1
|
Yang L, Li Q, Wang S, Ji Y, Ma X, Qin M, Gao Y, Yang Y. Sirtuin 3-activated superoxide dismutase 2 mediates fluoride-induced osteoblastic differentiation in vitro and in vivo by down-regulating reactive oxygen species. Arch Toxicol 2024; 98:3351-3363. [PMID: 39012504 DOI: 10.1007/s00204-024-03819-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 07/04/2024] [Indexed: 07/17/2024]
Abstract
Skeletal fluorosis is a chronic metabolic bone disease caused by long-term excessive fluoride intake. Abnormal differentiation of osteoblasts plays an important role in disease progression. Research on the mechanism of fluoride-mediated bone differentiation is necessary for the prevention and treatment of skeletal fluorosis. In the present study, a rat model of fluorosis was established by exposing it to drinking water containing 50 mg/L F-. We found that fluoride promoted Runt-related transcription factor 2 (RUNX2) as well as superoxide dismutase 2 (SOD2) and sirtuin 3 (SIRT3) expression in osteoblasts of rat bone tissue. In vitro, we also found that 4 mg/L sodium fluoride promoted osteogenesis-related indicators as well as SOD2 and SIRT3 expression in MG-63 and Saos-2 cells. In addition, we unexpectedly discovered that fluoride suppressed the levels of reactive oxygen species (ROS) and mitochondrial reactive oxygen species (mtROS) in osteoblasts. When SOD2 or SIRT3 was inhibited in MG-63 cells, fluoride-decreased ROS and mtROS were alleviated, which in turn inhibited fluoride-promoted osteogenic differentiation. In conclusion, our results suggest that SIRT3/SOD2 mediates fluoride-promoted osteoblastic differentiation by down-regulating reactive oxygen species.
Collapse
Affiliation(s)
- Liu Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Qiao Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Sa Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Yi Ji
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Xinbo Ma
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Ming Qin
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China
| | - Yanhui Gao
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| | - Yanmei Yang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health (23618504), Harbin Medical University, Harbin, 150081, Heilongjiang Province, China.
| |
Collapse
|
2
|
Takahashi R, Nojiri H, Ohara Y, Fujiwara T, Ishijima M. Decreased grip strength is associated with paraspinal muscular oxidative stress in female lumbar degenerative disease patients. J Orthop Res 2024; 42:2287-2295. [PMID: 38650087 DOI: 10.1002/jor.25863] [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: 04/30/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/25/2024]
Abstract
We aimed to investigate the relationship between superoxide dismutase 2-related oxidative stress in the paraspinal muscles and spinal alignment, clinical skeletal muscle parameters, and mitochondrial function. Multifidus muscle samples from patients who underwent posterior lumbar surgery were analyzed. Patients with diseases affecting oxidative stress and spinal alignment were excluded. The superoxide dismutase 2 redox index was defined as the ratio of reactive oxygen species (superoxide) to antioxidant enzymes (superoxide dismutase 2) and was used as an index of oxidative stress. Patients were divided into two groups based on the superoxide dismutase 2 redox index. Spinal alignment, clinical skeletal muscle parameters, and succinic dehydrogenase (SDH) mean grayscale value were compared between the groups, with analyzes for both sexes. Multiple regression analyzes were used to adjust for the confounding effect of age on variables showing a significant difference between the two groups. Thirty-five patients with lumbar degenerative diseases were included. No significant differences were observed between the two groups for any of the parameters in males; however, females with a higher superoxide dismutase 2 redox index had greater lumbar lordosis, lower grip strength, and higher SDH mean grayscale value than those with a lower index. Multiple regression analyzes revealed that the superoxide dismutase 2 redox index was an independent explanatory variable for lumbar lordosis, grip strength, and SDH mean grayscale value in female patients. In conclusion, superoxide dismutase 2-related oxidative stress in the paraspinal muscles was associated with mitochondrial dysfunction and decreased grip strength in female lumbar degenerative disease patients.
Collapse
Affiliation(s)
- Ryosuke Takahashi
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Orthopaedics, Juntendo University School of Medicine, Tokyo, Japan
- Spine and Spinal Cord Center, Juntendo University Hospital, Tokyo, Japan
| | - Hidetoshi Nojiri
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Orthopaedics, Juntendo University School of Medicine, Tokyo, Japan
- Spine and Spinal Cord Center, Juntendo University Hospital, Tokyo, Japan
| | - Yukoh Ohara
- Spine and Spinal Cord Center, Juntendo University Hospital, Tokyo, Japan
- Department of Neurosurgery, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshiyuki Fujiwara
- Department of Rehabilitation Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Muneaki Ishijima
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Orthopaedics, Juntendo University School of Medicine, Tokyo, Japan
- Spine and Spinal Cord Center, Juntendo University Hospital, Tokyo, Japan
| |
Collapse
|
3
|
Li H, Yu H, Liu D, Liao P, Gao C, Zhou J, Mei J, Zong Y, Ding P, Yao M, Wang B, Lu Y, Huang Y, Gao Y, Zhang C, Zheng M, Gao J. Adenosine diphosphate released from stressed cells triggers mitochondrial transfer to achieve tissue homeostasis. PLoS Biol 2024; 22:e3002753. [PMID: 39163396 PMCID: PMC11335167 DOI: 10.1371/journal.pbio.3002753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/12/2024] [Indexed: 08/22/2024] Open
Abstract
Cell-to-cell mitochondrial transfer has recently been shown to play a role in maintaining physiological functions of cell. We previously illustrated that mitochondrial transfer within osteocyte dendritic network regulates bone tissue homeostasis. However, the mechanism of triggering this process has not been explored. Here, we showed that stressed osteocytes in mice release adenosine diphosphate (ADP), resulting in triggering mitochondrial transfer from healthy osteocytes to restore the oxygen consumption rate (OCR) and to alleviate reactive oxygen species accumulation. Furthermore, we identified that P2Y2 and P2Y6 transduced the ADP signal to regulate osteocyte mitochondrial transfer. We showed that mitochondrial metabolism is impaired in aged osteocytes, and there were more extracellular nucleotides release into the matrix in aged cortical bone due to compromised membrane integrity. Conditioned medium from aged osteocytes triggered mitochondrial transfer between osteocytes to enhance the energy metabolism. Together, using osteocyte as an example, this study showed new insights into how extracellular ADP triggers healthy cells to rescue energy metabolism crisis in stressed cells via mitochondrial transfer in tissue homeostasis.
Collapse
Affiliation(s)
- Hao Li
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongping Yu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Delin Liu
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuan Gao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Zhou
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialun Mei
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Australia
| | - Peng Ding
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng Yao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingqi Wang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yafei Lu
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yigang Huang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youshui Gao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minghao Zheng
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, Australia
- Perron Institute for Neurological and Translational Science, Nedlands, Australia
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Zhu M, Zeiss C, Hamrick MW, Weinstein RS, Sun BH, Brotto M, Liu X, Siu E, Huttner A, Tommasini S, Simpson C, Insogna K. Mitofusin 2 plays a critical role in maintaining the functional integrity of the neuromuscular-skeletal axis. Bone 2024; 184:117086. [PMID: 38552893 DOI: 10.1016/j.bone.2024.117086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/22/2024] [Accepted: 03/23/2024] [Indexed: 05/12/2024]
Abstract
PURPOSE Mitofusin 2 (Mfn2) is one of two mitofusins involved in regulating mitochondrial size, shape and function, including mitophagy, an important cellular mechanism to limit oxidative stress. Reduced expression of Mfn2 has been associated with impaired osteoblast differentiation and function and a reduction in the number of viable osteocytes in bone. We hypothesized that the genetic absence of Mfn2 in these cells would increase their susceptibility to aging-associated metabolic stress, leading to a progressive impairment in skeletal homeostasis over time. METHODS Mfn2 was selectively deleted in vivo at three different stages of osteoblast lineage commitment by crossing mice in which the Mfn2 gene was floxed with transgenic mice expressing Cre under the control of the promoter for Osterix (OSX), collagen1a1, or DMP1 (Dentin Matrix Acidic Phosphoprotein 1). RESULTS Mice in which Mfn2 was deleted using DMP1-cre demonstrated a progressive and dramatic decline in bone mineral density (BMD) beginning at 10 weeks of age (n = 5 for each sex and each genotype from age 10 to 20 weeks). By 15 weeks, there was evidence for a functional decline in muscle performance as assessed using a rotarod apparatus (n = 3; 2 males/ 1 female for each genotype), accompanied by a decline in lean body mass. A marked reduction in trabecular bone mass was evident on bone histomorphometry, and biomechanical testing at 25 weeks (k/o: 2 male/1 female, control 2 male/2 female) revealed severely impaired femur strength. Extensive regional myofiber atrophy and degeneration was observed on skeletal muscle histology. Electron microscopy showed progressive disruption of cellular architecture, with disorganized sarcomeres and a bloated mitochondrial reticulum. There was also evidence of neurodegeneration within the ventral horn and roots of the lumbar spinal cord, which was accompanied by myelin loss and myofiber atrophy. Deletion of Mfn2 using OSX-cre or Col1a1-cre did not result in a musculoskeletal phenotype. Where possible, male and female animals were analyzed separately, but small numbers of animals in each group limited statistical power. For other outcomes, where sex was not considered, small sample sizes might still limit the strength of the observation. CONCLUSION Despite known functional overlap of Mfn1 and Mfn2 in some tissues, and their co-expression in bone, muscle and spinal cord, deletion of Mfn2 using the 8 kB DMP1 promoter uncovered an important non-redundant role for Mfn2 in maintaining the neuromuscular/bone axis.
