1
|
Wasi M, Wang S, Guerra RM, Chu T, Kooker R, Seaman K, Song XS, Sassi A, Li X, Xiong J, You L, Wang L. Different effects of moderate tibial loading and Yoda1 on breast cancer-induced osteolysis in aged mice. Bone 2025; 197:117517. [PMID: 40345567 DOI: 10.1016/j.bone.2025.117517] [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: 09/18/2024] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/11/2025]
Abstract
Elderly breast cancer patients and survivors are at high risk of bone loss but experience obstacles to harness the known benefits of exercise due to aging, cancer, and cancer treatment. Previously, we and others showed that moderate mechanical loading suppressed breast cancer-induced osteolysis in young adult mice. To overcome the mechano-transduction deficits in aged skeletons, we recently tested a dual therapy combining mechanical and Yoda1 activation of mechanosensitive Piezo1 channels. We found that the dual therapy was more effective in mitigating bone loss due to aging and doxorubicin in mature mice than the individual interventions. In the present study, we further tested the hypothesis that dual therapy combining moderate tibial loading and Yoda1 protects aged skeleton from breast cancer-induced osteolysis better than individual treatments. Aged female C57BL/6 J mice (∼74-week-old) receiving Py8119 breast cancer cells in both tibiae were assigned to the four experimental groups (n = 5-8 per group) to examine the effects of 4-week Yoda1 (dose 5 mg/kg, 5 times/week) and moderate tibial loading (4.5 N peak load, 4 Hz, 300 cycles per day, 5 days/week), individually or combined on bone structural integrity. At the end of 4 weeks' experiments, the dual therapy group had the lowest incidence of osteolytic perforation (56 %) compared to the non-treated group (80 %), loading only group (70 %), and Yoda1 only group (100 %). The relative drop of cortical polar moment of inertia (Ct.pMOI), calculated as [(Week 4- Week 0)/Week 0, %], were analyzed at the proximal end, mid-diaphysis, and tibial-fibular junction of the tibia. The average values over the three locations were - 12.7 %, -3.2 %, -24.0 %, -4.2 % for the non-treated, loading only, Yoda1 only, and dual therapy groups, respectively. Furthermore, the % of samples with decreased Ct.pMOI (indication of structural deterioration) was suppressed in the dual therapy group (33 %), compared with nontreated (100 %), loading only (80 %), and Yoda1 only (100 %) groups. Each treatment differentially affected the osteoclast activity, tumor proliferation, and apoptosis of osteocytes, marrow cells and tumor cells, revealing the complex interactions of bone, tumor, and mechanical stimulations. In summary, the dual therapy resulted in skeletal benefits comparable to or slightly better than loading only treatment. However, the exacerbated bone loss and cortical perforation associated with Yoda1 call for further investigation on safe and effective treatments of skeletal damages caused by metastatic breast cancers.
Collapse
Affiliation(s)
- Murtaza Wasi
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| | - Shubo Wang
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| | - Rosa M Guerra
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| | - Tiankuo Chu
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| | - Rory Kooker
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| | - Kimberly Seaman
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Xin Suzie Song
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Amel Sassi
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Xuehua Li
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA; Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA.
| |
Collapse
|
2
|
Jeon HH, Salas MCC, Park K, Fisher L, Ha S, Palmer C, Chan F, Graves DT. Comparison of the bone remodeling in the midpalatal suture during maxillary expansion between young and middle-aged mice. Bone 2025; 197:117512. [PMID: 40324615 DOI: 10.1016/j.bone.2025.117512] [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: 05/18/2024] [Revised: 04/17/2025] [Accepted: 05/02/2025] [Indexed: 05/07/2025]
Abstract
Maxillary expansion is a common orthodontic procedure for treating maxillary transverse deficiency. However, the cell responses to mechanical force may vary across different age groups, suggesting the need for age-specific treatment protocols. To compare the age-related responses to the mechanical force, we examined the 6-week- and 12-month-old mice undergoing maxillary expansion with 0.012-in. stainless steel orthodontic wire bonded to the maxillary first and second molars (25 g force). Mice were euthanized on days 0, 3, 7, and 14 for analysis. MicroCT analysis, tartrate-resistant acid phosphatase (TRAP) stain, and immunofluorescence/immunohistochemistry stain using antibodies to RUNX2, alkaline phosphatase (ALP), Gli1 and Ki67 along with the TUNEL assay, were conducted to evaluate suture width, osteoclast activity, new bone formation and mesenchymal stem cell (MSC) proliferation and apoptosis. Both 6-week- and 12-month-old mice exhibited successful midpalatal suture opening, but young mice demonstrated earlier and more intense osteoclast activity, along with higher expression of RUNX2 and ALP. Young mice also exhibited a higher percentage of Gli1+Ki67+ immunopositive cells, while middle-aged mice showed a higher percentage of Gli1+TUNEL+ positive cells on day 3 after maxillary expansion. Our findings suggest that aging negatively impacts mechanical force-induced bone remodeling by reducing osteoclastogenesis, osteogenesis, and MSC proliferation while increasing MSC apoptosis.
Collapse
Affiliation(s)
- Hyeran Helen Jeon
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mary Cruz Contreras Salas
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kyungjoon Park
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lindsay Fisher
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sara Ha
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Caroline Palmer
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Fionna Chan
- School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
3
|
Vallejo JA, Gray M, Klump J, Wacker A, Dallas M, Johnson ML, Wacker MJ. Bone mechanical loading reduces heart rate and increases heart rate variability in mice. Bone Rep 2025; 25:101844. [PMID: 40322617 PMCID: PMC12049822 DOI: 10.1016/j.bonr.2025.101844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Cardiovascular disease and osteoporosis are clinically associated. Bone adapts to mechanical forces by altering its overall structure and mass. In response to mechanical strain bone cells release signaling molecules and activate the nervous system. Bone also exhibits endocrine functions that modulate a number of tissues including the heart. We hypothesized that bone mechanical loading acutely alters cardiac function via neural and/or endocrine mechanisms. To test this hypothesis, we performed in vivo tibia mechanical loading in anesthetized mice while monitoring heart parameters using electrocardiogram (ECG). An immediate, transient reduction in resting heart rate was observed during tibial loading in both adult male and female mice (p < 0.01) with concurrent increases in heart rate variability (HRV) (p < 0.01). ECG intervals, PR, QRS and QTc were unaffected with loading. In further studies, we found that at least 3 N of load was necessary to elicit this heart response in adult mice. With aging to 11-12 months the responsiveness of the heart to loading was blunted, suggesting this bone-heart connection may weaken with age. Administration of lidocaine around the tibia significantly diminished the heart rate response to bone loading (p < 0.05). Moreover, pre-treatment with sympathetic antagonist propranolol inhibited this heart rate response to loading (p < 0.05), while parasympathetic antagonist atropine did not (p > 0.05). This suggests that a neuronal afferent pathway in the hindlimb and reduction in efferent sympathetic tone mediate this bone-neuro-heart reflex. In conclusion, the findings that tibia bone loading age-dependently modulates heart function support the concept of physiological coupling of the skeletal and cardiovascular systems.
Collapse
Affiliation(s)
- Julian A. Vallejo
- University of Missouri – Kansas City, School of Medicine, Department of Biomedical Sciences, USA
- University of Missouri – Kansas City, School of Dentistry, Department of Oral & Craniofacial Sciences, USA
| | - Mark Gray
- University of Missouri – Kansas City, School of Medicine, Department of Biomedical Sciences, USA
| | - Jackson Klump
- University of Missouri – Kansas City, School of Medicine, Department of Biomedical Sciences, USA
| | - Andrew Wacker
- University of Missouri – Kansas City, School of Medicine, Department of Biomedical Sciences, USA
| | - Mark Dallas
- University of Missouri – Kansas City, School of Dentistry, Department of Oral & Craniofacial Sciences, USA
| | - Mark L. Johnson
- University of Missouri – Kansas City, School of Dentistry, Department of Oral & Craniofacial Sciences, USA
| | - Michael J. Wacker
- University of Missouri – Kansas City, School of Medicine, Department of Biomedical Sciences, USA
| |
Collapse
|
4
|
Meslier QA, Oehrlein R, Shefelbine SJ. Combined Effects of Mechanical Loading and Piezo1 Chemical Activation on 22-Months-Old Female Mouse Bone Adaptation. Aging Cell 2025:e70087. [PMID: 40410950 DOI: 10.1111/acel.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/23/2025] [Accepted: 04/11/2025] [Indexed: 05/26/2025] Open
Abstract
With age, bones mechanosensitivity is reduced, which limits their ability to adapt to loading. The exact mechanism leading to this loss of mechanosensitvity is still unclear, making developing effective treatment challenging. Current treatments mostly focus on preventing bone mass loss (such as bisphosphonates) or promoting bone formation (such as Sclerostin inhibitors) to limit the decline of bones mass. However, treatments do not target the cause of bone mass loss which may be, in part, due to the bone's inability to initiate a normal bone mechanoadaptation response. In this work, we investigated the effects of 2 weeks of tibia loading, and Piezo1 agonist injection in vivo on 22-month-old mouse bone adaptation response. We used an optimized loading profile, which induced high fluid flow velocity and low strain magnitude in adult mouse tibia. We found that tibia loading and Yoda2 injection have an additive effect on increasing cortical bone parameters in 22-month-old mice. In vivo osteocytes calcium signaling imaging suggests that Yoda2 is able to reach osteocytes and activate Piezo1. This combination of mechanical and chemical stimulation could be a promising treatment strategy to help promote bone formation in patients who have low bone mass due to aging.
Collapse
Affiliation(s)
- Quentin A Meslier
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Robert Oehrlein
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Chu T, Wasi M, Guerra RM, Song X, Wang S, Sims-Mourtada J, You L, Wang L. Skeletal response to Yoda1 and whole-body vibration in mice varied with animal age, bone compartment, treatment duration, and radiation exposure. Bone 2025; 198:117525. [PMID: 40389188 DOI: 10.1016/j.bone.2025.117525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/13/2025] [Accepted: 05/13/2025] [Indexed: 05/21/2025]
Abstract
In this study, we investigated the skeletal effects of Yoda1, an agonist of the mechanosensitive Piezo1 channels, and whole-body vibration (WBV), alone and combined, in young mice (8-week-old) and in mature (31- to 36-week-old) mice after radiation exposure. Our goal was to determine whether the two mechanobiology-based interventions, known to induce anabolic response individually in young subjects, could promote bone health of older subjects undergoing cancer treatments such as radiotherapy. Our hypothesis was that the combination of Yoda1 and WBV could improve young skeletons and protect mature skeletons after radiotherapy better than Yoda1 or WBV alone. Our in vivo experiments demonstrated (1) that Yoda1 (5 mg/kg body weight) alone or combined with WBV (0.3 g, 13 Hz, 30 min/day, 5 days/week, 4 weeks) enhanced bone growth similarly (∼2 folds relative to nontreated controls) in young mice; (2) that mature mice were unresponsive to individual interventions but exhibited less polar moment of inertia loss (-56 %) in the tibiae receiving the combination of Yoda1 and WBV (15 min/day) but no radiation exposure; and (3) that the contralateral tibiae receiving fractionated radiation (2 × 8 Gy over three days) did not show different treatment responses in Week 4, while they responded to the combination therapy (increased cortical bone formation) in Week 2. Interestingly, pair comparisons of the irradiated and non-irradiated tibiae of the same animals revealed that radiation exposure resulted in decreased trabecular bone loss regardless of the treatments and increased the percentage of tibiae maintaining better cortical polar moment of inertia and cortical area in the groups receiving Yoda1 or the combination therapy. The complex skeletal responses to Yoda1 and/or WBV were compartment specific (cortical or trabecular bone) and dependent on animal age, radiation exposure, and treatment duration. This study partially supported our original hypothesis, while suggesting the need of finetuning the Yoda1 and WBV regimens and elucidating the underlying mechanisms in order to effectively treat age and radiation induced bone loss.
Collapse
Affiliation(s)
- Tiankuo Chu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Murtaza Wasi
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Rosa M Guerra
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Xin Song
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Shubo Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | | | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada; Department of Mechanical and Materials Engineering, Queens University, Ontario, Canada
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
6
|
Chougule A, Zhang C, Denbow J, Vinokurov N, Mendez D, Vojtisek E, Gardinier J. P2Y 2 Inhibition Modifies the Anabolic Response to Exercise in Adult Mice. Aging Cell 2025; 24:e14464. [PMID: 39741419 PMCID: PMC12074023 DOI: 10.1111/acel.14464] [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: 08/05/2024] [Revised: 11/04/2024] [Accepted: 12/08/2024] [Indexed: 01/03/2025] Open
Abstract
As the aging population continues to grow, the incidence of osteoporotic fractures increases and is compounded by our lack of therapeutic strategies that increase bone formation. Although exercise and physical activity play a key role in maintaining bone mass throughout our lives, the loads and exertion required to elicit an anabolic response becomes exceedingly difficult to achieve with age. Based on previous work, the P2Y2 receptor offers a unique therapeutic target to increasing bone mass by modifying the mechanotransduction. Others have also shown P2Y2 to have a negative effect on osteoblast function. However, the extent to which inhibiting P2Y2 pharmaceutically improves bone mass or the mechanotransduction of bone remains unknown. Our central hypothesis for this study states that inhibiting P2Y2 activity can enhance the anabolic response to loading in an aging population. To test this hypothesis, the anabolic response to exercise was examined by treating adult mice, which typically display a minimal response, with the P2Y2 inhibitor AR-C118925XX (ARC). Our findings from this study demonstrate that ARC treatment of adult mice increases periosteal bone formation in response to exercise. The enhanced response to exercise was characterized by a reduction in osteocytes' induction of osteoclast activity. Endocortical bone formation also increased with treatment independently of exercise, providing gains in mechanical strength and tissue level properties. Overall, inhibiting P2Y2 activation has a beneficial effect on bone formation and the anabolic response to loading, namely by limiting osteoclast activation.
