease of reduced bone mass that is particularly prevalent in post-menopausal women, but also affects the aged male population and is associated with increased fracture risk. Osteoporosis is the result of an imbalance whereby bone formation by osteoblasts no longer keeps pace with resorption of bone by osteoclasts. Osteocytes are the most abundant cells in bone and, although previously thought to be quiescent, they are now known to be active, multifunctional cells that play a key role in the maintenance of bone mass by regulating both osteoblast and osteoclast activity. They are also thought to regulate bone mass through their role as mechanoresponsive cells in bone that coordinate adaptive responses to mechanical loading. Osteocytes form an extensive interconnected network throughout the mineralized bone matrix and receive their nutrients as well as hormones and signaling factors through the lacunocanalicular system. Several studies have shown that the extent and connectivity of the lacunocanalicular system and osteocyte networks degenerates in aged humans as well as in animal models of aging. It is also known that the bone anabolic response to loading is decreased with aging. This review summarizes recent research on the degenerative changes that occur in osteocytes and their lacunocanalicular system as a result of aging and discusses the implications for skeletal health and homeostasis as well as potential mechanisms that may underlie these degenerative changes. Since osteocytes are such key regulators of skeletal homeostasis, maintaining the health of the osteocyte network would seem critical for maintenance of bone health. Therefore, a more complete understanding of the structure and function of the osteocyte network, its lacunocanalicular system, and the degenerative changes that occur with aging should lead to advances in our understanding of age related bone loss and potentially lead to improved therapies.
Collapse
Affiliation(s)
- LeAnn M Tiede-Lewis
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO 64108, United States of America
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO 64108, United States of America.
| |
Collapse
|
38
|
Gorustovich AA, Nielsen FH. Effects of Nutritional Deficiency of Boron on the Bones of the Appendicular Skeleton of Mice. Biol Trace Elem Res 2019; 188:221-229. [PMID: 30182352 DOI: 10.1007/s12011-018-1499-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 08/28/2018] [Indexed: 01/20/2023]
Abstract
Scientific evidence has shown the nutritional importance of boron (B) in the remodeling and repair of cancellous bone tissue. However, the effects of the nutritional deficiency of B on the cortical bone tissue of the appendicular skeleton have not yet been described. Thus, a study was performed to histomorphometrically evaluate the density of osteocyte lacunae of cortical bone of mouse femora under conditions of nutritional deficiency of B and to analyze the effects of the deficiency on the biomechanical properties of mouse tibiae. Weaning, 21-day-old male Swiss mice were assigned to the following two groups: controls (B+; n = 10) and experimental (B-; n = 10). Control mice were fed a basal diet containing 3 mg B/kg, whereas experimental mice were fed a B-deficient diet containing 0.07 mg B/kg for 9 weeks. The histological and histomorphometric evaluations of the mice fed a B-deficient diet showed a decrease in the density of osteocyte lacunae in the femoral cortical bone tissue and the evaluation of biomechanical properties showed lower bone rigidity in the tibia.
Collapse
Affiliation(s)
- Alejandro A Gorustovich
- Interdisciplinary Materials Group-IESIING-UCASAL, INTECIN UBA-CONICET, A4400EDD, Salta, Argentina.
| | | |
Collapse
|
39
|
Hemmatian H, Jalali R, Semeins CM, Hogervorst JMA, van Lenthe GH, Klein-Nulend J, Bakker AD. Mechanical Loading Differentially Affects Osteocytes in Fibulae from Lactating Mice Compared to Osteocytes in Virgin Mice: Possible Role for Lacuna Size. Calcif Tissue Int 2018; 103:675-685. [PMID: 30109376 PMCID: PMC6208961 DOI: 10.1007/s00223-018-0463-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/30/2018] [Indexed: 12/31/2022]
Abstract
Hormonal changes during lactation are associated with profound changes in bone cell biology, such as osteocytic osteolysis, resulting in larger lacunae. Larger lacuna shape theoretically enhances the transmission of mechanical signals to osteocytes. We aimed to provide experimental evidence supporting this theory by comparing the mechanoresponse of osteocytes in the bone of lactating mice, which have enlarged lacunae due to osteocytic osteolysis, with the response of osteocytes in bone from age-matched virgin mice. The osteocyte mechanoresponse was measured in excised fibulae that were cultured in hormone-free medium for 24 h and cyclically loaded for 10 min (sinusoidal compressive load, 3000 µε, 5 Hz) by quantifying loading-related changes in Sost mRNA expression (qPCR) and sclerostin and β-catenin protein expression (immunohistochemistry). Loading decreased Sost expression by ~ threefold in fibulae of lactating mice. The loading-induced decrease in sclerostin protein expression by osteocytes was larger in lactating mice (55% decrease ± 14 (± SD), n = 8) than virgin mice (33% decrease ± 15, n = 7). Mechanical loading upregulated β-catenin expression in osteocytes in lactating mice by 3.5-fold (± 0.2, n = 6) which is significantly (p < 0.01) higher than the 1.6-fold increase in β-catenin expression by osteocytes in fibulae from virgin mice (± 0.12, n = 4). These results suggest that osteocytes in fibulae from lactating mice with large lacunae may respond stronger to mechanical loading than those from virgin mice. This could indicate that osteocytes residing in larger lacuna show a stronger response to mechanical loading.