Collapse
Affiliation(s)
- Meiling Zhu
- Yale School of Medicine, Section of Endocrinology, New Haven, CT, USA
| | - Caroline Zeiss
- Yale School of Medicine, Section of Comparative Medicine, New Haven, CT, USA
| | - Mark W Hamrick
- Department of Orthopaedic Surgery, Institute of Molecular Medicine & Genetics, Medical College of Georgia, Augusta, GA, USA
| | - Robert S Weinstein
- Division of Endocrinology, Metabolic Bone Diseases, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Ben-Hua Sun
- Yale School of Medicine, Section of Endocrinology, New Haven, CT, USA
| | - Marco Brotto
- Bone-Muscle Research Center, College of Nursing & Health Innovation, University of Texas at Arlington, Arlington, TX, USA
| | - Xinran Liu
- Yale School of Medicine, Center for Cellular and Molecular Imaging, New Haven, CT, USA
| | - Edwin Siu
- Yale School of Medicine, Section of Endocrinology, New Haven, CT, USA
| | - Anita Huttner
- Yale School of Medicine, Department of Pathology, New Haven, CT, USA
| | - Steven Tommasini
- Yale School of Medicine, Department of Orthopaedics & Rehabilitation, New Haven, CT, USA
| | - Christine Simpson
- Yale School of Medicine, Section of Endocrinology, New Haven, CT, USA
| | - Karl Insogna
- Yale School of Medicine, Section of Endocrinology, New Haven, CT, USA.
| |
Collapse
|
5
|
Dang AT, Ono M, Wang Z, Tosa I, Hara ES, Mikai A, Kitagawa W, Yonezawa T, Kuboki T, Oohashi T. Local E-rhBMP-2/β-TCP Application Rescues Osteocyte Dendritic Integrity and Reduces Microstructural Damage in Alveolar Bone Post-Extraction in MRONJ-like Mouse Model. Int J Mol Sci 2024; 25:6648. [PMID: 38928355 PMCID: PMC11203997 DOI: 10.3390/ijms25126648] [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: 04/10/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The pathology of medication-related osteonecrosis of the jaw (MRONJ), often associated with antiresorptive therapy, is still not fully understood. Osteocyte networks are known to play a critical role in maintaining bone homeostasis and repair, but the exact condition of these networks in MRONJ is unknown. On the other hand, the local application of E-coli-derived Recombinant Human Bone Morphogenetic Protein 2/β-Tricalcium phosphate (E-rhBMP-2/β-TCP) has been shown to promote bone regeneration and mitigate osteonecrosis in MRONJ-like mouse models, indicating its potential therapeutic application for the treatment of MRONJ. However, the detailed effect of BMP-2 treatment on restoring bone integrity, including its osteocyte network, in an MRONJ condition remains unclear. Therefore, in the present study, by applying a scanning electron microscope (SEM) analysis and a 3D osteocyte network reconstruction workflow on the alveolar bone surrounding the tooth extraction socket of an MRONJ-like mouse model, we examined the effectiveness of BMP-2/β-TCP therapy on the alleviation of MRONJ-related bone necrosis with a particular focus on the osteocyte network and alveolar bone microstructure (microcrack accumulation). The 3D osteocyte dendritic analysis showed a significant decrease in osteocyte dendritic parameters along with a delay in bone remodeling in the MRONJ group compared to the healthy counterpart. The SEM analysis also revealed a notable increase in the number of microcracks in the alveolar bone surface in the MRONJ group compared to the healthy group. In contrast, all of those parameters were restored in the E-rhBMP-2/β-TCP-treated group to levels that were almost similar to those in the healthy group. In summary, our study reveals that MRONJ induces osteocyte network degradation and microcrack accumulation, while application of E-rhBMP-2/β-TCP can restore a compromised osteocyte network and abrogate microcrack accumulation in MRONJ.
Collapse
Affiliation(s)
- Anh Tuan Dang
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan; (I.T.); (T.K.)
| | - Mitsuaki Ono
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
- Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Ziyi Wang
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
| | - Ikue Tosa
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan; (I.T.); (T.K.)
- Cartilage Biology and Regenerative Medicine Laboratory, Section of Growth and Development, Division of Orthodontics, College of Dental Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Emilio Satoshi Hara
- Advanced Research Center for Oral and Craniofacial Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan;
| | - Akihiro Mikai
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
| | - Wakana Kitagawa
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan; (I.T.); (T.K.)
| | - Tomoko Yonezawa
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan; (I.T.); (T.K.)
- Department of Oral Rehabilitation and Implantology, Okayama University Hospital, Okayama 700-8558, Japan
| | - Toshitaka Oohashi
- Department of Molecular Biology and Biochemistry, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8558, Japan; (A.T.D.); (Z.W.); (A.M.); (W.K.); (T.Y.); (T.O.)
| |
Collapse
|
6
|
Kwon Y. YAP/TAZ as Molecular Targets in Skeletal Muscle Atrophy and Osteoporosis. Aging Dis 2024:AD.2024.0306. [PMID: 38502585 DOI: 10.14336/ad.2024.0306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/06/2024] [Indexed: 03/21/2024] Open
Abstract
Skeletal muscles and bones are closely connected anatomically and functionally. Age-related degeneration in these tissues is associated with physical disability in the elderly and significantly impacts their quality of life. Understanding the mechanisms of age-related musculoskeletal tissue degeneration is crucial for identifying molecular targets for therapeutic interventions for skeletal muscle atrophy and osteoporosis. The Hippo pathway is a recently identified signaling pathway that plays critical roles in development, tissue homeostasis, and regeneration. The Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are key downstream effectors of the mammalian Hippo signaling pathway. This review highlights the fundamental roles of YAP and TAZ in the homeostatic maintenance and regeneration of skeletal muscles and bones. YAP/TAZ play a significant role in stem cell function by relaying various environmental signals to stem cells. Skeletal muscle atrophy and osteoporosis are related to stem cell dysfunction or senescence triggered by YAP/TAZ dysregulation resulting from reduced mechanosensing and mitochondrial function in stem cells. In contrast, the maintenance of YAP/TAZ activation can suppress stem cell senescence and tissue dysfunction and may be used as a basis for the development of potential therapeutic strategies. Thus, targeting YAP/TAZ holds significant therapeutic potential for alleviating age-related muscle and bone dysfunction and improving the quality of life in the elderly.
Collapse
|
7
|
Niroobakhsh M, Laughrey LE, Dallas SL, Johnson ML, Ganesh T. Computational modeling based on confocal imaging predicts changes in osteocyte and dendrite shear stress due to canalicular loss with aging. Biomech Model Mechanobiol 2024; 23:129-143. [PMID: 37642807 DOI: 10.1007/s10237-023-01763-w] [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: 01/30/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
Exercise and physical activity exert mechanical loading on the bones which induces bone formation. However, the relationship between the osteocyte lacunar-canalicular morphology and mechanical stress experienced locally by osteocytes transducing signals for bone formation is not fully understood. In this study, we used computational modeling to predict the effect of canalicular density, the number of fluid inlets, and load direction on fluid flow shear stress (FFSS) and bone strains and how these might change following the microstructural deterioration of the lacunar-canalicular network that occurs with aging. Four distinct computational models were initially generated of osteocytes with either ten or eighteen dendrites using a fluid-structure interaction method with idealized geometries. Next, a young and a simulated aged osteocyte were developed from confocal images after FITC staining of the femur of a 4-month-old C57BL/6 mouse to estimate FFSS using a computational fluid dynamics approach. The models predicted higher fluid velocities in the canaliculi versus the lacunae. Comparison of idealized models with five versus one fluid inlet indicated that with four more inlets, one-half of the dendrites experienced FFSS greater than 0.8 Pa, which has been associated with osteogenic responses. Confocal image-based models of real osteocytes indicated a six times higher ratio of canalicular to lacunar surface area in the young osteocyte model than the simulated aged model and the average FFSS in the young model (FFSS = 0.46 Pa) was three times greater than the aged model (FFSS = 0.15 Pa). Interestingly, the surface area with FFSS values above 0.8 Pa was 23 times greater in the young versus the simulated aged model. These findings may explain the impaired mechano-responsiveness of osteocytes with aging.
Collapse
Affiliation(s)
- Mohammad Niroobakhsh
- Division of Natural and Built Environment, School of Science and Engineering, University of Missouri-Kansas City, 350 L Flarsheim Hall, 5100 Rockhill Road, Kansas City, MO, 64110, USA
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 620 E 25th Street, Kansas City, MO, 64108, USA
| | - Loretta E Laughrey
- Division of Natural and Built Environment, School of Science and Engineering, University of Missouri-Kansas City, 350 L Flarsheim Hall, 5100 Rockhill Road, Kansas City, MO, 64110, USA
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 620 E 25th Street, Kansas City, MO, 64108, USA
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 620 E 25th Street, Kansas City, MO, 64108, USA
| | - Mark L Johnson
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, 620 E 25th Street, Kansas City, MO, 64108, USA
| | - Thiagarajan Ganesh
- Division of Natural and Built Environment, School of Science and Engineering, University of Missouri-Kansas City, 350 L Flarsheim Hall, 5100 Rockhill Road, Kansas City, MO, 64110, USA.
| |
Collapse
|
8
|
Bi J, Zhang C, Lu C, Mo C, Zeng J, Yao M, Jia B, Liu Z, Yuan P, Xu S. Age-related bone diseases: Role of inflammaging. J Autoimmun 2024; 143:103169. [PMID: 38340675 DOI: 10.1016/j.jaut.2024.103169] [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: 11/08/2023] [Revised: 01/03/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024]
Abstract
Bone aging is characterized by an imbalance in the physiological and pathological processes of osteogenesis, osteoclastogenesis, adipogenesis, and chondrogenesis, resulting in exacerbated bone loss and the development of age-related bone diseases, including osteoporosis, osteoarthritis, rheumatoid arthritis, and periodontitis. Inflammaging, a novel concept in the field of aging research, pertains to the persistent and gradual escalation of pro-inflammatory reactions during the aging process. This phenomenon is distinguished by its low intensity, systemic nature, absence of symptoms, and potential for management. The mechanisms by which inflammaging contribute to age-related chronic diseases, particularly in the context of age-related bone diseases, remain unclear. The precise manner in which systemic inflammation induces bone aging and consequently contributes to the development of age-related bone diseases has yet to be fully elucidated. This article primarily examines the mechanisms underlying inflammaging and its association with age-related bone diseases, to elucidate the potential mechanisms of inflammaging in age-related bone diseases and offer insights for developing preventive and therapeutic strategies for such conditions.