Collapse
Affiliation(s)
- Amit Chougule
- Bone and Joint CenterHenry Ford Health SystemDetroitMichiganUSA
- Henry Ford Health + Michigan State University Health SciencesDetroitMichiganUSA
- Department of PhysiologyCollege of Human MedicineMichigan State UniversityEast LansingMichiganUSA
| | - Chunbin Zhang
- Bone and Joint CenterHenry Ford Health SystemDetroitMichiganUSA
| | - Jordan Denbow
- Bone and Joint CenterHenry Ford Health SystemDetroitMichiganUSA
| | | | - Devin Mendez
- School of MedicineWayne State UniversityDetroitMichiganUSA
| | | | - Joseph Gardinier
- Bone and Joint CenterHenry Ford Health SystemDetroitMichiganUSA
- Henry Ford Health + Michigan State University Health SciencesDetroitMichiganUSA
- Department of PhysiologyCollege of Human MedicineMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
7
|
Meslier QA, Hoffmann J, Oehrlein R, Kurczy D, Monaghan JR, Shefelbine SJ. 3D spatial distribution of Sost mRNA and sclerostin protein expression in response to in vivo tibia loading in female mice. Bone 2025; 193:117422. [PMID: 39978613 DOI: 10.1016/j.bone.2025.117422] [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: 09/26/2024] [Revised: 01/13/2025] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
Bones adapt to external mechanical loads through a process known as mechanoadaptation. Osteocytes are the bone cells that sense the mechanical environment and initiate a biological response. Investigating the changes in osteocyte molecular expression following mechanical loading has been instrumental in characterizing the regulatory pathways involved in bone adaptation. However, current methods for examining osteocyte molecular expression do not preserve the three-dimensional structure of the bone, which plays a critical role in the mechanical stimuli sensed by the osteocytes and their spatially controlled biological responses. In this study, we used WISH-BONE (Whole-mount In Situ Histology of Bone) to investigate the spatial distribution of Sost-mRNA transcripts and its encoded protein, sclerostin, in 3D mouse tibia midshaft following in vivo tibia loading. Our findings showed a decrease in the percentage of Sost-positive osteocytes, after loading, along the posterior-lateral side of the tibia. The number of sclerostin-positive osteocytes were found to significantly decrease at a very specific 2D location of the tibia after loading. However, in 3D, the total number of sclerostin-positive osteocytes was similar between loaded and control legs. This work is the first to provide a 3D analysis of Sost and sclerostin distribution in loaded versus contralateral mouse tibia midshafts. It also highlights the importance of the bone region analyzed and the method utilized when interpreting mechanoadaptation results. WISH-BONE represents a powerful tool for further characterization of mechanosensitive genes regulation in bone and holds the potential for advancing the development of new treatments targeting mechanosensitivity-related bone disorders.
Collapse
Affiliation(s)
- Quentin A Meslier
- Department of Bioengineering, Northeastern University, Boston, MA, United States; LifeCanvas Technologies, Cambridge, MA, United States.
| | - Jacy Hoffmann
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Robert Oehrlein
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Daniel Kurczy
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - James R Monaghan
- Department of Bioengineering, Northeastern University, Boston, MA, United States; Department of Biology, Northeastern University, Boston, MA, United States; Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, MA, United States
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, MA, United States; Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, MA, United States
| |
Collapse
|
8
|
Resende-Coelho A, Ali MM, James A, Warren A, Gatrell L, Kadhim I, Fu Q, Xiong J, Onal M, Almeida M. Mitochondrial oxidative stress or decreased autophagy in osteoblast lineage cells is not sufficient to mimic the deleterious effects of aging on bone mechanoresponsiveness. Aging (Albany NY) 2025; 17:610-629. [PMID: 40105873 PMCID: PMC11984430 DOI: 10.18632/aging.206213] [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: 06/07/2024] [Accepted: 01/23/2025] [Indexed: 03/20/2025]
Abstract
Exercise-induced mechanical load stimulates bone cells, including osteocytes, to promote bone formation. The bone response to loading is less effective with aging, but the cellular and molecular mechanisms responsible for the impaired mechanoresponsiveness remain unclear. Excessive mitochondrial reactive oxygen species (mtROS) and deficient autophagy are common aging mechanisms implicated in decreased bone formation in old mice. Here, we confirmed that the osteogenic effects of tibia compressive loading are lower in old versus young female mice. We also examined whether an increase in mtROS or decreased autophagy in osteoblast-lineage cells of adult female mice could mimic the deleterious effects of aging. To this end, we loaded mice lacking the antioxidant enzyme superoxide dismutase 2 (Sod2) or autophagy-related 7 (Atg7) in cells targeted by Osterix1 (Osx1)-Cre. Osteocytes in Atg7ΔOsx1 exhibited altered morphology and decreased osteocyte dendrite projections. Two weeks of loading increased cortical bone mass and bone formation rate at both periosteal and endosteal surfaces of Osx1-Cre control mice. Nonetheless, in both Atg7ΔOsx1 and Sod2ΔOsx1 mice the response to loading was identical to that observed in control mice, indicating that compromised Atg7-dependent autophagy or excessive mtROS are not sufficient to impair the bone response to tibial compressive loading. Thus, alternative mechanisms of aging might be responsible for the decreased response of the aged skeleton to mechanical stimuli. These findings also suggest that an intact osteocyte dendrite network is not required for the osteogenic response in this model of bone loading.
Collapse
Affiliation(s)
- Ana Resende-Coelho
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Md Mohsin Ali
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alicen James
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Aaron Warren
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Landon Gatrell
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ilham Kadhim
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Qiang Fu
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Melda Onal
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
9
|
Mathavan N, Singh A, Marques FC, Günther D, Kuhn GA, Wehrle E, Müller R. Spatial transcriptomics in bone mechanomics: Exploring the mechanoregulation of fracture healing in the era of spatial omics. SCIENCE ADVANCES 2025; 11:eadp8496. [PMID: 39742473 DOI: 10.1126/sciadv.adp8496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
In recent decades, the field of bone mechanobiology has sought experimental techniques to unravel the molecular mechanisms governing the phenomenon of mechanically regulated fracture healing. Each cell within a fracture site resides within different local microenvironments characterized by different levels of mechanical strain; thus, preserving the spatial location of each cell is critical in relating cellular responses to mechanical stimuli. Our spatial transcriptomics-based "mechanomics" platform facilitates spatially resolved analysis of the molecular profiles of cells with respect to their local in vivo mechanical environment by integrating time-lapsed in vivo micro-computed tomography, spatial transcriptomics, and micro-finite element analysis. We investigate the transcriptomic responses of cells as a function of the local strain magnitude by identifying the differential expression of genes in regions of high and low strain within a fracture site. Our platform thus has the potential to address fundamental open questions within the field and to discover mechano-responsive targets to enhance fracture healing.
Collapse
Affiliation(s)
| | - Amit Singh
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | | | - Denise Günther
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| | - Esther Wehrle
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
- AO Research Institute Davos, Davos Platz, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
10
|
Zhao D, Tu C, Zhang L, Guda T, Gu S, Jiang JX. Activation of connexin hemichannels enhances mechanosensitivity and anabolism in disused and aged bone. JCI Insight 2024; 9:e177557. [PMID: 39641271 PMCID: PMC11623949 DOI: 10.1172/jci.insight.177557] [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: 11/14/2023] [Accepted: 10/11/2024] [Indexed: 12/07/2024] Open
Abstract
Mechanical loading, essential for bone health, promotes bone formation and remodeling. However, the positive response diminishes in cases of disuse and aging, leading to bone loss and an increased fracture risk. This study demonstrates that activating hemichannels (HCs) using a connexin 43 (Cx43) antibody, Cx43(M2), in bone osteocytes revitalizes aging and disused bones. Using a hindlimb suspension (HLS) disuse model and a tibial mechanical loading model, we found that Cx43(M2) inhibited bone loss and osteocyte apoptosis induced by unloading in 16-week-old adult mice. Additionally, it enhanced bone mass in response to tibial loading in 22-month-old aged mice. The HC opening released bone anabolic factor prostaglandin E2 (PGE2) and suppressed catabolic factor sclerostin (SOST). This suppressed the increase of cortical bone formation and reduction of bone resorption during unloading and promoted trabecular and cortical bone formation during loading. Cx43(M2)-induced HC opening, coupled with PGE2 release, effectively rescued unloading-induced bone loss and restored the diminished anabolic response of aged bones to mechanical loading. Activating HCs with the Cx43 antibody holds promise as a de novo therapeutic approach, as it can overcome the limitations of existing treatment regimens for treating bone loss and osteoporosis associated with aging and disuse.
Collapse
Affiliation(s)
- Dezhi Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas, USA
- School of Medicine, Northwest University, Xi’an, China
| | - Chao Tu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas, USA
- The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lidan Zhang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas, USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas, USA
| | - Jean X. Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, Texas, USA
| |
Collapse
|
11
|
Wasi M, Chu T, Guerra RM, Kooker R, Maldonado K, Li X, Lin CY, Song X, Xiong J, You L, Wang L. Mitigating aging and doxorubicin induced bone loss in mature mice via mechanobiology based treatments. Bone 2024; 188:117235. [PMID: 39147353 PMCID: PMC11475016 DOI: 10.1016/j.bone.2024.117235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Aging leads to a reduced anabolic response to mechanical stimuli and a loss of bone mass and structural integrity. Chemotherapy agents such as doxorubicin exacerbate the degeneration of aging skeleton and further subject older cancer patients to a higher fracture risk. To alleviate this clinical problem, we proposed and tested a novel mechanobiology-based therapy. Building upon prior findings that i) Yoda1, the Piezo1 agonist, promoted bone growth in young adult mice and suppressed bone resorption markers in aged mice, and ii) moderate tibial loading protected bone from breast cancer-induced osteolysis, we hypothesized that combined Yoda1 and moderate loading would improve the structural integrity of adult and aged skeletons in vivo and protect bones from deterioration after chemotherapy. We first examined the effects of 4-week Yoda1 (dose 5 mg/kg, 5 times/week) and moderate tibial loading (4.5 N peak load, 4 Hz, 300 cycles for 5 days/week), individually and combined, on mature mice (∼50 weeks of age). Combined Yoda1 and loading was found to mitigate age-associated cortical and trabecular bone loss better than individual interventions. As expected, the non-treated controls experienced an average drop of cortical polar moment of inertia (Ct.pMOI) by -4.3 % over four weeks and the bone deterioration occurred in the majority (64 %) of the samples. Relative to no treatment, loading alone, Yoda1 alone, and combined Yoda1 and loading increased Ct.pMOI by +7.3 %, +9.5 %, +12.0 % and increased the % of samples with positive Ct.pMOI changes by +32 %, +26 %, and +43 %, respectively, suggesting an additive protection of aging-related bone loss for the combined therapy. We further tested if the treatment efficacy was preserved in mature mice following two weeks (six injections) of doxorubicin at the dose of 2.5 or 5 mg/kg. As expected, doxorubicin increased osteocyte apoptosis, altered bone remodeling, and impaired bone structure. However, the effects induced by DOX were too severe to be rescued by Yoda1 and loading, alone or combined, although loading and Yoda1 individually, or combined, increased the number of mice showing positive responsiveness by 0 %, +15 %, and +29 % relative to no intervention after doxorubicin exposure. Overall, this study supported the potentials and challenges of the Yoda1-based strategy in mitigating the detrimental skeletal effects caused by aging and doxorubicin.
Collapse
Affiliation(s)
- Murtaza Wasi
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Tiankuo Chu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Rosa M Guerra
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Rory Kooker
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Kenneth Maldonado
- Department of Biomedical Engineering, Kansas State University, Manhattan, KS, USA
| | - Xuehua Li
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Chun-Yu Lin
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Xin Song
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
12
|
Meslier QA, Duerr TJ, Guan W, Nguyen B, Monaghan JR, Shefelbine SJ. WISH-BONE: Whole-mount in situ histology, to label osteocyte mRNA and protein in 3D adult mouse bones. FASEB J 2024; 38:e70101. [PMID: 39387181 DOI: 10.1096/fj.202400635r] [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: 03/22/2024] [Revised: 07/19/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024]
Abstract
Bone is a three-dimensional (3D) highly dynamic tissue under constant remodeling. Commonly used tools to investigate bone biology require sample digestion for biomolecule extraction or provide only two-dimensional (2D) spatial information. There is a need for 3D tools to investigate spatially preserved biomarker expression in osteocytes. In this work, we present a new method, WISH-BONE, to label osteocyte messenger RNA (mRNA) and protein in whole-mount mouse bone. For mRNA labeling, we used hybridization chain reaction-fluorescence in situ hybridization (HCR-FISH) to label genes of interest in osteocytes. For protein labeling, samples were preserved using an epoxy-based solution that protects tissue structure and biomolecular components. Then an enzymatic matrix permeabilization step was performed to enable antibody penetration. Immunostaining was used to label various proteins involved in bone homeostasis. We also demonstrate the use of customized fluorescent nanobodies to target and label proteins in the cortical bone (CB). However, the relatively dim signal observed from nanobodies' staining limited detection. mRNA and protein labeling were performed in separate samples. In this study, we share protocols, highlight opportunities, and identify the challenges of this novel 3D labeling method. They are the first protocols for whole-mount osteocyte 3D labeling of mRNA and protein in mature mouse bones. WISH-BONE will allow the investigation of molecular signaling in bone cells in their 3D environment and could be applied to various bone-related fields of research.