Collapse
Affiliation(s)
- Haniyeh Hemmatian
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| | - Rozita Jalali
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| | - Cornelis M Semeins
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| | - Jolanda M A Hogervorst
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| | - G Harry van Lenthe
- Biomechanics Section, Department of Mechanical Engineering, KU Leuven, Leuven, Belgium
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Gustav Mahlerlaan 3004, 1081 LA, 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, Gustav Mahlerlaan 3004, 1081 LA, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Milovanovic P, Stojanovic M, Antonijevic D, Cirovic A, Radenkovic M, Djuric M. "Dangerous duo": Chronic nicotine exposure intensifies diabetes mellitus-related deterioration in bone microstructure - An experimental study in rats. Life Sci 2018; 212:102-108. [PMID: 30266406 DOI: 10.1016/j.lfs.2018.09.044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/03/2018] [Accepted: 09/24/2018] [Indexed: 01/19/2023]
Abstract
AIMS Bony complications of diabetes mellitus (DM) are still insufficiently understood. Our aims were to analyze the individual and combined effects of chronic hyperglycemia and nicotine exposure on the femoral trabecular and cortical microarchitecture on a rat experimental model. MAIN METHODS The micro-computed tomography based bone microstructural evaluation was performed on male Wistar rats divided into four groups: control (n = 7), experimentally-induced DM (n = 8), chronically exposed to nicotine (n = 9) and the DM group exposed chronically to nicotine (n = 9). KEY FINDINGS Chronic hyperglycemia caused mild trabecular deterioration; yet, the combination of hyperglycemia and nicotine exposure showed more deleterious effects on the trabecular bone. Namely, the DM + nicotine group had significantly lower bone volume fraction, fewer and more rod-like shaped trabeculae, along with higher trabecular separation and lower connectivity than the control group (p < 0.05). Nicotine alone did not show any significant deterioration compared to the control group. DM and DM + nicotine groups had lower cortical porosity than control and nicotine groups (p < 0.05). Cortical thickness did not show any significant intergroup differences, whereas bone perimeter and the mean polar moment of inertia were reduced in DM + nicotine group. SIGNIFICANCE Mild effects of chronic hyperglycemia on bone structure were accentuated by the chronic nicotine exposure, although nicotine alone did not cause any significant bone changes. That suggests a synergistic effect of hyperglycemia and nicotine on bone deterioration and increased propensity to fracture. Indeed, better understanding of risk factors driving bone structural deterioration is a precondition to limit the complications associated with DM.
Collapse
Affiliation(s)
- Petar Milovanovic
- Laboratory for Anthropology and Skeletal Biology, Institute of Anatomy, Faculty of Medicine, University of Belgrade, Dr Subotica 4/2, 11000 Belgrade, Serbia
| | - Marko Stojanovic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Dr Subotica 1, 11000 Belgrade, Serbia
| | - Djordje Antonijevic
- Laboratory for Anthropology and Skeletal Biology, Institute of Anatomy, Faculty of Medicine, University of Belgrade, Dr Subotica 4/2, 11000 Belgrade, Serbia
| | - Aleksandar Cirovic
- Laboratory for Anthropology and Skeletal Biology, Institute of Anatomy, Faculty of Medicine, University of Belgrade, Dr Subotica 4/2, 11000 Belgrade, Serbia
| | - Miroslav Radenkovic
- Department of Pharmacology, Clinical Pharmacology and Toxicology, Faculty of Medicine, University of Belgrade, Dr Subotica 1, 11000 Belgrade, Serbia
| | - Marija Djuric
- Laboratory for Anthropology and Skeletal Biology, Institute of Anatomy, Faculty of Medicine, University of Belgrade, Dr Subotica 4/2, 11000 Belgrade, Serbia.
| |
Collapse
|