Collapse
Affiliation(s)
- Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Caimei Zhang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Caihong Lu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Mingyan Yao
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, China; Department of Endocrinology, Baoding No.1 Central Hospital, Baoding, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Peiyan Yuan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Marques-Carvalho A, Kim HN, Almeida M. The role of reactive oxygen species in bone cell physiology and pathophysiology. Bone Rep 2023; 19:101664. [PMID: 38163012 PMCID: PMC10757300 DOI: 10.1016/j.bonr.2023.101664] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/26/2023] Open
Abstract
Hydrogen peroxide (H2O2), superoxide anion radical (O2-•), and other forms of reactive oxygen species (ROS) are produced by the vast majority of mammalian cells and can contribute both to cellular homeostasis and dysfunction. The NADPH oxidases (NOX) enzymes and the mitochondria electron transport chain (ETC) produce most of the cellular ROS. Multiple antioxidant systems prevent the accumulation of excessive amounts of ROS which cause damage to all cellular macromolecules. Many studies have examined the contribution of ROS to different bone cell types and to skeletal physiology and pathophysiology. Here, we discuss the role of H2O2 and O2-• and their major enzymatic sources in osteoclasts and osteoblasts, the fundamentally different ways via which these cell types utilize mitochondrial derived H2O2 for differentiation and function, and the molecular mechanisms that impact and are altered by ROS in these cells. Particular emphasis is placed on evidence obtained from mouse models describing the contribution of different sources of ROS or antioxidant enzymes to bone resorption and formation. Findings from studies using pharmacological or genetically modified mouse models indicate that an increase in H2O2 and perhaps other ROS contribute to the loss of bone mass with aging and estrogen deficiency, the two most important causes of osteoporosis and increased fracture risk in humans.
Collapse
Affiliation(s)
- Adriana Marques-Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Ha-Neui Kim
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Maria Almeida
- Division of Endocrinology and Metabolism, University of Arkansas for Medical Sciences, Little Rock, USA
- Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Orthopedic Surgery, University of Arkansas for Medical Sciences, Little Rock, USA
| |
Collapse
|
10
|
Yang T, Chen W, Gan K, Wang C, Xie X, Su Y, Lian H, Xu J, Zhao J, Liu Q. Myrislignan targets extracellular signal-regulated kinase (ERK) and modulates mitochondrial function to dampen osteoclastogenesis and ovariectomy-induced osteoporosis. J Transl Med 2023; 21:839. [PMID: 37993937 PMCID: PMC10664306 DOI: 10.1186/s12967-023-04706-2] [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: 04/07/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Activated osteoclasts cause excessive bone resorption, and disrupt bone homeostasis, leading to osteoporosis. The extracellular signal-regulated kinase (ERK) signaling is the classical pathway related to osteoclast differentiation, and mitochondrial reactive oxygen species are closely associated with the differentiation of osteoclasts. Myrislignan (MRL), a natural product derived from nutmeg, has multiple pharmacological activities; however, its therapeutic effect on osteoporosis is unclear. Here, we investigated whether MRL could inhibit osteoclastogenesis and bone mass loss in an ovariectomy mouse model by suppressing mitochondrial function and ERK signaling. METHODS Tartrate-resistant and phosphatase (TRAP) and bone resorption assays were performed to observe the effect of MRL on osteoclastogenesis of bone marrow macrophages. MitoSOX RED and tetramethyl rhodamine methyl ester (TMRM) staining was performed to evaluate the inhibitory effect of MRL on mitochondria. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) assay was performed to detect whether MRL suppressed the expression of osteoclast-specific genes. The impact of MRL on the protein involved in the mitogen-activated protein kinase (MAPK) and nuclear factor-κB pathways was evaluated using western blotting. In addition, a specific ERK agonist LM22B-10, was used to revalidate the inhibitory effect of MRL on ERK. Finally, we established an ovariectomy mouse model to assess the therapeutic effect of MRL on osteoporosis in vivo. RESULTS MRL inhibited osteoclast differentiation and the associated bone resorption, by significantly decreasing osteoclastic gene expression. Mechanistically, MRL inhibited the phosphorylation of ERK by suppressing the mitochondrial function, thereby downregulating the nuclear factor of activated T cells 1 (NFATc1) signaling. LM22B-10 treatment further verified the targeted inhibition effect of MRL on ERK. Microscopic computed tomographic and histologic analyses of the tibial tissue sections indicated that ovariectomized mice had lower bone mass and higher expression of ERK compared with normal controls. However, MRL treatment significantly reversed these effects, indicating the anti-osteoporosis effect of MRL. CONCLUSION We report for the first time that MRL inhibits ERK signaling by suppressing mitochondrial function, thereby ameliorating ovariectomy-induced osteoporosis. Our findings can provide a basis for the development of a novel therapeutic strategy for osteoporosis.
Collapse
Affiliation(s)
- Tao Yang
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Weiwei Chen
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Kai Gan
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chaofeng Wang
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiaoxiao Xie
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yuangang Su
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Haoyu Lian
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jiake Xu
- School of Biomedical Sciences, the University of Western Australia, Perth, WA, 6009, Australia.
| | - Jinmin Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
11
|
Catheline SE, Kaiser E, Eliseev RA. Mitochondrial Genetics and Function as Determinants of Bone Phenotype and Aging. Curr Osteoporos Rep 2023; 21:540-551. [PMID: 37542684 DOI: 10.1007/s11914-023-00816-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/12/2023] [Indexed: 08/07/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the recently published scientific literature regarding the effects of mitochondrial function and mitochondrial genome mutations on bone phenotype and aging. RECENT FINDINGS While aging and sex steroid levels have traditionally been considered the most important risk factors for development of osteoporosis, mitochondrial function and genetics are being increasingly recognized as important determinants of bone health. Recent studies indicate that mitochondrial genome variants found in different human populations determine the risk of complex degenerative diseases. We propose that osteoporosis should be among such diseases. Studies have shown the deleterious effects of mitochondrial DNA mutations and mitochondrial dysfunction on bone homeostasis. Mediators of such effects include oxidative stress, mitochondrial permeability transition, and dysregulation of autophagy. Mitochondrial health plays an important role in bone homeostasis and aging, and understanding underlying mechanisms is critical in leveraging this relationship clinically for therapeutic benefit.
Collapse
Affiliation(s)
- Sarah E Catheline
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Ethan Kaiser
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, USA
| | - Roman A Eliseev
- Center for Musculoskeletal Research, University of Rochester School of Medicine and Dentistry, Rochester, USA.
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, USA.
| |
Collapse
|
12
|
Yoon J, Kaya S, Matsumae G, Dole N, Alliston T. miR181a/b-1 controls osteocyte metabolism and mechanical properties independently of bone morphology. Bone 2023; 175:116836. [PMID: 37414200 PMCID: PMC11156520 DOI: 10.1016/j.bone.2023.116836] [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: 02/22/2023] [Revised: 06/30/2023] [Accepted: 07/01/2023] [Indexed: 07/08/2023]
Abstract
Bone derives its ability to resist fracture from bone mass and quality concurrently; however, many questions about the molecular mechanisms controlling bone quality remain unanswered, limiting the development of diagnostics and therapeutics. Despite the increasing evidence on the importance of miR181a/b-1 in bone homeostasis and disease, whether and how osteocyte-intrinsic miR181a/b-1 controls bone quality remains elusive. Osteocyte-intrinsic deletion of miR181a/b-1 in osteocytes in vivo resulted in compromised overall bone mechanical behavior in both sexes, although the parameters affected by miR181a/b-1 varied distinctly based on sex. Furthermore, impaired fracture resistance in both sexes was unexplained by cortical bone morphology, which was altered in female mice and intact in male mice with miR181a/b-1-deficient osteocytes. The role of miR181a/b-1 in the regulation of osteocyte metabolism was apparent in bioenergetic testing of miR181a/b-1-deficient OCY454 osteocyte-like cells and transcriptomic analysis of cortical bone from mice with osteocyte-intrinsic ablation of miR181a/b-1. Altogether, this study demonstrates the control of osteocyte bioenergetics and the sexually dimorphic regulation of cortical bone morphology and mechanical properties by miR181a/b-1, hinting at the role of osteocyte metabolism in the regulation of mechanical behavior.
Collapse
Affiliation(s)
- Jihee Yoon
- Department of Orthopaedic Surgery, University of California San Francisco, California, USA; Oral and Craniofacial Sciences Program, School of Dentistry, University of California San Francisco, California, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California San Francisco, California, USA
| | - Gen Matsumae
- Department of Orthopaedic Surgery, University of California San Francisco, California, USA
| | - Neha Dole
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, AR, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, California, USA; Oral and Craniofacial Sciences Program, School of Dentistry, University of California San Francisco, California, USA.
| |
Collapse
|
13
|
Riegger J, Schoppa A, Ruths L, Haffner-Luntzer M, Ignatius A. Oxidative stress as a key modulator of cell fate decision in osteoarthritis and osteoporosis: a narrative review. Cell Mol Biol Lett 2023; 28:76. [PMID: 37777764 PMCID: PMC10541721 DOI: 10.1186/s11658-023-00489-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/11/2023] [Indexed: 10/02/2023] Open
Abstract
During aging and after traumatic injuries, cartilage and bone cells are exposed to various pathophysiologic mediators, including reactive oxygen species (ROS), damage-associated molecular patterns, and proinflammatory cytokines. This detrimental environment triggers cellular stress and subsequent dysfunction, which not only contributes to the development of associated diseases, that is, osteoporosis and osteoarthritis, but also impairs regenerative processes. To counter ROS-mediated stress and reduce the overall tissue damage, cells possess diverse defense mechanisms. However, cellular antioxidative capacities are limited and thus ROS accumulation can lead to aberrant cell fate decisions, which have adverse effects on cartilage and bone homeostasis. In this narrative review, we address oxidative stress as a major driver of pathophysiologic processes in cartilage and bone, including senescence, misdirected differentiation, cell death, mitochondrial dysfunction, and impaired mitophagy by illustrating the consequences on tissue homeostasis and regeneration. Moreover, we elaborate cellular defense mechanisms, with a particular focus on oxidative stress response and mitophagy, and briefly discuss respective therapeutic strategies to improve cell and tissue protection.