Collapse
Affiliation(s)
- Quentin A Meslier
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- LifeCanvas Technologies, Cambridge, Massachusetts, USA
| | - Timothy J Duerr
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| | - Webster Guan
- LifeCanvas Technologies, Cambridge, Massachusetts, USA
| | - Brian Nguyen
- LifeCanvas Technologies, Cambridge, Massachusetts, USA
| | - James R Monaghan
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Department of Biology, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Chermside-Scabbo CJ, Shuster JT, Erdmann-Gilmore P, Tycksen E, Zhang Q, Townsend RR, Silva MJ. A proteomics approach to study mouse long bones: examining baseline differences and mechanical loading-induced bone formation in young-adult and old mice. Aging (Albany NY) 2024; 16:12726-12768. [PMID: 39400554 PMCID: PMC11501390 DOI: 10.18632/aging.206131] [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: 06/20/2023] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
With aging, bone mass declines and the anabolic effects of skeletal loading diminish. While much research has focused on gene transcription, how bone ages and loses its mechanoresponsiveness at the protein level remains unclear. We developed a novel proteomics approach and performed a paired mass spectrometry and RNA-seq analysis on tibias from young-adult (5-month) and old (22-month) mice. We report the first correlation estimate between the bone proteome and transcriptome (Spearman ρ = 0.40), which is in line with other tissues but indicates that a relatively low amount of variation in protein levels is explained by the variation in transcript levels. Of 71 shared targets that differed with age, eight were associated with bone mineral density in previous GWAS, including understudied targets Asrgl1 and Timp2. We used complementary RNA in situ hybridization to confirm that Asrgl1 and Timp2 had reduced expression in osteoblasts/osteocytes in old bones. We also found evidence for reduced TGF-beta signaling with aging, in particular Tgfb2. Next, we defined proteomic changes following mechanical loading. At the protein level, bone differed more with age than with loading, and aged bone had fewer loading-induced changes. Overall, our findings underscore the need for complementary protein-level assays in skeletal biology research.
Collapse
Affiliation(s)
- Christopher J. Chermside-Scabbo
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - John T. Shuster
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Petra Erdmann-Gilmore
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Eric Tycksen
- Department of Genetics, McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO 63108, USA
| | - Qiang Zhang
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - R. Reid Townsend
- Department of Medicine, Proteomics Core, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63105, USA
| |
Collapse
|
14
|
Li X, Zhang C, Vail CE, Sherrill JT, Xiong J. Piezo1 expression in mature osteocytes is dispensable for the skeletal response to mechanical loading. Bone 2024; 190:117276. [PMID: 39389439 DOI: 10.1016/j.bone.2024.117276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
Mechanical loading is essential for bone growth and homeostasis, with osteocytes considered the primary mechanosensors. Deleting the mechanosensitive ion channel Piezo1 from Dmp1-Cre-targeted cells, which include both osteoblasts and osteocytes, resulted in reduced bone mass and impaired skeletal responses to mechanical stimuli. To specifically isolate Piezo1's role in osteocytes, we used Sost-Cre mice to generate mice with Piezo1 deletion exclusively in mature osteocytes. These mice exhibited lower bone mineral density, decreased cancellous bone mass, and reduced cortical thickness with decrease periosteal expansion. However, unlike mice lacking Piezo1 in both osteoblasts and osteocytes, those with Piezo1 deletion in mature osteocytes responded normally to mechanical loading. These findings suggest that Piezo1 expression in mature osteocytes contributes to bone maintenance under normal physiological condition, but is dispensable for the skeletal response to mechanical loading.
Collapse
Affiliation(s)
- Xuehua Li
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Connie Zhang
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Cameron E Vail
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - John T Sherrill
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
15
|
Gilbert SJ, Jones R, Egan BJ, Bonnet CS, Evans SL, Mason DJ. Investigating mechanical and inflammatory pathological mechanisms in osteoarthritis using MSC-derived osteocyte-like cells in 3D. Front Endocrinol (Lausanne) 2024; 15:1359052. [PMID: 39157681 PMCID: PMC11328832 DOI: 10.3389/fendo.2024.1359052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 07/17/2024] [Indexed: 08/20/2024] Open
Abstract
Introduction Changes to bone physiology play a central role in the development of osteoarthritis with the mechanosensing osteocyte releasing factors that drive disease progression. This study developed a humanised in vitro model to detect osteocyte responses to either interleukin-6, a driver of degeneration and bone remodelling in animal and human joint injury, or mechanical loading, to mimic osteoarthritis stimuli in joints. Methods Human MSC cells (Y201) were differentiated in 3-dimensional type I collagen gels in osteogenic media and osteocyte phenotype assessed by RTqPCR and immunostaining. Gels were subjected to a single pathophysiological load or stimulated with interleukin-6 with unloaded or unstimulated cells as controls. RNA was extracted 1-hour post-load and assessed by RNAseq. Markers of pain, bone remodelling, and inflammation were quantified by RT-qPCR and ELISA. Results Y201 cells embedded within 3D collagen gels assumed dendritic morphology and expressed mature osteocytes markers. Mechanical loading of the osteocyte model regulated 7564 genes (Padj p<0.05, 3026 down, 4538 up). 93% of the osteocyte transcriptome signature was expressed in the model with 38% of these genes mechanically regulated. Mechanically loaded osteocytes regulated 26% of gene ontology pathways linked to OA pain, 40% reflecting bone remodelling and 27% representing inflammation. Load regulated genes associated with osteopetrosis, osteoporosis and osteoarthritis. 42% of effector genes in a genome-wide association study meta-analysis were mechanically regulated by osteocytes with 10 genes representing potential druggable targets. Interleukin-6 stimulation of osteocytes at concentrations reported in human synovial fluids from patients with OA or following knee injury, regulated similar readouts to mechanical loading including markers of pain, bone remodelling, and inflammation. Discussion We have developed a reproducible model of human osteocyte like cells that express >90% of the genes in the osteocyte transcriptome signature. Mechanical loading and inflammatory stimulation regulated genes and proteins implicated in osteoarthritis symptoms of pain as well as inflammation and degeneration underlying disease progression. Nearly half of the genes classified as 'effectors' in GWAS were mechanically regulated in this model. This model will be useful in identifying new mechanisms underlying bone and joint pathologies and testing drugs targeting those mechanisms.
Collapse
Affiliation(s)
- Sophie J. Gilbert
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Ryan Jones
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Ben J. Egan
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Cleo Selina Bonnet
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Sam L. Evans
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Engineering, Cardiff University, Cardiff, United Kingdom
| | - Deborah J. Mason
- Biomechanics and Bioengineering Centre Versus Arthritis, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| |
Collapse
|
16
|
Rindt WD, Krug M, Yamada S, Sennefelder F, Belz L, Cheng WH, Azeem M, Kuric M, Evers M, Leich E, Hartmann TN, Pereira AR, Hermann M, Hansmann J, Mussoni C, Stahlhut P, Ahmad T, Yassin MA, Mustafa K, Ebert R, Jundt F. A 3D bioreactor model to study osteocyte differentiation and mechanobiology under perfusion and compressive mechanical loading. Acta Biomater 2024; 184:210-225. [PMID: 38969078 DOI: 10.1016/j.actbio.2024.06.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/07/2024]
Abstract
Osteocytes perceive and process mechanical stimuli in the lacuno-canalicular network in bone. As a result, they secrete signaling molecules that mediate bone formation and resorption. To date, few three-dimensional (3D) models exist to study the response of mature osteocytes to biophysical stimuli that mimic fluid shear stress and substrate strain in a mineralized, biomimetic bone-like environment. Here we established a biomimetic 3D bone model by utilizing a state-of-art perfusion bioreactor platform where immortomouse/Dmp1-GFP-derived osteoblastic IDG-SW3 cells were differentiated into mature osteocytes. We evaluated proliferation and differentiation properties of the cells on 3D microporous scaffolds of decellularized bone (dBone), poly(L-lactide-co-trimethylene carbonate) lactide (LTMC), and beta-tricalcium phosphate (β-TCP) under physiological fluid flow conditions over 21 days. Osteocyte viability and proliferation were similar on the scaffolds with equal distribution of IDG-SW3 cells on dBone and LTMC scaffolds. After seven days, the differentiation marker alkaline phosphatase (Alpl), dentin matrix acidic phosphoprotein 1 (Dmp1), and sclerostin (Sost) were significantly upregulated in IDG-SW3 cells (p = 0.05) on LTMC scaffolds under fluid flow conditions at 1.7 ml/min, indicating rapid and efficient maturation into osteocytes. Osteocytes responded by inducing the mechanoresponsive genes FBJ osteosarcoma oncogene (Fos) and prostaglandin-endoperoxide synthase 2 (Ptgs2) under perfusion and dynamic compressive loading at 1 Hz with 5 % strain. Together, we successfully created a 3D biomimetic platform as a robust tool to evaluate osteocyte differentiation and mechanobiology in vitro while recapitulating in vivo mechanical cues such as fluid flow within the lacuno-canalicular network. STATEMENT OF SIGNIFICANCE: This study highlights the importance of creating a three-dimensional (3D) in vitro model to study osteocyte differentiation and mechanobiology, as cellular functions are limited in two-dimensional (2D) models lacking in vivo tissue organization. By using a perfusion bioreactor platform, physiological conditions of fluid flow and compressive loading were mimicked to which osteocytes are exposed in vivo. Microporous poly(L-lactide-co-trimethylene carbonate) lactide (LTMC) scaffolds in 3D are identified as a valuable tool to create a favorable environment for osteocyte differentiation and to enable mechanical stimulation of osteocytes by perfusion and compressive loading. The LTMC platform imitates the mechanical bone environment of osteocytes, allowing the analysis of the interaction with other cell types in bone under in vivo biophysical stimuli.
Collapse
Affiliation(s)
- Wyonna Darleen Rindt
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Melanie Krug
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig-Haus, University of Würzburg, Würzburg, Germany
| | - Shuntaro Yamada
- Centre of Translational Oral Research (TOR)-Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | | | - Louisa Belz
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Wen-Hui Cheng
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Muhammad Azeem
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Martin Kuric
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig-Haus, University of Würzburg, Würzburg, Germany
| | | | - Ellen Leich
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Tanja Nicole Hartmann
- Department of Medicine I, Medical Center-University Freiburg, and Faculty of Medicine, University of Freiburg, Freiburg im Breisgau, Germany
| | - Ana Rita Pereira
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
| | - Marietta Hermann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Würzburg, Würzburg, Germany
| | - Jan Hansmann
- Fraunhofer Institute for Silicate Research ISC, Translational Center Regenerative Therapies, Würzburg, Germany; Department of Electrical Engineering, University of Applied Sciences Würzburg-Schweinfurt, Schweinfurt, Germany
| | - Camilla Mussoni
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication (IFB), and Bavarian Polymer Institute (BPI), University of Würzburg, Würzburg, Germany
| | - Philipp Stahlhut
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication (IFB), and Bavarian Polymer Institute (BPI), University of Würzburg, Würzburg, Germany
| | - Taufiq Ahmad
- Department for Functional Materials in Medicine and Dentistry, Institute of Functional Materials and Biofabrication (IFB), and Bavarian Polymer Institute (BPI), University of Würzburg, Würzburg, Germany
| | - Mohammed Ahmed Yassin
- Centre of Translational Oral Research (TOR)-Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Kamal Mustafa
- Centre of Translational Oral Research (TOR)-Tissue Engineering Group, Department of Clinical Dentistry, University of Bergen, Bergen, Norway
| | - Regina Ebert
- Department of Musculoskeletal Tissue Regeneration, Orthopedic Clinic König-Ludwig-Haus, University of Würzburg, Würzburg, Germany
| | - Franziska Jundt
- Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
17
|
Bhadouria N, Holguin N. Osteoporosis treatments for intervertebral disc degeneration and back pain: a perspective. JBMR Plus 2024; 8:ziae048. [PMID: 38706880 PMCID: PMC11066806 DOI: 10.1093/jbmrpl/ziae048] [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: 10/02/2023] [Revised: 03/18/2024] [Accepted: 03/23/2024] [Indexed: 05/07/2024] Open
Abstract
Low back pain derived from intervertebral disc (IVD) degeneration is a debilitating spinal condition that, despite its prevalence, does not have any intermediary guidelines for pharmacological treatment between palliative care and invasive surgery. The development of treatments for the IVD is complicated by the variety of resident cell types needed to maintain the regionally distinct structural properties of the IVD that permit the safe, complex motions of the spine. Osteoporosis of the spine increases the risk of vertebral bone fracture that can increase the incidence of back pain. Fortunately, there are a variety of pharmacological treatments for osteoporosis that target osteoblasts, osteoclasts and/or osteocytes to build bone and prevent vertebral fracture. Of particular note, clinical and preclinical studies suggest that commonly prescribed osteoporosis drugs like bisphosphonates, intermittent parathyroid hormone, anti-sclerostin antibody, selective estrogen receptor modulators and anti-receptor activator of nuclear factor-kappa B ligand inhibitor denosumab may also relieve back pain. Here, we cite clinical and preclinical studies and include unpublished data to support the argument that a subset of these therapeutics for osteoporosis may alleviate low back pain by also targeting the IVD.
Collapse
Affiliation(s)
- Neharika Bhadouria
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, United States
| | - Nilsson Holguin
- Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| |
Collapse
|
18
|
Tetrault E, Aaronson B, Gilbert MC, Albertson RC. Foraging-induced craniofacial plasticity is associated with an early, robust and dynamic transcriptional response. Proc Biol Sci 2024; 291:20240215. [PMID: 38654651 PMCID: PMC11040245 DOI: 10.1098/rspb.2024.0215] [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: 02/12/2024] [Accepted: 03/19/2024] [Indexed: 04/26/2024] Open
Abstract
Phenotypic plasticity is the ability of a single genotype to vary its phenotype in response to the environment. Plasticity of the skeletal system in response to mechanical input is widely studied, but the timing of its transcriptional regulation is not well understood. Here, we used the cichlid feeding apparatus to examine the transcriptional dynamics of skeletal plasticity over time. Using three closely related species that vary in their ability to remodel bone and a panel of 11 genes, including well-studied skeletal differentiation markers and newly characterized environmentally sensitive genes, we examined plasticity at one, two, four and eight weeks following the onset of alternate foraging challenges. We found that the plastic species exhibited environment-specific bursts in gene expression beginning at one week, followed by a sharp decline in levels, while the species with more limited plasticity exhibited consistently low levels of gene expression. This trend held across nearly all genes, suggesting that it is a hallmark of the larger plasticity regulatory network. We conclude that plasticity of the cichlid feeding apparatus is not the result of slowly accumulating gene expression difference over time, but rather is stimulated by early bursts of environment-specific gene expression followed by a return to homeostatic levels.