Collapse
Affiliation(s)
- Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081, Ulm, Germany.
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Leonie Ruths
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| |
Collapse
|
14
|
Hu G, Yu Y, Sharma D, Pruett-Miller SM, Ren Y, Zhang GF, Karner CM. Glutathione limits RUNX2 oxidation and degradation to regulate bone formation. JCI Insight 2023; 8:e166888. [PMID: 37432749 PMCID: PMC10543723 DOI: 10.1172/jci.insight.166888] [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: 11/03/2022] [Accepted: 07/06/2023] [Indexed: 07/12/2023] Open
Abstract
Reactive oxygen species (ROS) are natural products of mitochondrial oxidative metabolism and oxidative protein folding. ROS levels must be well controlled, since elevated ROS has been shown to have deleterious effects on osteoblasts. Moreover, excessive ROS is thought to underlie many of the skeletal phenotypes associated with aging and sex steroid deficiency in mice and humans. The mechanisms by which osteoblasts regulate ROS and how ROS inhibits osteoblasts are not well understood. Here, we demonstrate that de novo glutathione (GSH) biosynthesis is essential in neutralizing ROS and establish a proosteogenic reduction and oxidation reaction (REDOX) environment. Using a multifaceted approach, we demonstrate that reducing GSH biosynthesis led to acute degradation of RUNX2, impaired osteoblast differentiation, and reduced bone formation. Conversely, reducing ROS using catalase enhanced RUNX2 stability and promoted osteoblast differentiation and bone formation when GSH biosynthesis was limited. Highlighting the therapeutic implications of these findings, in utero antioxidant therapy stabilized RUNX2 and improved bone development in the Runx2+/- haplo-insufficient mouse model of human cleidocranial dysplasia. Thus, our data establish RUNX2 as a molecular sensor of the osteoblast REDOX environment and mechanistically clarify how ROS negatively impacts osteoblast differentiation and bone formation.
Collapse
Affiliation(s)
- Guoli Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yilin Yu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Deepika Sharma
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Shondra M. Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Yinshi Ren
- Center for Excellence in Hip Disorders, Texas Scottish Rite Hospital for Children, Dallas, Texas, USA
| | - Guo-Fang Zhang
- Department of Medicine, Division of Endocrinology, Metabolism Nutrition, and
- Sarah W. Stedman Nutrition and Metabolism Center & Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, USA
| | - Courtney M. Karner
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, USA
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
15
|
Sang W, Ural A. Evaluating the Role of Canalicular Morphology and Perilacunar Region Properties on Local Mechanical Environment of Lacunar-Canalicular Network Using Finite Element Modeling. J Biomech Eng 2023; 145:1156059. [PMID: 36629002 DOI: 10.1115/1.4056655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 01/07/2023] [Indexed: 01/12/2023]
Abstract
Physiological and pathological processes such as aging, diseases, treatments, and lactation can alter lacunar-canalicular network (LCN) morphology and perilacunar region properties. These modifications can impact the mechanical environment of osteocytes which in turn can influence osteocyte mechanosensitivity and the remodeling process. In this study, we aim to evaluate how the modifications in the canalicular morphology, lacunar density, and the perilacunar region properties influence the local mechanical environment of LCN and the apparent bone properties using three-dimensional finite element (FE) modeling. The simulation results showed that a 50% reduction in perilacunar elastic modulus led to about 7% decrease in apparent elastic modulus of the bone. The increase in canalicular density, length, and diameter did not influence the strain amplification in the models but they increased the amount of highly strained bone around LCN. Change in lacunar density did not influence the strain amplification and the amount of highly strained regions on LCN surfaces. Reduction in perilacunar elastic modulus increased both the strain amplification and the volume of highly strained tissue around and on the surface of LCN. The FE models of LCN in this study can be utilized to quantify the influence of modifications in canalicular morphology, lacunar density, and perilacunar region properties on the apparent bone properties and the local mechanical environment of LCN. Although this is a numerical study with idealized models, it provides important information on how mechanical environment of osteocytes is influenced by the modifications in LCN morphology and perilacunar region properties due to physiological and pathological processes.
Collapse
Affiliation(s)
- Wen Sang
- Department of Mechanical Engineering, Villanova University, 800 Lancaster Avenue, Villanova, PA 19085
| | - Ani Ural
- Department of Mechanical Engineering, Villanova University, 800 Lancaster Avenue, Villanova, PA 19085
| |
Collapse
|
16
|
Zhang C, Li H, Li J, Hu J, Yang K, Tao L. Oxidative stress: A common pathological state in a high-risk population for osteoporosis. Biomed Pharmacother 2023; 163:114834. [PMID: 37163779 DOI: 10.1016/j.biopha.2023.114834] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/29/2023] [Accepted: 05/01/2023] [Indexed: 05/12/2023] Open
Abstract
Osteoporosis is becoming a major concern in the field of public health. The process of bone loss is insidious and does not directly induce obvious symptoms. Complications indicate an irreversible decrease in bone mass. The high-risk populations of osteoporosis, including postmenopausal women, elderly men, diabetic patients and obese individuals need regular bone mineral density testing and appropriate preventive treatment. However, the primary changes in these populations are different, increasing the difficulty of effective treatment of osteoporosis. Determining the core pathogenesis of osteoporosis helps improve the efficiency and efficacy of treatment among these populations. Oxidative stress is a common pathological state secondary to estrogen deficiency, aging, hyperglycemia and hyperlipemia. In this review, we divided oxidative stress into the direct effect of reactive oxygen species (ROS) and the reduction of antioxidant enzyme activity to discuss their roles in the development of osteoporosis. ROS initiated mitochondrial apoptotic signaling and suppressed osteogenic marker expression to weaken osteogenesis. MAPK and NF-κB signaling pathways mediated the positive effect of ROS on osteoclast differentiation. Antioxidant enzymes not only eliminate the negative effects of ROS, but also directly participate in the regulation of bone metabolism. Additionally, we also described the roles of proinflammatory factors and HIF-1α under the pathophysiological changes of inflammation and hypoxia, which provided a supplement of oxidative stress-induced osteoporosis. In conclusion, our review showed that oxidative stress was a common pathological state in a high-risk population for osteoporosis. Targeted oxidative stress treatment would greatly optimize the therapeutic schedule of various osteoporosis treatments.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Hao Li
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Jie Li
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China
| | - Jiajin Hu
- Health Sciences Institute, China Medical University, Shenyang 110122, China
| | - Keda Yang
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China.
| | - Lin Tao
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nanjing North Street, Shenyang, China.
| |
Collapse
|
17
|
Li F, Lun D, Liu D, Jia Z, Zhu Z, Liu Z, Li X. Melatonin activates mitochondrial unfolded protein response to preserve osteogenic potential of senescent BMSCs via upregulating PDI-6. Biochimie 2023; 209:44-51. [PMID: 36708867 DOI: 10.1016/j.biochi.2023.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/04/2023] [Accepted: 01/18/2023] [Indexed: 01/27/2023]
Abstract
Bone marrow stromal cells (BMSCs) possess the capability to differentiate into osteogenic or adipogenic lineages. With aging, BMSCs suffer from mitochondrial dysfunction and undergo senescence, favoring adipogenesis at the expense of osteoblastogenesis. It leads to decreased bone formation and contributes to senile osteoporosis (SOP). In the current study, RNA-seq analysis unveiled that senescent BMSCs from mice exhibited a significant suppression in the expression of the protein disulfide isomerase PDI-6, an important regulator of mitochondrial unfolded protein response (UPRmt) as well as maintenance of mitochondrial homeostasis. Overexpression of PDI-6 in senescent BMSCs partially rescued mitochondrial function and enhanced osteogenic differentiation. In contrast, osteoblastogenesis of BMSCs remarkably deteriorated under the condition of PDI-6 silencing. Furthermore, melatonin, an endocrine hormone, effectively enhanced PDI-6 expression and repaired injured mitochondria, and the effect of melatonin on PDI-6 expression was melatonin receptor dependent. We further identified that PDI-6 was a downstream effector of Wnt/β-catenin pathway, as the inhibitor of Wnt3A/TCF signaling, Wnt-C59, inhibited PDI-6 expression. Potential β-catenin-TCF/LEF binding sites on the promoter of PDI-6 gene were also validated by chromatin immunoprecipitation (ChIP) assay. Thus, our study suggests that PDI-6 is a pharmacological target of melatonin for the intervention of age-related osteoporosis via mitigating mitochondrial dysfunction in senescent BMSCs.
Collapse
Affiliation(s)
- Feng Li
- Department of Orthopaedics, Weifang People's Hospital, Weifang, Shandong, China
| | - Dengxing Lun
- Department of Orthopaedics, Weifang People's Hospital, Weifang, Shandong, China
| | - Dayong Liu
- Department of Orthopaedics, Weifang People's Hospital, Weifang, Shandong, China
| | - Zesen Jia
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Rd, Heping, Tianjin, China
| | - Zhenye Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Rd, Heping, Tianjin, China
| | - Zhiqiang Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Rd, Heping, Tianjin, China
| | - Xiaopeng Li
- Department of Orthopaedics, Weifang People's Hospital, Weifang, Shandong, China.
| |
Collapse
|
18
|
Wang JS, Wein MN. Pathways Controlling Formation and Maintenance of the Osteocyte Dendrite Network. Curr Osteoporos Rep 2022; 20:493-504. [PMID: 36087214 PMCID: PMC9718876 DOI: 10.1007/s11914-022-00753-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 01/30/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to discuss the molecular mechanisms involved in osteocyte dendrite formation, summarize the similarities between osteocytic and neuronal projections, and highlight the importance of osteocyte dendrite maintenance in human skeletal disease. RECENT FINDINGS It is suggested that there is a causal relationship between the loss of osteocyte dendrites and the increased osteocyte apoptosis during conditions including aging, microdamage, and skeletal disease. A few mechanisms are proposed to control dendrite formation and outgrowth, such as via the regulation of actin polymerization dynamics. This review addresses the impact of osteocyte dendrites in bone health and disease. Recent advances in multi-omics, in vivo and in vitro models, and microscopy-based imaging have provided novel approaches to reveal the underlying mechanisms that regulate dendrite development. Future therapeutic approaches are needed to target the process of osteocyte dendrite formation.