Collapse
Affiliation(s)
- Emily Tetrault
- Molecular and Cell Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - Ben Aaronson
- Department of Biology, University of Massachusetts, Amherst, MA 01003, USA
| | - Michelle C. Gilbert
- Department of Biology, Pennsylvania State University, State College, PA 16802, USA
| | - R. Craig Albertson
- Department of Biology, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
19
|
Buck HV, Stains JP. Osteocyte-mediated mechanical response controls osteoblast differentiation and function. Front Physiol 2024; 15:1364694. [PMID: 38529481 PMCID: PMC10961341 DOI: 10.3389/fphys.2024.1364694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/29/2024] [Indexed: 03/27/2024] Open
Abstract
Low bone mass is a pervasive global health concern, with implications for osteoporosis, frailty, disability, and mortality. Lifestyle factors, including sedentary habits, metabolic dysfunction, and an aging population, contribute to the escalating prevalence of osteopenia and osteoporosis. The application of mechanical load to bone through physical activity and exercise prevents bone loss, while sufficient mechanical load stimulates new bone mass acquisition. Osteocytes, cells embedded within the bone, receive mechanical signals and translate these mechanical cues into biological signals, termed mechano-transduction. Mechano-transduction signals regulate other bone resident cells, such as osteoblasts and osteoclasts, to orchestrate changes in bone mass. This review explores the mechanisms through which osteocyte-mediated response to mechanical loading regulates osteoblast differentiation and bone formation. An overview of bone cell biology and the impact of mechanical load will be provided, with emphasis on the mechanical cues, mechano-transduction pathways, and factors that direct progenitor cells toward the osteoblast lineage. While there are a wide range of clinically available treatments for osteoporosis, the majority act through manipulation of the osteoclast and may have significant disadvantages. Despite the central role of osteoblasts to the deposition of new bone, few therapies directly target osteoblasts for the preservation of bone mass. Improved understanding of the mechanisms leading to osteoblastogenesis may reveal novel targets for translational investigation.
Collapse
Affiliation(s)
| | - Joseph Paul Stains
- School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
20
|
Ahmad M, Haffner-Luntzer M, Schoppa A, Najafova Z, Lukic T, Yorgan TA, Amling M, Schinke T, Ignatius A. Mechanical induction of osteoanabolic Wnt1 promotes osteoblast differentiation via Plat. FASEB J 2024; 38:e23489. [PMID: 38407813 DOI: 10.1096/fj.202301424rr] [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: 07/13/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Physical activity-induced mechanical stimuli play a crucial role in preserving bone mass and structure by promoting bone formation. While the Wnt pathway is pivotal for mediating the osteoblast response to loading, the exact mechanisms are not fully understood. Here, we found that mechanical stimulation induces osteoblastic Wnt1 expression, resulting in an upregulation of key osteogenic marker genes, including Runx2 and Sp7, while Wnt1 knockdown using siRNA prevented these effects. RNAseq analysis identified Plat as a major target through which Wnt1 exerts its osteogenic influence. This was corroborated by Plat depletion using siRNA, confirming its positive role in osteogenic differentiation. Moreover, we demonstrated that mechanical stimulation enhances Plat expression, which, in turn leads to increased expression of osteogenic markers like Runx2 and Sp7. Notably, Plat depletion by siRNA prevented this effect. We have established that Wnt1 regulates Plat expression by activating β-Catenin. Silencing Wnt1 impairs mechanically induced β-Catenin activation, subsequently reducing Plat expression. Furthermore, our findings showed that Wnt1 is essential for osteoblasts to respond to mechanical stimulation and induce Runx2 and Sp7 expression, in part through the Wnt1/β-Catenin/Plat signaling pathway. Additionally, we observed significantly reduced Wnt1 and Plat expression in bones from ovariectomy (OVX)-induced and age-related osteoporotic mouse models compared with non-OVX and young mice, respectively. Overall, our data suggested that Wnt1 and Plat play significant roles in mechanically induced osteogenesis. Their decreased expression in bones from OVX and aged mice highlights their potential involvement in post-menopausal and age-related osteoporosis, respectively.
Collapse
Affiliation(s)
- Mubashir Ahmad
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | | | - Teodora Lukic
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
21
|
Zhang L, Guan Q, Wang Z, Feng J, Zou J, Gao B. Consequences of Aging on Bone. Aging Dis 2023; 15:2417-2452. [PMID: 38029404 PMCID: PMC11567267 DOI: 10.14336/ad.2023.1115] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023] Open
Abstract
With the aging of the global population, the incidence of musculoskeletal diseases has been increasing, seriously affecting people's health. As people age, the microenvironment within skeleton favors bone resorption and inhibits bone formation, accompanied by bone marrow fat accumulation and multiple cellular senescence. Specifically, skeletal stem/stromal cells (SSCs) during aging tend to undergo adipogenesis rather than osteogenesis. Meanwhile, osteoblasts, as well as osteocytes, showed increased apoptosis, decreased quantity, and multiple functional limitations including impaired mechanical sensing, intercellular modulation, and exosome secretion. Also, the bone resorption function of macrophage-lineage cells (including osteoclasts and preosteoclasts) was significantly enhanced, as well as impaired vascularization and innervation. In this study, we systematically reviewed the effect of aging on bone and the within microenvironment (including skeletal cells as well as their intracellular structure variations, vascular structures, innervation, marrow fat distribution, and lymphatic system) caused by aging, and mechanisms of osteoimmune regulation of the bone environment in the aging state, and the causal relationship with multiple musculoskeletal diseases in addition with their potential therapeutic strategy.
Collapse
Affiliation(s)
- Lingli Zhang
- College of Athletic Performance, Shanghai University of Sport, Shanghai, China
| | - Qiao Guan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Zhikun Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jie Feng
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jun Zou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Bo Gao
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| |
Collapse
|
22
|
He X, Hu W, Zhang Y, Chen M, Ding Y, Yang H, He F, Gu Q, Shi Q. Cellular senescence in skeletal disease: mechanisms and treatment. Cell Mol Biol Lett 2023; 28:88. [PMID: 37891477 PMCID: PMC10612178 DOI: 10.1186/s11658-023-00501-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The musculoskeletal system supports the movement of the entire body and provides blood production while acting as an endocrine organ. With aging, the balance of bone homeostasis is disrupted, leading to bone loss and degenerative diseases, such as osteoporosis, osteoarthritis, and intervertebral disc degeneration. Skeletal diseases have a profound impact on the motor and cognitive abilities of the elderly, thus creating a major challenge for both global health and the economy. Cellular senescence is caused by various genotoxic stressors and results in permanent cell cycle arrest, which is considered to be the underlying mechanism of aging. During aging, senescent cells (SnCs) tend to aggregate in the bone and trigger chronic inflammation by releasing senescence-associated secretory phenotypic factors. Multiple signalling pathways are involved in regulating cellular senescence in bone and bone marrow microenvironments. Targeted SnCs alleviate age-related degenerative diseases. However, the association between senescence and age-related diseases remains unclear. This review summarises the fundamental role of senescence in age-related skeletal diseases, highlights the signalling pathways that mediate senescence, and discusses potential therapeutic strategies for targeting SnCs.
Collapse
Affiliation(s)
- Xu He
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China
| | - Wei Hu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China
| | - Yuanshu Zhang
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214026, People's Republic of China
| | - Mimi Chen
- Department of Orthopedics, Children Hospital of Soochow University, No. 92 Zhongnan Street, Suzhou, Jiangsu, 215000, People's Republic of China
| | - Yicheng Ding
- Xuzhou Medical University, 209 Copper Mountain Road, Xuzhou, 221004, People's Republic of China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China
| | - Fan He
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China.
| | - Qiaoli Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China.
| | - Qin Shi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute of Soochow University, Medical College of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, People's Republic of China.
- Department of Orthopedics, Wuxi Ninth People's Hospital Affiliated to Soochow University, Wuxi, Jiangsu, 214026, People's Republic of China.
| |
Collapse
|
23
|
Wang Y, Ren L, Xu L, Wang J, Zhai J, Zhu G. Radiation Induces Bone Microenvironment Disruption by Activating the STING-TBK1 Pathway. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1316. [PMID: 37512126 PMCID: PMC10386124 DOI: 10.3390/medicina59071316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/30/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023]
Abstract
Background and Objectives: Damage to normal bone tissue following therapeutic irradiation (IR) represents a significant concern, as IR-induced bone microenvironment disruption can cause bone loss and create a more favorable environment for tumor metastases. The aim of the present study was to explore the cellular regulatory mechanism of IR-induced bone microenvironment disruption to effectively prevent radiotherapy-associated adverse effects in the future. Materials and Methods: In this study, a mouse model of local IR was established via local irradiation of the left hind limb of BALB/c mice with 12 Gy X-rays, and an in vitro osteocyte (OCY) model was established by exposing osteocyte-like MLO-Y4 cells to 2, 4, and 8 Gy irradiation to analyze multicellular biological injuries and cellular senescence. Small interfering RNA (siRNA) transfection at the cellular level and a selective antagonist intervention C-176 at the animal level were used to explore the potential role of the stimulator of interferon genes (STING) on IR-induced bone microenvironment disruption. Results: The results showed that 12 Gy local IR induces multicellular dysfunction, manifested as ascension of OCYs exfoliation, activation of osteoclastogenesis, degeneration of osteogenesis and fate conversion of adipogenesis, as well as cellular senescence and altered senescence-associated secretory phenotype (SASP) secretion. Furthermore, the expression of STING was significantly elevated, both in the primary OCYs harvested from locally irradiated mice and in vitro irradiated MLO-Y4 cells, accompanied by the markedly upregulated levels of phosphorylated TANK-binding kinase 1 (P-TBK1), RANKL and sclerostin (SOST). STING-siRNA transfection in vitro restored IR-induced upregulated protein expression of P-TBK1 and RANKL, as well as the mRNA expression levels of inflammatory cytokines, such as IL-1α, IL-6 and NF-κB, accompanied by the alleviation of excessive osteoclastogenesis. Finally, administration of the STING inhibitor C-176 mitigated IR-induced activation of osteoclastogenesis and restraint of osteogenesis, ameliorating the IR-induced biological damage of OCYs, consistent with the inhibition of P-TBK1, RANKL and SOST. Conclusions: The STING-P-TBK1 signaling pathway plays a crucial role in the regulation of the secretion of inflammatory cytokines and osteoclastogenesis potential in IR-induced bone microenvironment disruption. The selective STING antagonist can be used to intervene to block the STING pathway and, thereby, repair IR-induced multicellular biological damage and mitigate the imbalance between osteoclastogenesis and osteoblastgenesis.
Collapse
Affiliation(s)
- Yuyang Wang
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
- Shanghai Municipal Center for Disease Control & Prevention, Shanghai 200051, China
| | - Li Ren
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
| | - Linshan Xu
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
| | - Jianping Wang
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
| | - Jianglong Zhai
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
| | - Guoying Zhu
- Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China
| |
Collapse
|
24
|
Kitase Y, Prideaux M. Regulation of the Osteocyte Secretome with Aging and Disease. Calcif Tissue Int 2023; 113:48-67. [PMID: 37148298 DOI: 10.1007/s00223-023-01089-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/21/2023] [Indexed: 05/08/2023]
Abstract
As the most numerous and long-lived of all bone cells, osteocytes have essential functions in regulating skeletal health. Through the lacunar-canalicular system, secreted proteins from osteocytes can reach cells throughout the bone. Furthermore, the intimate connectivity between the lacunar-canalicular system and the bone vasculature allows for the transport of osteocyte-secreted factors into the circulation to reach the entire body. Local and endocrine osteocyte signaling regulates physiological processes such as bone remodeling, bone mechanoadaptation, and mineral homeostasis. However, these processes are disrupted by impaired osteocyte function induced by aging and disease. Dysfunctional osteocyte signaling is now associated with the pathogenesis of many disorders, including chronic kidney disease, cancer, diabetes mellitus, and periodontitis. In this review, we focus on the targeting of bone and extraskeletal tissues by the osteocyte secretome. In particular, we highlight the secreted osteocyte proteins, which are known to be dysregulated during aging and disease, and their roles during disease progression. We also discuss how therapeutic or genetic targeting of osteocyte-secreted proteins can improve both skeletal and systemic health.
Collapse
Affiliation(s)
- Yukiko Kitase
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA
| | - Matthew Prideaux
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
- Department of Anatomy, Cell Biology and Physiology, School of Medicine, Indiana University, Indianapolis, IN, 46202, USA.
| |
Collapse
|
25
|
Zagrodna A, Książek A, Słowińska-Lisowska M, Chmura J, Ponikowski P, Lombardi G. Effects of running a marathon on sclerostin and parathyroid hormone concentration in males aged over 50. J Sports Sci 2023; 41:796-802. [PMID: 37506230 DOI: 10.1080/02640414.2023.2240618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
The aim of our study was to verify whether running a marathon (32nd Wroclaw Marathon) was associated with changes in sclerostin and intact PTH (iPTH) concentration in middle-aged males. We enrolled 33 males who completed the marathon race. Blood samples were taken 60 minutes before (V1), immediately after (V2), and 7 days after the run (V3). The mean serum sclerostin concentration was 42.4 ± 10.8 pmol/L at V1, increased to 62.9 ± 12.6 pmol/L at V2 (t= -11.206; p < 0.001) and returned to baseline in V3 (t = 8.344; p < 0.001, V3 vs. V2). A similar trend was recorded for iPTH (t= -7.440; p < 0.001, for V2 vs. V1; t = 6.229; p < 0.001, for V3 vs. V2), at V3, iPTH levels remained significantly higher than V1 (t= -2.759; p = 0.010). The results of our study suggest that, in middle-aged males, running a marathon affects skeletal metabolism by activating two counteracting mechanisms, although temporarily overlapping: first, by a sudden inhibition of bone formation, through induction sclerostin expression and, secondly, by a long-lasting induction of PTH, which also guarantees the maintenance of adequate circulating levels of calcium. The net effect would be the maintenance of adequately high levels of circulating calcium to be used for neuromuscular activity and muscle contraction.