Collapse
Affiliation(s)
- Jialiang S Wang
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
19
|
Natural Compounds That Enhance Motor Function in a Mouse Model of Muscle Fatigue. Biomedicines 2022; 10:biomedicines10123073. [PMID: 36551829 PMCID: PMC9775528 DOI: 10.3390/biomedicines10123073] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
Musculoskeletal disease can be a serious condition associated with aging that may lead to fractures and a bedridden state due to decreased motor function. In addition to exercise training to increase muscle mass, increasing muscle function with the intake of functional foods is an effective treatment strategy for musculoskeletal disease. Muscle-specific SOD2-deficient mice (muscle-Sod2-/-) show a severe disturbance in exercise in association with increased mitochondrial reactive oxygen species, as well as mitochondrial dysfunction and muscle damage. In the present study, to develop a therapeutic strategy for musculoskeletal disease, we searched for substances that enhanced motor function among functional compounds by in vivo screening using muscle-Sod2-/- mice as a muscle fatigue model. We administered 96 compounds, including antioxidants, to muscle-Sod2-/- mice and assessed their effects on treadmill performance. Among the administered compounds, gossypin, genistein, kaempferol, taxifolin, fumaric acid, β-hydroxy-β-methylbutyrate Ca, and astaxanthin, which are dietary functional food factors, increased forced running time in muscle-Sod2-/- mice. In addition, troglitazone, tempol, trolox, and MnTE-2-PyP, which are antioxidants, also significantly increased the running ability of muscle-Sod2-/- mice. These results suggest that the intake of functional foods with antioxidant activity can improve motor function. Muscle-Sod2-/- mice, as a muscle fatigue model, are suitable for the in vivo screening of functional substances that promote improvements in exercise and muscle performance.
Collapse
|
20
|
Ma J, Wang A, Zhang H, Liu B, Geng Y, Xu Y, Zuo G, Jia P. Iron overload induced osteocytes apoptosis and led to bone loss in Hepcidin -/- mice through increasing sclerostin and RANKL/OPG. Bone 2022; 164:116511. [PMID: 35933095 DOI: 10.1016/j.bone.2022.116511] [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: 05/18/2022] [Revised: 07/22/2022] [Accepted: 08/01/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Numerous studies have demonstrated that iron overload is a risk factor of osteoporosis. However, there has been no systematic and in-depth studies on the effect of iron overload on osteocytes and its role in iron overload-induced bone loss. Therefore, to address this problem, we carried out in vitro and in vivo studies using MLO-Y4 osteocyte-like cells and Hepcidin-/- mice as iron overload models. METHODS (1) MLO-Y4 cells were treated with ferric ammonium citrate (FAC). Intracellular reactive oxygen species (ROS) levels and apoptosis of MLO-Y4 cells were determined by flow cytometry. Western blotting was performed to evaluate the effect of FAC on the expression of sclerostin and RANKL/OPG. (2) The conditioned medium of MLO-Y4 cells after treatment with FAC was collected and used to treat pre-osteoblasts and monocytes. Alkaline phosphatase (ALP) staining and alizarin red (AR) staining were used to evaluate osteogenic differentiation capacity, and tartrate-resistant acid phosphatase (TRAP) staining was performed to demonstrate osteoclast differentiation capacity. (3) In vivo studies included a wild type mouse, Hepcidin-/- mice, Hepcidin-/- mice + deferoxamine (DFO), and Hepcidin-/- mice + N-actyl-l-cysteine (NAC) group. Micro-CT was performed to evaluate the bone mineral density (BMD), bone volume, and bone micro-architecture of the mice, and three bending tests were used to assess bone strength. Histological analysis was used to detect alterations in bone turnover. TUNEL staining and scanning electron microscopy (SEM) were performed to evaluate the apoptosis and morphology of osteocytes. Immunohistochemical staining and Western blotting were used to determine alterations in sclerostin and RANKL/OPG expression levels in mice. RESULTS (1) FAC increased intracellular ROS and apoptosis in MLO-Y4 cells, while FAC enhanced the expression of sclerostin and RANKL/OPG in MLO-Y4 cells. (2) Conditioned medium of MLO-Y4 cells inhibited the osteogenic capacity of osteoblasts while stimulating osteoclast differentiation. (3) By increasing oxidative stress, iron overload promotes the apoptosis of osteocytes and undermines the morphology of osteocytes in Hepcidin-/- mice, further increasing the expression levels of sclerostin and RANKL/OPG in osteocytes, which is considered to be the causative factor for reduced bone formation and enhanced bone resorption. DFO administration reduced iron levels, and NAC treatment decreased oxidative stress in Hepcidin-/- mice. Therefore, DFO or NAC treatment rescued the decrease in BMD, bone volume, and bone strength and attenuated the deterioration of bone architecture in Hepcidin-/- mice by attenuating the effect of iron overload on osteocytes. CONCLUSION Osteocyte apoptosis due to increased ROS and resultant sclerostin and RANKL/OPG expression alteration was the main reason for bone loss in Hepcidin-/- mice. Osteocytes are the main targets for the prevention and treatment of iron overload-induced osteoporosis.
Collapse
Affiliation(s)
- Jiawei Ma
- Second Affiliated Hospital of Soochow University, Orthopedic Department, China; Osteoporosis Research Institute of Soochow University, China
| | - Aifei Wang
- Second Affiliated Hospital of Soochow University, Orthopedic Department, China; Osteoporosis Research Institute of Soochow University, China
| | - Hui Zhang
- Second Affiliated Hospital of Soochow University, Orthopedic Department, China; Osteoporosis Research Institute of Soochow University, China
| | - Baoshan Liu
- Second Affiliated Hospital of Soochow University, Orthopedic Department, China; Osteoporosis Research Institute of Soochow University, China
| | - Yu Geng
- Second Affiliated Hospital of Soochow University, Orthopedic Department, China; Osteoporosis Research Institute of Soochow University, China
| | - Youjia Xu
- Second Affiliated Hospital of Soochow University, Orthopedic Department, China; Osteoporosis Research Institute of Soochow University, China
| | - Guilai Zuo
- Shandong Provincial Qianfoshan Hospital, The First Affiliated Hospital of Shandong First Medical University, Orthopedic Department, China.
| | - Peng Jia
- Second Affiliated Hospital of Soochow University, Orthopedic Department, China; Osteoporosis Research Institute of Soochow University, China.
| |
Collapse
|
21
|
Wang H, Cao X, Guo J, Yang X, Sun X, Fu Z, Qin A, Wu Y, Zhao J. BNTA alleviates inflammatory osteolysis by the SOD mediated anti-oxidation and anti-inflammation effect on inhibiting osteoclastogenesis. Front Pharmacol 2022; 13:939929. [PMID: 36249770 PMCID: PMC9559729 DOI: 10.3389/fphar.2022.939929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 09/09/2022] [Indexed: 11/15/2022] Open
Abstract
Abnormal activation and overproliferation of osteoclast in inflammatory bone diseases lead to osteolysis and bone mass loss. Although current pharmacological treatments have made extensive advances, limitations still exist. N-[2-bromo-4-(phenylsulfonyl)-3-thienyl]-2-chlorobenzamide (BNTA) is an artificially synthesized molecule compound that has antioxidant and anti-inflammatory properties. In this study, we presented that BNTA can suppress intracellular ROS levels through increasing ROS scavenging enzymes SOD1 and SOD2, subsequently attenuating the MARK signaling pathway and the transcription of NFATc1, leading to the inhibition of osteoclast formation and osteolytic resorption. Moreover, the results also showed an obvious restrained effect of BNTA on RANKL-stimulated proinflammatory cytokines, which indirectly mediated osteoclastogenesis. In line with the in vitro results, BNTA protected LPS-induced severe bone loss in vivo by enhancing scavenging enzymes, reducing proinflammatory cytokines, and decreasing osteoclast formation. Taken together, all of the results demonstrate that BNTA effectively represses oxidation, regulates inflammatory activity, and inhibits osteolytic bone resorption, and it may be a potential and exploitable drug to prevent inflammatory osteolytic bone diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yujie Wu
- *Correspondence: Yujie Wu, ; Jie Zhao,
| | - Jie Zhao
- *Correspondence: Yujie Wu, ; Jie Zhao,
| |
Collapse
|
22
|
Buettmann EG, Goldscheitter GM, Hoppock GA, Friedman MA, Suva LJ, Donahue HJ. Similarities Between Disuse and Age-Induced Bone Loss. J Bone Miner Res 2022; 37:1417-1434. [PMID: 35773785 PMCID: PMC9378610 DOI: 10.1002/jbmr.4643] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 11/07/2022]
Abstract
Disuse and aging are known risk factors associated with low bone mass and quality deterioration, resulting in increased fracture risk. Indeed, current and emerging evidence implicate a large number of shared skeletal manifestations between disuse and aging scenarios. This review provides a detailed overview of current preclinical models of musculoskeletal disuse and the clinical scenarios they seek to recapitulate. We also explore and summarize the major similarities between bone loss after extreme disuse and advanced aging at multiple length scales, including at the organ/tissue, cellular, and molecular level. Specifically, shared structural and material alterations of bone loss are presented between disuse and aging, including preferential loss of bone at cancellous sites, cortical thinning, and loss of bone strength due to enhanced fragility. At the cellular level bone loss is accompanied, during disuse and aging, by increased bone resorption, decreased formation, and enhanced adipogenesis due to altered gap junction intercellular communication, WNT/β-catenin and RANKL/OPG signaling. Major differences between extreme short-term disuse and aging are discussed, including anatomical specificity, differences in bone turnover rates, periosteal modeling, and the influence of subject sex and genetic variability. The examination also identifies potential shared mechanisms underlying bone loss in aging and disuse that warrant further study such as collagen cross-linking, advanced glycation end products/receptor for advanced glycation end products (AGE-RAGE) signaling, reactive oxygen species (ROS) and nuclear factor κB (NF-κB) signaling, cellular senescence, and altered lacunar-canalicular connectivity (mechanosensation). Understanding the shared structural alterations, changes in bone cell function, and molecular mechanisms common to both extreme disuse and aging are paramount to discovering therapies to combat both age-related and disuse-induced osteoporosis. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Evan G Buettmann
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Galen M Goldscheitter
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Gabriel A Hoppock
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Michael A Friedman
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| | - Larry J Suva
- Department of Veterinary Physiology and Pharmacology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Henry J Donahue
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
23
|
Ghomashchi S, Clement A, Whyne CM, Akens MK. Establishment and Image based evaluation of a New Preclinical Rat Model of Osteoblastic Bone Metastases. Clin Exp Metastasis 2022; 39:833-840. [PMID: 35819644 DOI: 10.1007/s10585-022-10175-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/08/2022] [Indexed: 11/28/2022]
Abstract
Bone remodeling is disrupted in the presence of metastases and can present as osteolytic, osteoblastic or a mixture of the two. Established rat models of osteolytic and mixed metastases have been identified changes in structural and tissue-level properties of bone. The aim of this work was to establish a preclinical rat model of osteoblastic metastases and characterize bone quality changes through image-based evaluation. Female athymic rats (n = 22) were inoculated with human breast cancer cells ZR-75-1 and tumor development tracked over 3-4 months with bioluminescence and in-vivo µCT imaging. Bone tissue-level stereological features were quantified on ex-vivo µCT imaging. Histopathology verified the presence of osteoblastic bone. Bone mineral density distribution was assessed via backscattered electron microscopy. Newly formed osteoblastic bone was associated with reduced mineral content and increased heterogeneity leading to an overall degraded bone quality. Characterizing changes in osteoblastic bone properties is relevant to pre-clinical therapeutic testing and treatment planning.