Collapse
Affiliation(s)
- Aleksandra Zagrodna
- Department of Biological and Medical Basis of Sport, Faculty of Physical Education and Sports, Wroclaw University of Health and Sport Sciences, Wroclaw, Poland
| | - Anna Książek
- Department of Biological and Medical Basis of Sport, Faculty of Physical Education and Sports, Wroclaw University of Health and Sport Sciences, Wroclaw, Poland
| | - Małgorzata Słowińska-Lisowska
- Department of Biological and Medical Basis of Sport, Faculty of Physical Education and Sports, Wroclaw University of Health and Sport Sciences, Wroclaw, Poland
| | - Jan Chmura
- Department of Biological and Motor Sport Bases, Faculty of Physical Education and Sports, Wroclaw University of Health and Sport Sciences, Wroclaw, Poland
| | - Piotr Ponikowski
- Department of Heart Diseases, Medical University, Wrocław, Poland
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry and Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| |
Collapse
|
26
|
Prideaux M, Smargiassi A, Peng G, Brotto M, Robling AG, Bonewald LF. L-BAIBA Synergizes with Sub-Optimal Mechanical Loading to Promote New Bone Formation. JBMR Plus 2023; 7:e10746. [PMID: 37283651 PMCID: PMC10241089 DOI: 10.1002/jbm4.10746] [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: 02/15/2023] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 06/08/2023] Open
Abstract
The L-enantiomer of β-aminoisobutyric acid (BAIBA) is secreted by contracted muscle in mice, and exercise increases serum levels in humans. In mice, L-BAIBA reduces bone loss with unloading, but whether it can have a positive effect with loading is unknown. Since synergism can be more easily observed with sub-optimal amounts of factors/stimulation, we sought to determine whether L-BAIBA could potentiate the effects of sub-optimal loading to enhance bone formation. L-BAIBA was provided in drinking water to C57Bl/6 male mice subjected to either 7 N or 8.25 N of sub-optimal unilateral tibial loading for 2 weeks. The combination of 8.25 N and L-BAIBA significantly increased the periosteal mineral apposition rate and bone formation rate compared to loading alone or BAIBA alone. Though L-BAIBA alone had no effect on bone formation, grip strength was increased, suggesting a positive effect on muscle function. Gene expression analysis of the osteocyte-enriched bone showed that the combination of L-BAIBA and 8.25 N induced the expression of loading-responsive genes such as Wnt1, Wnt10b, and the TGFb and BMP signaling pathways. One dramatic change was the downregulation of histone genes in response to sub-optimal loading and/or L-BAIBA. To determine early gene expression, the osteocyte fraction was harvested within 24 hours of loading. A dramatic effect was observed with L-BAIBA and 8.25 N loading as genes were enriched for pathways regulating the extracellular matrix (Chad, Acan, Col9a2), ion channel activity (Scn4b, Scn7a, Cacna1i), and lipid metabolism (Plin1, Plin4, Cidec). Few changes in gene expression were observed with sub-optimal loading or L-BAIBA alone after 24 hours. These results suggest that these signaling pathways are responsible for the synergistic effects between L-BAIBA and sub-optimal loading. Showing that a small muscle factor can enhance the effects of sub-optimal loading of bone may be of relevance for individuals unable to benefit from optimal exercise. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Matt Prideaux
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of MedicineIndiana UniversityIndianapolisINUSA
| | - Alberto Smargiassi
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of MedicineIndiana UniversityIndianapolisINUSA
| | - Gang Peng
- Indiana Center for Musculoskeletal Health, Department of Medicine and Molecular Genetics, School of MedicineIndiana UniversityIndianapolisINUSA
| | - Marco Brotto
- Bone‐Muscle Research Center, College of Nursing and Health InnovationUniversity of Texas‐ArlingtonArlingtonTXUSA
| | - Alexander G Robling
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of MedicineIndiana UniversityIndianapolisINUSA
| | - Lynda F Bonewald
- Indiana Center for Musculoskeletal Health, Department of Anatomy, Cell Biology and Physiology, School of MedicineIndiana UniversityIndianapolisINUSA
| |
Collapse
|
27
|
Zhao D, Wu J, Acosta FM, Xu H, Jiang JX. Connexin 43 hemichannels and prostaglandin E 2 release in anabolic function of the skeletal tissue to mechanical stimulation. Front Cell Dev Biol 2023; 11:1151838. [PMID: 37123401 PMCID: PMC10133519 DOI: 10.3389/fcell.2023.1151838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
Bone adapts to changes in the physical environment by modulating remodeling through bone resorption and formation to maintain optimal bone mass. As the most abundant connexin subtype in bone tissue, connexin 43 (Cx43)-forming hemichannels are highly responsive to mechanical stimulation by permitting the exchange of small molecules (<1.2 kDa) between bone cells and the extracellular environment. Upon mechanical stimulation, Cx43 hemichannels facilitate the release of prostaglandins E2 (PGE2), a vital bone anabolic factor from osteocytes. Although most bone cells are involved in mechanosensing, osteocytes are the principal mechanosensitive cells, and PGE2 biosynthesis is greatly enhanced by mechanical stimulation. Mechanical stimulation-induced PGE2 released from osteocytic Cx43 hemichannels acts as autocrine effects that promote β-catenin nuclear accumulation, Cx43 expression, gap junction function, and protects osteocytes against glucocorticoid-induced osteoporosis in cultured osteocytes. In vivo, Cx43 hemichannels with PGE2 release promote bone formation and anabolism in response to mechanical loading. This review summarizes current in vitro and in vivo understanding of Cx43 hemichannels and extracellular PGE2 release, and their roles in bone function and mechanical responses. Cx43 hemichannels could be a significant potential new therapeutic target for treating bone loss and osteoporosis.
Collapse
Affiliation(s)
- Dezhi Zhao
- School of Medicine, Northwest University, Xi’an, China
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Jiawei Wu
- School of Medicine, Northwest University, Xi’an, China
| | - Francisca M. Acosta
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| | - Huiyun Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an, China
| | - Jean X. Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, United States
| |
Collapse
|
28
|
Yan L, Liao L, Su X. Role of mechano-sensitive non-coding RNAs in bone remodeling of orthodontic tooth movement: recent advances. Prog Orthod 2022; 23:55. [PMID: 36581789 PMCID: PMC9800683 DOI: 10.1186/s40510-022-00450-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 11/15/2022] [Indexed: 12/31/2022] Open
Abstract
Orthodontic tooth movement relies on bone remodeling and periodontal tissue regeneration in response to the complicated mechanical cues on the compressive and tensive side. In general, mechanical stimulus regulates the expression of mechano-sensitive coding and non-coding genes, which in turn affects how cells are involved in bone remodeling. Growing numbers of non-coding RNAs, particularly mechano-sensitive non-coding RNA, have been verified to be essential for the regulation of osteogenesis and osteoclastogenesis and have revealed how they interact with signaling molecules to do so. This review summarizes recent findings of non-coding RNAs, including microRNAs and long non-coding RNAs, as crucial regulators of gene expression responding to mechanical stimulation, and outlines their roles in bone deposition and resorption. We focused on multiple mechano-sensitive miRNAs such as miR-21, - 29, -34, -103, -494-3p, -1246, -138-5p, -503-5p, and -3198 that play a critical role in osteogenesis function and bone resorption. The emerging roles of force-dependent regulation of lncRNAs in bone remodeling are also discussed extensively. We summarized mechano-sensitive lncRNA XIST, H19, and MALAT1 along with other lncRNAs involved in osteogenesis and osteoclastogenesis. Ultimately, we look forward to the prospects of the novel application of non-coding RNAs as potential therapeutics for tooth movement and periodontal tissue regeneration.
Collapse
Affiliation(s)
- Lichao Yan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry and Engineering Research Center of Oral Translational Medicine and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Li Liao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry and Engineering Research Center of Oral Translational Medicine and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Xiaoxia Su
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Pediatric Dentistry and Engineering Research Center of Oral Translational Medicine and National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
29
|
Chlebek C, Moore JA, Ross FP, van der Meulen MCH. Molecular Identification of Spatially Distinct Anabolic Responses to Mechanical Loading in Murine Cortical Bone. J Bone Miner Res 2022; 37:2277-2287. [PMID: 36054133 DOI: 10.1002/jbmr.4686] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 08/05/2022] [Accepted: 08/20/2022] [Indexed: 11/08/2022]
Abstract
Osteoporosis affects over 200 million women worldwide, one-third of whom are predicted to suffer from an osteoporotic fracture in their lifetime. The most promising anabolic drugs involve administration of expensive antibodies. Because mechanical loading stimulates bone formation, our current data, using a mouse model, replicates the anabolic effects of loading in humans and may identify novel pathways amenable to oral treatment. Murine tibial compression produces axially varying deformations along the cortical bone, inducing highest strains at the mid-diaphysis and lowest at the metaphyseal shell. To test the hypothesis that load-induced transcriptomic responses at different axial locations of cortical bone would vary as a function of strain magnitude, we loaded the left tibias of 10-week-old female C57Bl/6 mice in vivo in compression, with contralateral limbs as controls. Animals were euthanized at 1, 3, or 24 hours post-loading or loaded for 1 week (n = 4-5/group). Bone marrow and cancellous bone were removed, cortical bone was segmented into the metaphyseal shell, proximal diaphysis, and mid-diaphysis, and load-induced differential gene expression and enriched biological processes were examined for the three segments. At each time point, the mid-diaphysis (highest strain) had the greatest transcriptomic response. Similarly, biological processes regulating bone formation and turnover increased earlier and to the greatest extent at the mid-diaphysis. Higher strain induced greater levels of osteoblast and osteocyte genes, whereas expression was lower in osteoclasts. Among the top differentially expressed genes at 24-hours post-loading, 17 had known functions in bone biology, of which 12 were present only in osteoblasts, 3 exclusively in osteoclasts, and 2 were present in both cell types. Based on these results, we conclude that murine tibial loading induces spatially unique transcriptomic responses correlating with strain magnitude in cortical bone. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Carolyn Chlebek
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jacob A Moore
- College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | | | - Marjolein C H van der Meulen
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA.,Hospital for Special Surgery, New York, NY, USA
| |
Collapse
|
30
|
Yue Z, Nie L, Zhao P, Ji N, Liao G, Wang Q. Senescence-associated secretory phenotype and its impact on oral immune homeostasis. Front Immunol 2022; 13:1019313. [PMID: 36275775 PMCID: PMC9581398 DOI: 10.3389/fimmu.2022.1019313] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/15/2022] [Indexed: 09/09/2023] Open
Abstract
The senescence-associated secretory phenotype (SASP), which accumulates over the course of normal aging and in age-related diseases, is a crucial driver of chronic inflammation and aging phenotypes. It is also responsible for the pathogenesis of multiple oral diseases. However, the pathogenic mechanism underlying SASP has not yet been fully elucidated. Here, relevant articles on SASP published over the last five years (2017-2022) were retrieved and used for bibliometric analysis, for the first time, to examine SASP composition. More than half of the relevant articles focus on various cytokines (27.5%), growth factors (20.9%), and proteases (20.9%). In addition, lipid metabolites (13.1%) and extracellular vesicles (6.5%) have received increasing attention over the past five years, and have been recognized as novel SASP categories. Based on this, we summarize the evidences demonstrating that SASP plays a pleiotropic role in oral immunity and propose a four-step hypothetical framework for the progression of SASP-related oral pathology-1) oral SASP development, 2) SASP-related oral pathological alterations, 3) pathological changes leading to oral immune homeostasis disruption, and 4) SASP-mediated immune dysregulation escalating oral disease. By targeting specific SASP factors, potential therapies can be developed to treat oral and age-related diseases.
Collapse
Affiliation(s)
- Ziqi Yue
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Lulingxiao Nie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Pengfei Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Faculty of Dentistry, The University of Hong Kong, Sai Ying Pun, Hong Kong SAR, China
| | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ga Liao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Information Management, Department of Stomatology Informatics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
- Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Lawson LY, Migotsky N, Chermside-Scabbo CJ, Shuster JT, Joeng KS, Civitelli R, Lee B, Silva MJ. Loading-induced bone formation is mediated by Wnt1 induction in osteoblast-lineage cells. FASEB J 2022; 36:e22502. [PMID: 35969160 PMCID: PMC9430819 DOI: 10.1096/fj.202200591r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/11/2022] [Accepted: 08/02/2022] [Indexed: 11/11/2022]
Abstract
Mechanical loading on the skeleton stimulates bone formation. Although the exact mechanism underlying this process remains unknown, a growing body of evidence indicates that the Wnt signaling pathway is necessary for the skeletal response to loading. Recently, we showed that Wnts produced by osteoblast lineage cells mediate the osteo-anabolic response to tibial loading in adult mice. Here, we report that Wnt1 specifically plays a crucial role in mediating the mechano-adaptive response to loading. Independent of loading, short-term loss of Wnt1 in the Osx-lineage resulted in a decreased cortical bone area in the tibias of 5-month-old mice. In females, strain-matched loading enhanced periosteal bone formation in Wnt1F/F controls, but not in Wnt1F/F; OsxCreERT2 knockouts. In males, strain-matched loading increased periosteal bone formation in both control and knockout mice; however, the periosteal relative bone formation rate was 65% lower in Wnt1 knockouts versus controls. Together, these findings show that Wnt1 supports adult bone homeostasis and mediates the bone anabolic response to mechanical loading.
Collapse
Affiliation(s)
- Lisa Y. Lawson
- Department of Orthopaedic Surgery, Washington University School of Medicine, Saint Louis, MO, United States
- Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Nicole Migotsky
- Department of Orthopaedic Surgery, Washington University School of Medicine, Saint Louis, MO, United States
- Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Biomedical Engineering, Washington University, Saint Louis, MO, United States
| | - Christopher J. Chermside-Scabbo
- Department of Orthopaedic Surgery, Washington University School of Medicine, Saint Louis, MO, United States
- Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, USA
| | - John T. Shuster
- Department of Orthopaedic Surgery, Washington University School of Medicine, Saint Louis, MO, United States
- Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Kyu Sang Joeng
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Roberto Civitelli
- Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, Saint Louis, MO, United States
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Waco, TX, United States
| | - Matthew J. Silva
- Department of Orthopaedic Surgery, Washington University School of Medicine, Saint Louis, MO, United States
- Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Biomedical Engineering, Washington University, Saint Louis, MO, United States
| |
Collapse
|
32
|
Zhang Y, Li X, Li J, Liu D, Zhai L, Wang X, Abdurahman A, Yokota H, Zhang P. Knee Loading Enhances the Migration of Adipose-Derived Stem Cells to the Osteoarthritic Sites Through the SDF-1/CXCR4 Regulatory Axis. Calcif Tissue Int 2022; 111:171-184. [PMID: 35429248 DOI: 10.1007/s00223-022-00976-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/30/2022] [Indexed: 11/02/2022]
Abstract
Osteoarthritis (OA) is a whole joint disorder that is characterized by cartilage damage and abnormal remodeling of subchondral bone. Injecting adipose-derived stem cells (ASCs) into the knee joint cavity can assist in repairing osteoarthritic joints, but their ability to migrate to the damaged site is limited. Our previous studies have shown that knee loading can improve the symptoms of OA, but the effect and mechanism of knee loading on the migration of ASCs in OA remain unclear. We employed a mouse model of OA in the knee and applied knee loading (1 N at 5 Hz for 6 min/day for 2 weeks) after the intra-articular injection of ASCs. The cartilage and subchondral bone repair were assessed by histopathological analysis. Immunofluorescence assays were also used to analyze the migration of ASCs. Using cell cultures, we evaluated the migration of ASCs using the transwell migration and wound healing assays. In vivo experiments showed that knee loading promoted the migration of ASCs, increased the local SDF-1 level, and accelerated the repair of the OA-damaged sites. Mechanistically, the observed effects were blocked by the SDF-1/CXCR4 inhibitor. The in vitro results further revealed that knee loading promoted the migration of ASCs and the inhibition of SDF-1/CXCR4 significantly suppressed the beneficial loading effect. The results herein suggested that the migration of ASCs was enhanced by knee loading through the SDF-1/CXCR4 regulatory axis, and mechanical loading promoted the joint-protective effect of ASCs.
Collapse
Affiliation(s)
- Yifan Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China
| | - Jie Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China
| | - Daquan Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China
| | - Lidong Zhai
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Xuetong Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Abdusami Abdurahman
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University-Purdue University, Indianapolis, IN, 46202, USA
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China.
- Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, 300070, China.
- Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin Medical University, Tianjin, 300052, China.
| |
Collapse
|
33
|
Ko FC, Moran MM, Ross RD, Sumner DR. Activation of canonical Wnt signaling accelerates intramembranous bone regeneration in male mice. J Orthop Res 2022; 40:1834-1843. [PMID: 34811780 PMCID: PMC9124233 DOI: 10.1002/jor.25217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/25/2021] [Accepted: 11/09/2021] [Indexed: 02/04/2023]
Abstract
Canonical Wnt signaling plays an important role in skeletal development, homeostasis, and both endochondral and intramembranous repair. While studies have demonstrated that the inhibition of Wnt signaling impairs intramembranous bone regeneration, how its activation affects intramembranous bone regeneration has been underexplored. Therefore, we sought to determine the effects of activation of canonical Wnt signaling on intramembranous bone regeneration by using the well-established marrow ablation model. We hypothesized that mice with a mutation in the Wnt ligand coreceptor gene Lrp5 would have accelerated intramembranous bone regeneration. Male and female wild-type and Lrp5-mutant mice underwent unilateral femoral bone marrow ablation surgery in the right femur at 4 weeks of age. Both the left intact and right operated femurs were assessed at Days 3, 5, 7, 10, and 14. The intact femur of Lrp5 mutant mice of both sexes had higher bone mass than wild-type littermates, although to a greater degree in males than females. Overall, the regenerated bone volume in Lrp5 mutant male mice was 1.8-fold higher than that of littermate controls, whereas no changes were observed between female Lrp5 mutant and littermate control mice. In addition, the rate of intramembranous bone regeneration (from Day 3 to Day 7) was higher in Lrp5 mutant male mice compared to their same-sex littermate controls with no difference in the females. Thus, activation of canonical Wnt signaling increases bone mass in intact bones of both sexes, but accelerates intramembranous bone regeneration following an injury challenge only in male mice.
Collapse
Affiliation(s)
- Frank C. Ko
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612,Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612
| | - Meghan M. Moran
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612,Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612
| | - Ryan D. Ross
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612,Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612
| | - D. Rick Sumner
- Department of Anatomy & Cell Biology, Rush University Medical Center, Chicago, IL, 60612,Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612
| |
Collapse
|
34
|
Zhao D, Hua R, Riquelme MA, Cheng H, Guda T, Xu H, Gu S, Jiang JX. Osteocytes regulate bone anabolic response to mechanical loading in male mice via activation of integrin α5. Bone Res 2022; 10:49. [PMID: 35851577 PMCID: PMC9293884 DOI: 10.1038/s41413-022-00222-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/25/2022] [Accepted: 06/13/2022] [Indexed: 12/18/2022] Open
Abstract
Physical mechanical stimulation can maintain and even increase bone mass. Here, we report an important role of osteocytic integrin α5 in regulating the anabolic response of bone to mechanical loading using an Itga5 conditional gene knockout (cKO) mouse model. Integrin α5 gene deletion increased apoptotic osteocytes and reduced cortical anabolic responses to tibial compression including decreased endosteal osteoblasts and bone formation, and increased endosteal osteoclasts and bone resorption, contributing to the decreased bone area fraction and biomechanical properties, leading to an enlarged bone marrow area in cKO mice. Similar disruption of anabolic responses to mechanical loading was also detected in cKO trabecular bone. Moreover, integrin α5 deficiency impeded load-induced Cx43 hemichannel opening, and production and release of PGE2, an anabolic factor, resulting in attenuated effects of the loading on catabolic sclerostin (SOST) reduction and anabolic β-catenin increase. Together, this study shows an indispensable role of integrin α5 in osteocytes in the anabolic action of mechanical loading on skeletal tissue through activation of hemichannels and PGE2-evoked gene expression. Integrin α5 could act as a potential new therapeutic target for bone loss, especially in the elderly population with impeded mechanical sensitivity.
Collapse
Affiliation(s)
- Dezhi Zhao
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Rui Hua
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Manuel A Riquelme
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Hongyun Cheng
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Teja Guda
- Department of Biomedical Engineering and Chemical Engineering, University of Texas, San Antonio, TX, USA
| | - Huiyun Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Sumin Gu
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
35
|
Kroon T, Bhadouria N, Niziolek P, Edwards D, Choi R, Clinkenbeard EL, Robling A, Holguin N. Suppression of Sost/Sclerostin and Dickkopf-1 Augment Intervertebral Disc Structure in Mice. J Bone Miner Res 2022; 37:1156-1169. [PMID: 35278242 PMCID: PMC9320845 DOI: 10.1002/jbmr.4546] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 03/02/2022] [Accepted: 03/10/2022] [Indexed: 11/06/2022]
Abstract
Intervertebral disc (IVD) degeneration is a leading cause of low back pain, characterized by accelerated extracellular matrix breakdown and IVD height loss, but there is no approved pharmacological therapeutic. Deletion of Wnt ligand competitor Lrp5 induces IVD degeneration, suggesting that Wnt signaling is essential for IVD homeostasis. Therefore, the IVD may respond to neutralization of Wnt ligand competitors sost(gene)/sclerostin(protein) and/or dickkopf-1 (dkk1). Anti-sclerostin antibody (scl-Ab) is an FDA-approved bone therapeutic that activates Wnt signaling. We aimed to (i) determine if pharmacological neutralization of sclerostin, dkk1, or their combination would stimulate Wnt signaling and augment IVD structure and (ii) determine the prolonged adaptation of the IVD to global, persistent deletion of sost. Nine-week-old C57Bl/6J female mice (n = 6-7/group) were subcutaneously injected 2×/week for 5.5 weeks with scl-Ab (25 mg/kg), dkk1-Ab (25 mg/kg), 3:1 scl-Ab/dkk1-Ab (18.75:6.25 mg/kg), or vehicle (veh). Separately, IVD of sost KO and wild-type (WT) mice (n = 8/group) were harvested at 16 weeks of age. First, compared with vehicle, injection of scl-Ab, dkk1-Ab, and 3:1 scl-Ab/dkk1-Ab similarly increased lumbar IVD height and β-catenin gene expression. Despite these similarities, only injection of scl-Ab alone strengthened IVD mechanical properties and decreased heat shock protein gene expressions. Genetically and compared with WT, sost KO enlarged IVD height, increased proteoglycan staining, and imbibed IVD hydration. Notably, persistent deletion of sost was compensated by upregulation of dkk1, which consequently reduced the cell nuclear fraction for Wnt signaling co-transcription factor β-catenin in the IVD. Lastly, RNA-sequencing pathway analysis confirmed the compensatory suppression of Wnt signaling and revealed a reduction of cellular stress-related pathways. Together, suppression of sost/sclerostin or dkk1 each augmented IVD structure by stimulating Wnt signaling, but scl-Ab outperformed dkk1-Ab in strengthening the IVD. Ultimately, postmenopausal women prescribed scl-Ab injections to prevent vertebral fracture may also benefit from a restoration of IVD height and health. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Tori Kroon
- Department of Biomedical EngineeringIUPUIIndianapolisINUSA
| | - Neharika Bhadouria
- Department of Mechanical EngineeringPurdue UniversityWest LafayetteINUSA
| | | | - Daniel Edwards
- Indiana Center of Musculoskeletal HealthIndianapolisINUSA
| | - Roy Choi
- Department for Anatomy and Cell BiologyIUPUIIndianapolisINUSA
| | | | - Alexander Robling
- Indiana Center of Musculoskeletal HealthIndianapolisINUSA
- Department for Anatomy and Cell BiologyIUPUIIndianapolisINUSA
| | - Nilsson Holguin
- Indiana Center of Musculoskeletal HealthIndianapolisINUSA
- Department for Anatomy and Cell BiologyIUPUIIndianapolisINUSA
- Department of Mechanical and Energy EngineeringIUPUIIndianapolisINUSA
| |
Collapse
|
36
|
Rooney AM, Ayobami OO, Kelly NH, Schimenti JC, Ross FP, van der Meulen MCH. Bone mass and adaptation to mechanical loading are sexually dimorphic in adult osteoblast-specific ERα knockout mice. Bone 2022; 158:116349. [PMID: 35123146 DOI: 10.1016/j.bone.2022.116349] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/20/2022] [Accepted: 01/31/2022] [Indexed: 12/14/2022]
Abstract
Estrogen receptor-alpha (ERα) regulates bone mass and is implicated in bone tissue's response to mechanical loading. The effects of ERα deletion in mice depend on sex, anatomical location, and the cellular stage at which ERα is removed. Few studies have investigated the effect of age on the role of ERα in skeletal maintenance and functional adaptation. We previously demonstrated that bone mass and adaptation to loading were altered in growing 10-week-old female and male mice lacking ERα in mature osteoblasts and osteocytes (pOC-ERαKO). Here our goal was to determine the effects of ERα and mechanical loading in skeletally-mature adult mice. We subjected 26-week-old skeletally-mature adult pOC-ERαKO and littermate control (LC) mice of both sexes to two weeks of in vivo cyclic tibial loading. ERα deletion in male mice did not alter bone mass or the response to loading. Adult female pOC-ERαKO mice had reduced cancellous and cortical bone mass and increased adaptation to high-magnitude mechanical loading compared to LC mice. Thus, ERα deletion from mature osteoblasts reduced the bone mass and increased the mechanoadaptation of adult female but not male mice. Additionally, compared to our previous work in young mice, adult female mice had greatly reduced mechanoadaptation and adult male mice retained most of their mechanoadaptation with age.
Collapse
Affiliation(s)
- Amanda M Rooney
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - Olufunmilayo O Ayobami
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - Natalie H Kelly
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA.
| | - John C Schimenti
- College of Veterinary Medicine, Cornell University, Ithaca 14853, NY, USA.
| | - F Patrick Ross
- Research Division, Hospital for Special Surgery, New York, NY 10021, USA.
| | - Marjolein C H van der Meulen
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA; Research Division, Hospital for Special Surgery, New York, NY 10021, USA.
| |
Collapse
|
37
|
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: 78] [Impact Index Per Article: 26.0] [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
|
38
|
Abdurahman A, Li X, Li J, Liu D, Zhai L, Wang X, Zhang Y, Meng Y, Yokota H, Zhang P. Loading-driven PI3K/Akt signaling and erythropoiesis enhanced angiogenesis and osteogenesis in a postmenopausal osteoporosis mouse model. Bone 2022; 157:116346. [PMID: 35114427 DOI: 10.1016/j.bone.2022.116346] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/21/2022]
Abstract
Bone vasculature influences osteogenesis and haematopoiesis in the bone microenviroment. Mechanical loading has been shown to stimulate the formation of osteogenesis-related type H vessels in an ovariectomy (OVX)-induced osteoporosis mouse model. To determine the loading-driven mechanism of angiogenesis and the formation of type H vessels in bone, we evaluated the roles of PI3K/Akt signaling and erythropoiesis in the bone marrow. The daily application of mechanical loading (1 N at 5 Hz for 6 min/day) for 2 weeks on OVX mice inhibited osteoclast activity, associated with an increase in the number of osteoblasts and trabecular volume ratio. Mechanical loading enhanced bone vasculature and vessel formation, as well as PI3K/Akt phosphorylation and erythropoiesis in the bone marrow. Notably, LY294002, an inhibitor of PI3K signaling, blocked the tube formation by endothelial progenitor cells, as well as their migration and wound healing. The conditioned medium, derived from erythroblasts, also promoted the function of HUVECs with elevated levels of VEGF, CD31, and Emcn. Collectively, this study demonstrates that mechanical loading prevents osteoporotic bone loss by promoting angiogenesis and type H vessel formation. This load-driven preventing effect is in part mediated by PI3K/Akt signaling and erythropoiesis in the bone marrow.