Collapse
Affiliation(s)
- Soroush Ghomashchi
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Allison Clement
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| | - Cari M Whyne
- Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Surgery, University of Toronto, 101 College Street, Rm 15-311, M5G 1L7, Toronto, ON, Canada
| | - Margarete K Akens
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada. .,Department of Surgery, University of Toronto, 101 College Street, Rm 15-311, M5G 1L7, Toronto, ON, Canada. .,Techna Institute, University Health Network, 101 College Street, Rm 15-311, M5G 1L7, Toronto, ON, Canada.
| |
Collapse
|
24
|
Goff E, Cohen A, Shane E, Recker RR, Kuhn G, Müller R. Large-scale osteocyte lacunar morphological analysis of transiliac bone in normal and osteoporotic premenopausal women. Bone 2022; 160:116424. [PMID: 35460961 DOI: 10.1016/j.bone.2022.116424] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 11/29/2022]
Abstract
Bone's ability to adapt is governed by the network of embedded osteocytes, which inhabit individual pores called lacunae. The morphology of these lacunae and their resident osteocytes are known to change with age and diseases such as postmenopausal osteoporosis. However, it is unclear whether alterations in lacunar morphology are present in younger populations with osteoporosis. To investigate this, we implemented a previously validated methodology to image and quantify the three-dimensional morphometries of lacunae on a large scale with ultra-high-resolution micro-computed tomography (microCT) in transiliac bone biopsies from three groups of premenopausal women: control n = 39; idiopathic osteoporosis (IOP) n = 45; idiopathic low BMD (ILBMD) n = 19. Lacunar morphometric parameters were measured in both trabecular and cortical bone such as lacunar density (Lc.N/BV), lacunar volume (Lc.V), and lacunar sphericity (Lc.Sr). These were then compared against each other and also with previously measured tissue morphometries such as bone volume density (BV/TV), trabecular separation (Tb.Sp), trabecular number (Tb.N), and others. We detected no differences in lacunar morphology between the IOP, ILBMD and healthy premenopausal women. In contrast, we did find significant differences between lacunar morphologies including Lc.N/BV, Lc. V, and Lc. Sr in cortical and trabecular regions within all three groups (p < 0.001), which was consistent with our previous findings on a subgroup of the healthy group. Furthermore, we discovered strong correlations between Lc. Sr from trabecular regions with the measured BV/TV (R = -0.90, p < 0.05). The findings and comprehensive lacunar dataset we present here will be a crucial foundation for future investigations of the relationship between osteocyte lacunar morphology and disease.
Collapse
Affiliation(s)
- Elliott Goff
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Adi Cohen
- Department of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Elizabeth Shane
- Department of Medicine, Columbia University Vagelos College of Physicians & Surgeons, New York, NY, USA
| | - Robert R Recker
- Department of Medicine, Creighton University Medical Center, Omaha, NE, USA
| | - Gisela Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
25
|
Allison H, O'Sullivan L, McNamara L. Temporal changes in cortical microporosity during estrogen deficiency associated with perilacunar resorption and osteocyte apoptosis: A pilot study. Bone Rep 2022; 16:101590. [PMID: 35663377 PMCID: PMC9156983 DOI: 10.1016/j.bonr.2022.101590] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/13/2022] [Accepted: 05/14/2022] [Indexed: 02/07/2023] Open
Abstract
Osteocytes can actively regulate bone microporosity, through either perilacunar resorption or micropetrosis following apoptosis. Osteocyte apoptosis is more prevalent in estrogen deficiency and changes in the lacunar-canalicular network of osteocytes have been reported. Temporal changes in bone mineralisation and osteocytes cellular strains occur, which might be associated with osteocyte-driven microporosity changes, although time dependant changes in bone microporosity are not yet fully understood. In this pilot study we conducted micro-CT analysis, backscatter electron imaging and histological analysis of femoral cortical bone form an ovariectomized rat model of osteoporosis to investigate whether estrogen deficiency causes temporal changes in lacunar and vascular porosity. We also assessed MMP14 expression, lacunar occupancy and mineral infilling, as indicators of perilacunar resorption and micropetrosis. We report temporal changes in cortical microporosity in estrogen deficiency. Specifically, canalicular and vascular porosity initially increased (4 weeks post-OVX), coinciding with the period of rapid bone loss, whereas in the longer term (14 weeks post-OVX) lacunar and canalicular diameter decreased. Interestingly, these changes coincided with an increased prevalence of empty lacunae and osteocyte lacunae were observed to be more circular with a mineralised border around the lacunar space. In addition we report an increase in MMP14+ osteocytes, which also suggests active matrix degradation by these cells. Together these results provide an insight into the temporal changes in cortical microporosity during estrogen deficiency and suggest the likelihood of occurrence of both perilacunar resorption and osteocyte apoptosis leading to micropetrosis. We propose that microporosity changes arise due to processes driven by distinct populations of osteocytes, which are either actively resorbing their matrix or have undergone apoptosis and are infilling lacunae by micropetrosis.
Collapse
Key Words
- BMDD, Bone mineral density distribution
- BSEM
- BSEM, Backscattered scanning electron microscopy
- BV, Bone volume
- Dm, Diameter
- Estrogen deficiency
- Lacunar
- Lc, Lacunar
- MMP, Matrix metalloproteases
- Micro-CT, Micro computed tomography
- Microporosity
- OVX, Ovariectomized
- PLR, Perilacunar resorption
- Sp, spacing
- TRAP, Tartrate-resistant acid phosphatase
- TV, Total volume
- Tb, Trabecular
- Th, Thickness
- V Ca, Vascular canal
- Vascular canals
- micro-CT
- με, Microstrain (ε ×10−6)
Collapse
Affiliation(s)
- H. Allison
- Mechanobiology and Medical Devices Research Group (MMDRG), Centre for Biomechanics Research (BioMEC), Biomedical Engineering, College of Science and Engineering, National University of Ireland Galway, Ireland
| | - L.M. O'Sullivan
- Mechanobiology and Medical Devices Research Group (MMDRG), Centre for Biomechanics Research (BioMEC), Biomedical Engineering, College of Science and Engineering, National University of Ireland Galway, Ireland
| | - L.M. McNamara
- Mechanobiology and Medical Devices Research Group (MMDRG), Centre for Biomechanics Research (BioMEC), Biomedical Engineering, College of Science and Engineering, National University of Ireland Galway, Ireland
| |
Collapse
|
26
|
Koike M, Nagao M, Iwase Y, Kaneko K, Ishijima M, Nojiri H. Clinical Efficacy of Melon GliSODin ® for the Treatment of Aging-Related Dysfunction in Motor Organs-A Double Blind, Randomized Placebo-Controlled Study. J Clin Med 2022; 11:2747. [PMID: 35628874 PMCID: PMC9143343 DOI: 10.3390/jcm11102747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/29/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Locomotive syndrome is a concept proposed in Japan involving decreased mobility due to osteoarthritis, osteoporosis, and sarcopenia. This double-blind, randomized study aimed to investigate the effects of superoxide dismutase (SOD)-rich melon extract (Melon GliSODin®) on locomotive syndrome. METHODS For 6 months, we administered oral Melon GliSODin® (500.4 mg/day) or a placebo to 24 and 22 women, respectively (aged 50-80 years), with knee or lower back discomfort or pain. Using baseline and 6-month data, changes in the Verbal Rating Scale and in subjective symptoms (determined using the Japanese Knee Osteoarthritis Measure, Locomo 25, the Roland-Morris Disability questionnaire, and the Chalder Fatigue Scale) were assessed, along with various oxidative markers, antioxidants, inflammatory markers, renal and liver function biochemical markers, bone metabolism markers, body composition, and motor function. RESULTS Oral Melon GliSODin® administration tended to be associated with a larger improvement in subjective symptom scores, a reduction in oxidative markers (malondialdehyde and diacron reactive oxygen metabolites) and tumor necrosis factor-α, and a significant increase in non-fat mass between baseline and 6 months. However, no statistically significant differences were observed between the groups for outcomes at 6 months. CONCLUSIONS Melon GliSODin® tended to improve the subjective symptoms of participants who had knee or lower back pain or discomfort. Melon GliSODin® administration may help to prevent the progression of locomotive syndrome. Future studies involving larger sample sizes and more stringent randomization protocols are needed to determine differences between the placebo and Melon GliSODin® groups.