Collapse
Affiliation(s)
- Abdusami Abdurahman
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300070, China
| | - Jie Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300070, China
| | - Daquan Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300070, China
| | - Lidong Zhai
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xuetong Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yifan Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yao Meng
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University Purdue University Indianapolis, IN 46202, USA
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300070, China; Tianjin Key Laboratory of Spine and Spinal Cord, Tianjin Medical University, Tianjin 300052, China.
| |
Collapse
|
39
|
Abstract
This is a review of evidence and practical tips on exercise for individuals with osteoporosis, including individuals with hip and vertebral fractures. Balance and functional training, with or without strength training, can prevent falls. Several types of exercise can improve outcomes that are important to patients, such as physical functioning or quality of life. Individuals with osteoporosis should prioritize balance, functional and resistance training ≥ twice weekly, where exercises, volume, intensity, and progression are aligned with the patient's goals and abilities. Patients who want to participate in other activities (e.g., walking, impact exercise, yoga, Pilates) can do them in addition to, but not instead of, balance and functional or strength training, if they can be done safely or modified. Avoid generic advice like "Don't bend or twist", which is difficult or impossible to operationalize, and may create fear and activity avoidance. Instead, be specific about the types of activities to avoid or modify, and provide tips on how to make daily activities safer, or signpost to resources from national osteoporosis societies. For example, not all bending or twisting is bad; it is activities that involve rapid, repetitive, sustained, weighted, or end range of motion twisting or flexion of the spine that may need to be modified, especially in individuals at high risk of fracture.
Collapse
Affiliation(s)
- L M Giangregorio
- Department of Kinesiology and Health Sciences, University of Waterloo, 200 University Ave W, Waterloo, Ontario, N2K 2N1, Canada; Schlegel-UW Research Institute for Aging, Waterloo, Ontario, Canada.
| | - Matteo Ponzano
- Department of Kinesiology and Health Sciences, University of Waterloo, 200 University Ave W, Waterloo, Ontario, N2K 2N1, Canada
| |
Collapse
|
40
|
McGregor NE, Walker EC, Chan AS, Poulton IJ, Cho EHJ, Windahl SH, Sims NA. STAT3 Hyperactivation Due to SOCS3 Deletion in Murine Osteocytes Accentuates Responses to Exercise- and Load-Induced Bone Formation. J Bone Miner Res 2022; 37:547-558. [PMID: 34870348 DOI: 10.1002/jbmr.4484] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/07/2021] [Accepted: 11/27/2021] [Indexed: 12/16/2022]
Abstract
Cortical bone develops and changes in response to mechanical load, which is sensed by bone-embedded osteocytes. The bone formation response to load depends on STAT3 intracellular signals, which are upregulated after loading and are subject to negative feedback from Suppressor of Cytokine Signaling 3 (Socs3). Mice with Dmp1Cre-targeted knockout of Socs3 have elevated STAT3 signaling in osteocytes and display delayed cortical bone maturation characterized by impaired accrual of high-density lamellar bone. This study aimed to determine whether these mice exhibit an altered response to mechanical load. The approach used was to test both treadmill running and tibial compression in female Dmp1Cre.Socs3f/f mice. Treadmill running for 5 days per week from 6 to 11 weeks of age did not change cortical bone mass in control mice, but further delayed cortical bone maturation in Dmp1Cre.Socs3f/f mice; accrual of high-density bone was suppressed, and cortical thickness was less than in genetically-matched sedentary controls. When strain-matched anabolic tibial loading was tested, both control and Dmp1Cre.Socs3f/f mice exhibited a significantly greater cortical thickness and periosteal perimeter in loaded tibia compared with the contralateral non-loaded bone. At the site of greatest compressive strain, the loaded Dmp1Cre.Socs3f/f tibias showed a significantly greater response than controls, indicated by a greater increase in cortical thickness. This was due to a greater bone formation response on both periosteal and endocortical surfaces, including formation of abundant woven bone on the periosteum. This suggests a greater sensitivity to mechanical load in Dmp1Cre.Socs3f/f bone. In summary, mice with targeted SOCS3 deletion and immature cortical bone have an exaggerated response to both physiological and experimental mechanical loads. We conclude that there is an optimal level of osteocytic response to mechanical load required for cortical bone maturation and that load-induced bone formation may be increased by augmenting STAT3 signaling within osteocytes. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
| | - Emma C Walker
- St. Vincent's Institute of Medical Research, Fitzroy, Australia
| | - Audrey Sm Chan
- Centre for Muscle Research, The University of Melbourne, Melbourne, Australia
| | | | - Ellie H-J Cho
- Biological Optical Microscopy Platform, The University of Melbourne, Melbourne, Australia
| | - Sara H Windahl
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Huddinge, Sweden
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Australia.,Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, Australia
| |
Collapse
|
41
|
Liu P, Tu J, Wang W, Li Z, Li Y, Yu X, Zhang Z. Effects of Mechanical Stress Stimulation on Function and Expression Mechanism of Osteoblasts. Front Bioeng Biotechnol 2022; 10:830722. [PMID: 35252138 PMCID: PMC8893233 DOI: 10.3389/fbioe.2022.830722] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Osteoclasts and osteoblasts play a major role in bone tissue homeostasis. The homeostasis and integrity of bone tissue are maintained by ensuring a balance between osteoclastic and osteogenic activities. The remodeling of bone tissue is a continuous ongoing process. Osteoclasts mainly play a role in bone resorption, whereas osteoblasts are mainly involved in bone remodeling processes, such as bone cell formation, mineralization, and secretion. These cell types balance and restrict each other to maintain bone tissue metabolism. Bone tissue is very sensitive to mechanical stress stimulation. Unloading and loading of mechanical stress are closely related to the differentiation and formation of osteoclasts and bone resorption function as well as the differentiation and formation of osteoblasts and bone formation function. Consequently, mechanical stress exerts an important influence on the bone microenvironment and bone metabolism. This review focuses on the effects of different forms of mechanical stress stimulation (including gravity, continuously compressive pressure, tensile strain, and fluid shear stress) on osteoclast and osteoblast function and expression mechanism. This article highlights the involvement of osteoclasts and osteoblasts in activating different mechanical transduction pathways and reports changings in their differentiation, formation, and functional mechanism induced by the application of different types of mechanical stress to bone tissue. This review could provide new ideas for further microscopic studies of bone health, disease, and tissue damage reconstruction.
Collapse
Affiliation(s)
- Pan Liu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Ji Tu
- Spine Labs, St. George & Sutherland Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Wenzhao Wang
- Department of Orthopedics, West China Hospital of Sichuan University, Chengdu, China
| | - Zheng Li
- People’s Hospital of Jiulongpo District, Chongqing, China
| | - Yao Li
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Xiaoping Yu
- School of Public Health, Chengdu Medical College, Chengdu, China
- Basic Medical College of Chengdu University, Chengdu, China
- *Correspondence: Xiaoping Yu, ; Zhengdong Zhang,
| | - Zhengdong Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, China
- The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- Department of Orthopedics, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
- *Correspondence: Xiaoping Yu, ; Zhengdong Zhang,
| |
Collapse
|
42
|
Gardinier JD, Chougule A, Zhang C. The mechanotransduction of MLO-Y4 cells is disrupted by the senescence-associated secretory phenotype of neighboring cells. J Cell Physiol 2022; 237:2249-2257. [PMID: 35102547 PMCID: PMC9052359 DOI: 10.1002/jcp.30690] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 01/14/2022] [Accepted: 01/19/2022] [Indexed: 01/10/2023]
Abstract
Age-related bone loss is attributed to the accumulation of senescent cells and their increasing production of inflammatory cytokines as part of the senescence-associated secretory phenotype (SASP). In otherwise healthy individuals, osteocytes play a key role in maintaining bone mass through their primary function of responding to skeletal loading. Given that osteocytes' response to loading is known to steadily decline with age, we hypothesized that the increasing presence of senescent cells and their SASP inhibit osteocytes' response to loading. To test this hypothesis, we developed two in vitro models of senescent osteocytes and osteoblasts derived from MLO-Y4 and MC3T3 cell lines, respectively. The senescent phenotype was unique to each cell type based on distinct changes in cell cycle inhibitors and SASP profile. The SASP profile of senescent osteocytes was in part dependent on nuclear factor-κB signaling and presents a new potential mechanism to target the SASP in bone. Nonsenescent MLO-Y4 cells cultured with the SASP of each senescent cell type failed to exhibit changes in gene expression as well as ERK phosphorylation and prostaglandin E2 release. The SASP of senescent osteocytes had the largest effect and neutralizing interleukin-6 (IL-6) as part of the SASP restored osteocytes' response to loading. The loss in mechanotransduction due to IL-6 was attributed to a decrease in P2X7 expression and overall sensitivity to purinergic signaling. Altogether, these findings demonstrate that the SASP of senescent cells have a negative effect on the mechanotransduction of osteocytes and that IL-6 is a key SASP component that contributes to the loss in mechanotransduction.
Collapse
Affiliation(s)
- Joseph D Gardinier
- Bone and Joint Center, Henry Ford Health System, Henry Ford Hospital, Detroit, Michigan, USA
| | - Amit Chougule
- Bone and Joint Center, Henry Ford Health System, Henry Ford Hospital, Detroit, Michigan, USA
| | - Chunbin Zhang
- Bone and Joint Center, Henry Ford Health System, Henry Ford Hospital, Detroit, Michigan, USA
| |
Collapse
|
43
|
Martínez-Gil N, Ugartondo N, Grinberg D, Balcells S. Wnt Pathway Extracellular Components and Their Essential Roles in Bone Homeostasis. Genes (Basel) 2022; 13:genes13010138. [PMID: 35052478 PMCID: PMC8775112 DOI: 10.3390/genes13010138] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 12/11/2022] Open
Abstract
The Wnt pathway is involved in several processes essential for bone development and homeostasis. For proper functioning, the Wnt pathway is tightly regulated by numerous extracellular elements that act by both activating and inhibiting the pathway at different moments. This review aims to describe, summarize and update the findings regarding the extracellular modulators of the Wnt pathway, including co-receptors, ligands and inhibitors, in relation to bone homeostasis, with an emphasis on the animal models generated, the diseases associated with each gene and the bone processes in which each member is involved. The precise knowledge of all these elements will help us to identify possible targets that can be used as a therapeutic target for the treatment of bone diseases such as osteoporosis.
Collapse
|
44
|
Nagu P, Sharma V, Behl T, Pathan AKA, Mehta V. Molecular Insights to the Wnt Signaling During Alzheimer's Disorder: a Potential Target for Therapeutic Interventions. J Mol Neurosci 2022; 72:679-690. [PMID: 34997460 DOI: 10.1007/s12031-021-01940-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/30/2021] [Indexed: 11/25/2022]
Abstract
In the adult brain, Wnt signaling is crucial for neurogenesis, and it also regulates neuronal development, neuronal maturation, neuronal differential, and proliferation. Impaired Wnt signaling pathways are associated with enhanced levels of amyloid-β, reduced β-catenin levels, and increased expression of GSK-3β enzyme, suggesting its direct association with the pathogenesis of Alzheimer's disorder (AD). These findings are consolidated by reports where activation of Wnt signaling by genetic factors and pharmacological intervention has improved the cognitive functions in animals and restored neurogenesis in the adult brain. Various natural and synthetic molecules have been identified that modulate Wnt signaling in the adult brain and promote neurogenesis and alleviate behavioral dysfunction. These molecules include lithium, valproic acid, ethosuximide, selenomethionine, curcumin, andrographolide, xanthoceraside, huperzine A, pyridostigmine, ginkgolide-B, ricinine, cannabidiol, and resveratrol. These molecules are associated with the DKK1 and GSK-3β inhibition and β-catenin stabilization along with their effects on neurogenesis, neuronal proliferation, and differentiation in the hippocampus through modulation of Wnt signaling and thereby could prove beneficial in the management of AD pathogenesis. Although modulation of the Wnt signaling seems to suggest to be promising in the management of AD, unfortunately, most of the literature available for the association of Wnt signaling and AD pathogenesis is either from preclinical studies or post-mortem brain. Therefore, it will be interesting to understand the role of Wnt signaling in AD patients, and a rigorous investigation could provide us with a better understanding of AD pathogenesis and the identification of novel targets for therapeutic interventions.
Collapse
Affiliation(s)
- Priyanka Nagu
- Department of Pharmacy, Shri Jagdishprasad Jhabarmal Tibrewala University, Jhunjhunu, Rajasthan, India.,Department of Pharmaceutics, Government College of Pharmacy, Rohru, Himachal Pradesh, India
| | - Vivek Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.,Department of Pharmacology, Government College of Pharmacy, Himachal Pradesh 171207, Rohru, District Shimla, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Amjad Khan A Pathan
- Department of Pharmacy, Shri Jagdishprasad Jhabarmal Tibrewala University, Jhunjhunu, Rajasthan, India
| | - Vineet Mehta
- Department of Pharmacology, Government College of Pharmacy, Himachal Pradesh 171207, Rohru, District Shimla, India.
| |
Collapse
|
45
|
Takata T, Matsumura M. The LINC Complex Assists the Nuclear Import of Mechanosensitive Transcriptional Regulators. Results Probl Cell Differ 2022; 70:315-337. [PMID: 36348113 DOI: 10.1007/978-3-031-06573-6_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Mechanical forces play pivotal roles in directing cell functions and fate. To elicit gene expression, either intrinsic or extrinsic mechanical information are transmitted into the nucleus beyond the nuclear envelope via at least two distinct pathways, possibly more. The first and well-known pathway utilizes the canonical nuclear transport of mechanoresponsive transcriptional regulators through the nuclear pore complex, which is an exclusive route for macromolecular trafficking between the cytoplasm and nucleoplasm. The second pathway depends on the linker of the nucleoskeleton and cytoskeleton (LINC) complex, which is a molecular bridge traversing the nuclear envelope between the cytoskeleton and nucleoskeleton. This protein complex is a central component in mechanotransduction at the nuclear envelope that transmits mechanical information from the cytoskeleton into the nucleus to influence the nuclear structure, nuclear stiffness, chromatin organization, and gene expression. Besides the mechanical force transducing function, recent increasing evidence shows that the LINC complex plays a role in controlling nucleocytoplasmic transport of mechanoresponsive transcriptional regulators. Here we discuss recent findings regarding the contribution of the LINC complex to the regulation of intracellular localization of the most-notable mechanosensitive transcriptional regulators, β-catenin, YAP, and TAZ.
Collapse
Affiliation(s)
- Tomoyo Takata
- Ehime Prefectural University of Health Sciences, Tobe, Ehime, Japan
| | - Miki Matsumura
- Ehime Prefectural University of Health Sciences, Tobe, Ehime, Japan.
| |
Collapse
|
46
|
Lawson LY, Brodt MD, Migotsky N, Chermside-Scabbo CJ, Palaniappan R, Silva MJ. Osteoblast-Specific Wnt Secretion Is Required for Skeletal Homeostasis and Loading-Induced Bone Formation in Adult Mice. J Bone Miner Res 2022; 37:108-120. [PMID: 34542191 PMCID: PMC8770559 DOI: 10.1002/jbmr.4445] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/10/2021] [Accepted: 08/28/2021] [Indexed: 01/03/2023]
Abstract
Wnt signaling is critical to many aspects of skeletal regulation, but the importance of Wnt ligands in the bone anabolic response to mechanical loading is not well established. Recent transcriptome profiling studies by our laboratory and others show that mechanical loading potently induces genes encoding Wnt ligands, including Wnt1 and Wnt7b. Based on these findings, we hypothesized that mechanical loading stimulates adult bone formation by inducing Wnt ligand expression. To test this hypothesis, we inhibited Wnt ligand secretion in adult (5 months old) mice using a systemic (drug) and a bone-targeted (conditional gene knockout) approach, and subjected them to axial tibial loading to induce lamellar bone formation. Mice treated with the Wnt secretion inhibitor WNT974 exhibited a decrease in bone formation in non-loaded bones as well as a 54% decline in the periosteal bone formation response to tibial loading. Next, osteoblast-specific Wnt secretion was inhibited by dosing 5-month-old Osx-CreERT2; WlsF/F mice with tamoxifen. Within 1 to 2 weeks of Wls deletion, skeletal homeostasis was altered with decreased bone formation and increased resorption, and the anabolic response to loading was reduced 65% compared to control (WlsF/F ). Together, these findings show that Wnt ligand secretion is required for adult bone homeostasis, and furthermore establish a role for osteoblast-derived Wnts in mediating the bone anabolic response to tibial loading. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Lisa Y. Lawson
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Michael D. Brodt
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Nicole Migotsky
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Biomedical Engineering, Washington University, Saint Louis, MO, United States
| | - Christopher J. Chermside-Scabbo
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
- Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Ramya Palaniappan
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
| | - Matthew J. Silva
- Department of Orthopaedic Surgery and Musculoskeletal Research Center, Washington University School of Medicine, Saint Louis, MO, United States
- Department of Biomedical Engineering, Washington University, Saint Louis, MO, United States
| |
Collapse
|
47
|
Aguirre JI, Castillo EJ, Kimmel DB. Biologic and pathologic aspects of osteocytes in the setting of medication-related osteonecrosis of the jaw (MRONJ). Bone 2021; 153:116168. [PMID: 34487892 PMCID: PMC8478908 DOI: 10.1016/j.bone.2021.116168] [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: 06/08/2021] [Revised: 08/02/2021] [Accepted: 08/31/2021] [Indexed: 02/08/2023]
Abstract
Medication-related osteonecrosis of the jaw (MRONJ) is a potentially severe, debilitating condition affecting patients with cancer and patients with osteoporosis who have been treated with powerful antiresorptives (pARs) or angiogenesis inhibitors (AgIs). Oral risk factors associated with the development of MRONJ include tooth extraction and inflammatory dental disease (e.g., periodontitis, periapical infection). In bone tissues, osteocytes play a bidirectional role in which they not only act as the "receiver" of systemic signals from blood vessels, such as hormones and drugs, or local signals from the mineralized matrix as it is deformed, but they also play a critical role as "transmitter" of signals to the cells that execute bone modeling and remodeling (osteoclasts, osteoblasts and lining cells). When the survival capacity of osteocytes is overwhelmed, they can die. Osteocyte death has been associated with several pathological conditions. Whereas the causes and mechanisms of osteocyte death have been studied in conditions like osteonecrosis of the femoral head (ONFH), few studies of the causes and mechanisms of osteocyte death have been done in MRONJ. The three forms of cell death that affect most of the different cells in the body (apoptosis, autophagy, and necrosis) have been recognized in osteocytes. Notably, necroptosis, a form of regulated cell death with "a necrotic cell death phenotype," has also been identified as a form of cell death in osteocytes under certain pathologic conditions. Improving the understanding of osteocyte death in MRONJ may be critical for preventing disease and developing treatment approaches. In this review, we intend to provide insight into the biology of osteocytes, cell death, in general, and osteocyte death, in particular, and discuss hypothetical mechanisms involved in osteocyte death associated with MRONJ.
Collapse
Affiliation(s)
- J I Aguirre
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - E J Castillo
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America.
| | - D B Kimmel
- Department of Physiological Sciences, University of Florida (UF), Gainesville, FL, United States of America
| |
Collapse
|
48
|
Choi RB, Robling AG. The Wnt pathway: An important control mechanism in bone's response to mechanical loading. Bone 2021; 153:116087. [PMID: 34271473 PMCID: PMC8478810 DOI: 10.1016/j.bone.2021.116087] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/01/2021] [Accepted: 06/21/2021] [Indexed: 10/25/2022]
Abstract
The conversion of mechanical energy into biochemical changes within living cells is process known as mechanotransduction. Bone is a quintessential tissue for studying the molecular mechanisms of mechanotransduction, as the skeleton's mechanical competence is crucial for vertebrate movement. Bone cell mechanotransduction is facilitated by a number of cell biological pathways, one of the most prominent of which is the Wnt signaling cascade. The Wnt co-receptor Lrp5 has been identified as a crucial protein for mechanical signaling in bone, and modifiers of Lrp5 activity play important roles in mediating signaling efficiency through Lrp5, including sclerostin, Dkk1, and the co-receptor Lrp4. Mechanical regulation of sclerostin is mediated by certain members of the Hdac family. Other mechanisms that influence Wnt signaling-some of which are mechanoresponsive-are coming to light, including R-spondins and their role in organizing the Rnf43/Znrf3 and Lgr4/5/6 complex that liberates Lrp5. While the identity of the key Wnt proteins involved in bone cell mechanical signaling are elusive, the likely pool of key players is narrowing. Identification of Wnt-based molecular targets that can be modulated pharmacologically to make mechanical stimulation (e.g., exercise) more beneficial is an emerging approach to improving skeletal integrity and reducing fracture risk.
Collapse
Affiliation(s)
- Roy B Choi
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Alexander G Robling
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Biomedical Engineering, Indiana University-Purdue University at Indianapolis, Indianapolis, IN, USA; Richard L. Roudebush Veterans Affairs Medical Center, Indianapolis, IN, USA; Indiana Center for Musculoskeletal Health, Indianapolis, IN, USA.
| |
Collapse
|
49
|
Nokhbatolfoghahaei H, Rad MR, Paknejad Z, Ardeshirylajimi A, Khojasteh A. Identification osteogenic signaling pathways following mechanical stimulation: A systematic review. Curr Stem Cell Res Ther 2021; 17:772-792. [PMID: 34615453 DOI: 10.2174/1574888x16666211006105915] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 07/17/2021] [Accepted: 07/26/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION It has been shown that mechanical forces can induce or promote osteogenic differentiation as well as remodeling of the new created bone tissues. To apply this characteristic in bone tissue engineering, it is important to know which mechanical stimuli through which signaling pathway has a more significant impact on osteogenesis. METHODS In this systematic study, an electronic search was conducted using PubMed and Google Scholar databases. This study has been prepared and organized according to the preferred reporting items for systematic reviews and meta-analyses (PRISMA) guidelines. Included studies were first categorized according to the in vivo and in vitro studies. RESULTS Six types of mechanical stresses were used in these articles and the most commonly used mechanical force and cell source were tension and bone marrow-derived mesenchymal stem cells (BMMSCs), respectively. These forces were able to trigger twelve signaling pathways in which Wnt pathway was so prominent. CONCLUSION 1) Although specific signaling pathways are induced through specific mechanical forces, Wnt signaling pathways are predominantly activated by almost all types of force/stimulation, 2) All signaling pathways regulate expression of RUNX2, which is known as a master regulator of osteogenesis, 3) In Tension force, the mode of force administration, i.e, continuous or non-continuous tension is more important than the percentage of elongation.
Collapse
Affiliation(s)
- Hanieh Nokhbatolfoghahaei
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran. Iran
| | - Maryam Rezai Rad
- Dental Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran. Iran
| | - Zahrasadat Paknejad
- Medical nanotechnology and tissue engineering research Center, Shahid Beheshti University of Medical Sciences, Tehran. Iran
| | - Abdolreza Ardeshirylajimi
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran. Iran
| | - Arash Khojasteh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran. Iran
| |
Collapse
|
50
|
Jackson E, Lara-Castillo N, Akhter MP, Dallas M, Scott JM, Ganesh T, Johnson ML. Osteocyte Wnt/β-catenin pathway activation upon mechanical loading is altered in ovariectomized mice. Bone Rep 2021; 15:101129. [PMID: 34584905 PMCID: PMC8455641 DOI: 10.1016/j.bonr.2021.101129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/09/2021] [Indexed: 11/23/2022] Open
Abstract
Estrogen levels decline in both sexes with age, but more dramatically in females. Activation of the Wnt/β-catenin signaling pathway is central to the regulation of bone mass accrual and maintenance and in response to mechanical loading. Using the ovariectomized mouse model we examined the effect of estrogen loss on the osteocyte's ability to activate the Wnt/β-catenin pathway following mechanical loading. Female TOPGAL mice underwent ovariectomy (OVX) (n = 10) or sham surgery (n = 10) at 16 weeks of age. Four weeks post-surgery, a single loading session (global strain of 2200 με for 100 cycles at 2 Hz) was performed on the right forearm with the left as a non-loaded control. Mice (n = 5) were sacrificed at 1 or 24 hr post-load. Ulnae were stained for β-catenin activation, femurs were used for μCT and 3-pt bending/biomechanical testing, and tibiae were used for histology analysis and to determine osteocyte lacunar size using SEM and high resolution micro-XCT. A 2.2-fold increase in β-catenin signaling activation was observed 24 hr post-load in the Sham group but did not occur in the OVX group. The OVX group versus control had significant losses (p < 0.05) in trabecular BMD (−8%), BV/TV (−35%) and thickness (−23%), along with cortical thickness (−6%) and periosteal perimeter (−4%). The OVX group had significantly higher trabecular bone osteoclast numbers (63%), OCS/BS (77%) and N.OC/BPm (94%) and a significant decrease in osteoblast number (53%), OBS/BS (37%) and N.OB/BPm (40%) compared to the sham group (p < 0.05). Cortical bone lacunar number/lacunar volume and bone biomechanical properties did not change between groups. Given that the ulna is a cortical bone loading model and the lack of changes in osteocyte lacunar number/volume in cortical bone, which would alter strains experienced by osteocytes, these data suggest the absence of estrogen resulted in intrinsic changes in the ability of the osteocyte to respond to mechanical load, rather than changes in the biomechanical and architectural properties of bone. In vivo mechanical loading activates β-catenin signaling in osteocytes. Ovariectomy induced estrogen loss attenuates in vivo loading induced β-catenin signaling in osteocytes. Changes in bone material and architectural properties do not appear to explain attenuated pathway activation. Our data suggests estrogen loss alters the intrinsic ability of the osteocyte to respond to mechanical load.
Collapse
Affiliation(s)
- Erica Jackson
- UMKC, School of Dentistry, Kansas City, MO 64108, United States of America
| | | | - Mohammed P. Akhter
- Creighton University, Osteoporosis Research Center, Omaha, NE 68122, United States of America
| | - Mark Dallas
- UMKC, School of Dentistry, Kansas City, MO 64108, United States of America
| | - JoAnna M. Scott
- UMKC, School of Dentistry, Kansas City, MO 64108, United States of America
| | - Thiagarajan Ganesh
- UMKC, School of Computing and Engineering, Kansas City, MO 64110, United States of America
| | - Mark L. Johnson
- UMKC, School of Dentistry, Kansas City, MO 64108, United States of America
- Corresponding author.
| |
Collapse
|