Collapse
Affiliation(s)
- Masato Koike
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan; (M.K.); (Y.I.)
| | - Masashi Nagao
- Department of Orthopaedic Surgery, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.N.); (K.K.); (M.I.)
- Department of Orthopaedic Surgery, Medical Technology Innovation Center, Clinical Research & Trial Center, Graduate School of Health and Sports Science, Juntendo University, Tokyo 113-8421, Japan
| | - Yoshiyuki Iwase
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan; (M.K.); (Y.I.)
| | - Kazuo Kaneko
- Department of Orthopaedic Surgery, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.N.); (K.K.); (M.I.)
| | - Muneaki Ishijima
- Department of Orthopaedic Surgery, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.N.); (K.K.); (M.I.)
| | - Hidetoshi Nojiri
- Department of Orthopaedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan; (M.K.); (Y.I.)
- Department of Orthopaedic Surgery, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan; (M.N.); (K.K.); (M.I.)
| |
Collapse
|
27
|
Schoppa AM, Chen X, Ramge JM, Vikman A, Fischer V, Haffner-Luntzer M, Riegger J, Tuckermann J, Scharffetter-Kochanek K, Ignatius A. Osteoblast lineage Sod2 deficiency leads to an osteoporosis-like phenotype in mice. Dis Model Mech 2022; 15:274992. [PMID: 35394023 PMCID: PMC9118037 DOI: 10.1242/dmm.049392] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/25/2022] [Indexed: 12/19/2022] Open
Abstract
Osteoporosis is a systemic metabolic skeletal disease characterized by low bone mass and strength associated with fragility fractures. Oxidative stress, which results from elevated intracellular reactive oxygen species (ROS) and arises in the aging organism, is considered one of the critical factors contributing to osteoporosis. Mitochondrial (mt)ROS, as the superoxide anion (O2−) generated during mitochondrial respiration, are eliminated in the young organism by antioxidant defense mechanisms, including superoxide dismutase 2 (SOD2), the expression and activity of which are decreased in aging mesenchymal progenitor cells, accompanied by increased mtROS production. Using a mouse model of osteoblast lineage cells with Sod2 deficiency, we observed significant bone loss in trabecular and cortical bones accompanied by decreased osteoblast activity, increased adipocyte accumulation in the bone marrow and augmented osteoclast activity, suggestive of altered mesenchymal progenitor cell differentiation and osteoclastogenesis. Furthermore, osteoblast senescence was increased. To date, there are only a few studies suggesting a causal association between mtROS and cellular senescence in tissue in vivo. Targeting SOD2 to improve redox homeostasis could represent a potential therapeutic strategy for maintaining bone health during aging. Summary: Osteoblast-lineage specific Sod2 deficiency in mice leads to increased mtROS, impaired osteoblast function, increased adipogenesis, increased osteoclast activity and increased osteoblast senescence, resulting in bone loss.
Collapse
Affiliation(s)
- Astrid M Schoppa
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Xiangxu Chen
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Jan-Moritz Ramge
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Anna Vikman
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Verena Fischer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081 Ulm, Germany
| | - Jana Riegger
- Department of Orthopedics, Division for Biochemistry of Joint and Connective Tissue Diseases, Ulm University Medical Center, 89081 Ulm, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, Ulm University, 89081 Ulm, Germany
| | | | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
28
|
Murshid SA. Bone permeability and mechanotransduction: Some current insights into the function of the lacunar-canalicular network. Tissue Cell 2022; 75:101730. [PMID: 35032785 DOI: 10.1016/j.tice.2022.101730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 10/19/2022]
Abstract
Lacunar-canalicular (LC) permeability involves the passage of fluids, nutrients, oxygen, ions, and signalling molecules through bone tissue, facilitating the maintenance of bone vitality and function and responses to various physiological conditions and diseases. LC permeability and fluid flow-shear stress/drag force play important roles in mechanotransduction in bone tissue by inducing mechanical stimuli in osteocytes, modulating cellular functions, and determining bone adaptation. Alterations in LC structure may therefore influence the fluid flow pattern through the LC network, thereby affecting the ability of osteocytes to sense and translate mechanical signals and possibly contributing to bone remodelling. Several bone-health conditions are associated with changes in LC structure and function and may affect mechanotransduction and responses, although the mechanisms underlying these associations are still not fully understood. In this review, recent studies of LC networks, their formation and transfer mechanical stimuli, and changes in structure, functional permeability, and mechanotransduction that result from age, pathology, and mechanical loading are discussed. Additionally, applications of vibration and low-intensity pulsed ultrasound in bone healthcare and regeneration fields are also presented.
Collapse
Affiliation(s)
- Sakhr Ahmed Murshid
- Institute for Globally Distributed Open Research and Education (IGDORE); Ilmajoki Health Public Dental Clinics, Social and Health Care Services in Jalasjärvi, Ilmajoki, Kurikka, Finland.
| |
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW Osteoblasts are responsible for bone matrix production during bone development and homeostasis. Much is known about the transcriptional regulation and signaling pathways governing osteoblast differentiation. However, less is known about how osteoblasts obtain or utilize nutrients to fulfill the energetic demands associated with osteoblast differentiation and bone matrix synthesis. The goal of this review is to highlight and discuss what is known about the role and regulation of bioenergetic metabolism in osteoblasts with a focus on more recent studies. RECENT FINDINGS Bioenergetic metabolism has emerged as an important regulatory node in osteoblasts. Recent studies have begun to identify the major nutrients and bioenergetic pathways favored by osteoblasts as well as their regulation during differentiation. Here, we highlight how osteoblasts obtain and metabolize glucose, amino acids, and fatty acids to provide energy and other metabolic intermediates. In addition, we highlight the signals that regulate nutrient uptake and metabolism and focus on how energetic metabolism promotes osteoblast differentiation. Bioenergetic metabolism provides energy and other metabolites that are critical for osteoblast differentiation and activity. This knowledge contributes to a more comprehensive understanding of osteoblast biology and may inform novel strategies to modulate osteoblast differentiation and bone anabolism in patients with bone disorders.
Collapse
Affiliation(s)
- Leyao Shen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Guoli Hu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Courtney M Karner
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
30
|
Khosla S, Farr JN, Monroe DG. Cellular senescence and the skeleton: pathophysiology and therapeutic implications. J Clin Invest 2022; 132:154888. [PMID: 35104801 PMCID: PMC8803328 DOI: 10.1172/jci154888] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a fundamental aging mechanism that is currently the focus of considerable interest as a pathway that could be targeted to ameliorate aging across multiple tissues, including the skeleton. There is now substantial evidence that senescent cells accumulate in the bone microenvironment with aging and that targeting these cells prevents age-related bone loss, at least in mice. Cellular senescence also plays important roles in mediating the skeletal fragility associated with diabetes mellitus, radiation, and chemotherapy. As such, there are ongoing efforts to develop "senolytic" drugs that kill senescent cells by targeting key survival mechanisms in these cells without affecting normal cells. Because senescent cells accumulate across tissues with aging, senolytics offer the attractive possibility of treating multiple age-related comorbidities simultaneously.
Collapse
|
31
|
Emerging Therapeutic Potential of Short Mitochondrial-produced Peptides for Anabolic Osteogenesis. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-021-10353-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
32
|
Spatz JM, Ko FC, Ayturk UM, Warman ML, Bouxsein ML. RNAseq and RNA molecular barcoding reveal differential gene expression in cortical bone following hindlimb unloading in female mice. PLoS One 2021; 16:e0250715. [PMID: 34637435 PMCID: PMC8509868 DOI: 10.1371/journal.pone.0250715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/12/2021] [Indexed: 11/24/2022] Open
Abstract
Disuse-induced bone loss is seen following spinal cord injury, prolonged bed rest, and exposure to microgravity. We performed whole transcriptomic profiling of cortical bone using RNA sequencing (RNAseq) and RNA molecular barcoding (NanoString) on a hindlimb unloading (HLU) mouse model to identify genes whose mRNA transcript abundances change in response to disuse. Eleven-week old female C57BL/6 mice were exposed to ambulatory loading or HLU for 7 days (n = 8/group). Total RNA from marrow-flushed femoral cortical bone was analyzed on HiSeq and NanoString platforms. The expression of several previously reported genes associated with Wnt signaling and metabolism was altered by HLU. Furthermore, the increased abundance of transcripts, such as Pfkfb3 and Mss51, after HLU imply these genes also have roles in the cortical bone’s response to altered mechanical loading. Our study demonstrates that an unbiased approach to assess the whole transcriptomic profile of cortical bone can reveal previously unidentified mechanosensitive genes and may eventually lead to novel targets to prevent disuse-induced osteoporosis.
Collapse
Affiliation(s)
- Jordan M Spatz
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America.,University of California San Francisco School of Medicine, San Francisco, California, United States of America.,Harvard Medical School, Boston, Massachusetts, United States of America
| | - Frank C Ko
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America.,Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ugur M Ayturk
- Harvard Medical School, Boston, Massachusetts, United States of America.,Boston Children's Hospital, Boston, Massachusetts, United States of America
| | - Matthew L Warman
- Harvard Medical School, Boston, Massachusetts, United States of America.,Boston Children's Hospital, Boston, Massachusetts, United States of America
| | - Mary L Bouxsein
- Center for Advanced Orthopedic Studies, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America.,Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
33
|
Cooper ID, Brookler KH, Crofts CAP. Rethinking Fragility Fractures in Type 2 Diabetes: The Link between Hyperinsulinaemia and Osteofragilitas. Biomedicines 2021; 9:1165. [PMID: 34572351 PMCID: PMC8472634 DOI: 10.3390/biomedicines9091165] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/27/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with type 2 diabetes mellitus (T2DM) and/or cardiovascular disease (CVD), conditions of hyperinsulinaemia, have lower levels of osteocalcin and bone remodelling, and increased rates of fragility fractures. Unlike osteoporosis with lower bone mineral density (BMD), T2DM bone fragility "hyperinsulinaemia-osteofragilitas" phenotype presents with normal to increased BMD. Hyperinsulinaemia and insulin resistance positively associate with increased BMD and fragility fractures. Hyperinsulinaemia enforces glucose fuelling, which decreases NAD+-dependent antioxidant activity. This increases reactive oxygen species and mitochondrial fission, and decreases oxidative phosphorylation high-energy production capacity, required for osteoblasto/cytogenesis. Osteocytes directly mineralise and resorb bone, and inhibit mineralisation of their lacunocanalicular space via pyrophosphate. Hyperinsulinaemia decreases vitamin D availability via adipocyte sequestration, reducing dendrite connectivity, and compromising osteocyte viability. Decreased bone remodelling and micropetrosis ensues. Trapped/entombed magnesium within micropetrosis fossilisation spaces propagates magnesium deficiency (MgD), potentiating hyperinsulinaemia and decreases vitamin D transport. Vitamin D deficiency reduces osteocalcin synthesis and favours osteocyte apoptosis. Carbohydrate restriction/fasting/ketosis increases beta-oxidation, ketolysis, NAD+-dependent antioxidant activity, osteocyte viability and osteocalcin, and decreases excess insulin exposure. Osteocalcin is required for hydroxyapatite alignment, conferring bone structural integrity, decreasing fracture risk and improving metabolic/endocrine homeodynamics. Patients presenting with fracture and normal BMD should be investigated for T2DM and hyperinsulinaemia.
Collapse
Affiliation(s)
- Isabella D. Cooper
- Translational Physiology Research Group, School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK
| | - Kenneth H. Brookler
- Research Collaborator, Aerospace Medicine and Vestibular Research Laboratory, Mayo Clinic, Scottsdale, AZ 85259, USA;
| | - Catherine A. P. Crofts
- School of Public Health and Interdisciplinary Studies, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland 0627, New Zealand;
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW In this review, we provide a recent update on bioenergetic pathways in osteocytes and identify potential future areas of research interest. Studies have identified a role for regulation of bone formation and bone resorption through osteocyte mechanosensing and osteocyte secreted factors. Nevertheless, there is a paucity of studies on the bioenergetics and energy metabolism of osteocytes, which are required for the regulation of bone remodeling. RECENT FINDINGS Osteocytes are cells of the osteoblast lineage embedded in bone. The osteocyte lacunocanalicular network within the skeletal matrix is exposed to a unique hypoxic environment. Therefore, the bioenergetic requirements of these cells could differ from other bone cells due to its location in the ossified matrix and its role in bone regulation transduced by mechanical signals. Recent findings highlighted in this review provide some evidence that metabolism of these cells is dependent on their location due to the substrates present in the microenvironment and metabolic cues from stress pathways. Both glycolysis (glucose metabolism) and oxidative phosphorylation (mitochondrial dynamics, ROS generation) affect osteocyte function and viability. In this review, we provide evidence that is currently available about information regarding bioenergetics pathways in osteocytes. We discuss published studies showing a role for hypoxia-driven glucose metabolism in regulating osteocyte bioenergetics. We also provide information on various substrates that osteocytes could utilize to fuel energetic needs, namely pyruvate, amino acids, and fatty acids. This is based on some preliminary experimental evidence that is available in literature. The role of parathyroid hormone PTH and parathryoid hormone-related peptide PTHrP in bone anabolism and resorption, along with regulation of metabolic pathways in the cells of the skeletal niche, needs to be explored further. Mitochondrial metabolism has a role in osteocyte bioenergetics through substrate utilization, location of the osteocyte in the bone cortex, and mitochondrial biogenesis. While there are limitations in studying metabolic flux in traditional cell lines, there are now novel cell lines and sophisticated tools available to study osteocyte bioenergetics to help harness its potential in vivo in the future.
Collapse
Affiliation(s)
- Vivin Karthik
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA
| | - Anyonya R Guntur
- Center for Molecular Medicine, Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA.
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, USA.
- Tufts University School of Medicine, Tufts University, Boston, MA, USA.
| |
Collapse
|
35
|
Pant A, Paul E, Niebur GL, Vahdati A. Integration of mechanics and biology in computer simulation of bone remodeling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 164:33-45. [PMID: 33965425 DOI: 10.1016/j.pbiomolbio.2021.05.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/27/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022]
Abstract
Bone remodeling is a complex physiological process that spans across multiple spatial and temporal scales and is regulated by both mechanical and hormonal cues. An imbalance between bone resorption and bone formation in the process of bone remodeling may lead to various bone pathologies. One powerful and non-invasive approach to gain new insights into mechano-adaptive bone remodeling is computer modeling and simulation. Recent findings in bone physiology and advances in computer modeling have provided a unique opportunity to study the integration of mechanics and biology in bone remodeling. Our objective in this review is to critically appraise recent advances and developments and discuss future research opportunities in computational bone remodeling approaches that enable integration of mechanics and cellular and molecular pathways. Based on the critical appraisal of the relevant recent published literature, we conclude that multiscale in silico integration of personalized bone mechanics and mechanobiology combined with data science and analytics techniques offer the potential to deepen our knowledge of bone remodeling and provide ample opportunities for future research.
Collapse
Affiliation(s)
- Anup Pant
- Multi-disciplinary Mechanics and Modeling Laboratory, Department of Engineering, East Carolina University, Greenville, NC 27858, USA
| | - Elliot Paul
- Multi-disciplinary Mechanics and Modeling Laboratory, Department of Engineering, East Carolina University, Greenville, NC 27858, USA
| | - Glen L Niebur
- Tissue Mechanics Laboratory, Bioengineering Graduate Program, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Ali Vahdati
- Multi-disciplinary Mechanics and Modeling Laboratory, Department of Engineering, East Carolina University, Greenville, NC 27858, USA.
| |
Collapse
|
36
|
Watanabe K, Shibuya S, Ozawa Y, Toda T, Shimizu T. Pathological Relationship between Intracellular Superoxide Metabolism and p53 Signaling in Mice. Int J Mol Sci 2021; 22:3548. [PMID: 33805584 PMCID: PMC8037821 DOI: 10.3390/ijms22073548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/23/2021] [Accepted: 03/23/2021] [Indexed: 11/16/2022] Open
Abstract
Intracellular superoxide dismutases (SODs) maintain tissue homeostasis via superoxide metabolism. We previously reported that intracellular reactive oxygen species (ROS), including superoxide accumulation caused by cytoplasmic SOD (SOD1) or mitochondrial SOD (SOD2) insufficiency, induced p53 activation in cells. SOD1 loss also induced several age-related pathological changes associated with increased oxidative molecules in mice. To evaluate the contribution of p53 activation for SOD1 knockout (KO) (Sod1-/-) mice, we generated SOD1 and p53 KO (double-knockout (DKO)) mice. DKO fibroblasts showed increased cell viability with decreased apoptosis compared with Sod1-/- fibroblasts. In vivo experiments revealed that p53 insufficiency was not a great contributor to aging-like tissue changes but accelerated tumorigenesis in Sod1-/- mice. Furthermore, p53 loss failed to improve dilated cardiomyopathy or the survival in heart-specific SOD2 conditional KO mice. These data indicated that p53 regulated ROS-mediated apoptotic cell death and tumorigenesis but not ROS-mediated tissue degeneration in SOD-deficient models.
Collapse
Affiliation(s)
- Kenji Watanabe
- Aging Stress Response Research Project Team, National Center for Geriatrics and Gerontology, Obu 474-8511, Aichi, Japan; (K.W.); (S.S.)
| | - Shuichi Shibuya
- Aging Stress Response Research Project Team, National Center for Geriatrics and Gerontology, Obu 474-8511, Aichi, Japan; (K.W.); (S.S.)
| | - Yusuke Ozawa
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba 260-8677, Chiba, Japan; (Y.O.); (T.T.)
| | - Toshihiko Toda
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba 260-8677, Chiba, Japan; (Y.O.); (T.T.)
| | - Takahiko Shimizu
- Aging Stress Response Research Project Team, National Center for Geriatrics and Gerontology, Obu 474-8511, Aichi, Japan; (K.W.); (S.S.)
- Department of Endocrinology, Hematology and Gerontology, Chiba University Graduate School of Medicine, Chiba 260-8677, Chiba, Japan; (Y.O.); (T.T.)
| |
Collapse
|
37
|
Li Q, Cheng JC, Jiang Q, Lee WY. Role of sirtuins in bone biology: Potential implications for novel therapeutic strategies for osteoporosis. Aging Cell 2021; 20:e13301. [PMID: 33393735 PMCID: PMC7884050 DOI: 10.1111/acel.13301] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/16/2022] Open
Abstract
The decline in bone mass and bone strength and musculoskeletal problems associated with aging constitute a major challenge for affected individuals and the healthcare system globally. Sirtuins 1-7 (SIRT1-SIRT7) are a family of nicotinamide adenine dinucleotide-dependent deacetylases with remarkable abilities to promote longevity and counteract age-related diseases. Sirtuin knockout and transgenic models have provided novel insights into the function and signaling of these proteins in bone homeostasis. Studies have revealed that sirtuins play a critical role in normal skeletal development and homeostasis through their direct action on bone cells and that their dysregulation might contribute to different bone diseases. Preclinical studies have demonstrated that mice treated with sirtuin agonists show protection against age-related, postmenopausal, and immobilization-induced osteoporosis. These findings suggest that sirtuins could be potential targets for the modulation of the imbalance in bone remodeling and treatment of osteoporosis and other bone disorders. The aim of this review was to provide a comprehensive updated review of the current knowledge on sirtuin biology, focusing specifically on their roles in bone homeostasis and osteoporosis, and potential pharmacological interventions targeting sirtuins for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Qiangqiang Li
- SH Ho Scoliosis Research LaboratoryDepartment of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing UniversityThe Chinese University of Hong KongHong Kong SARChina
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARChina
| | - Jack Chun‐yiu Cheng
- SH Ho Scoliosis Research LaboratoryDepartment of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing UniversityThe Chinese University of Hong KongHong Kong SARChina
| | - Qing Jiang
- Department of Sports Medicine and Adult Reconstructive SurgeryDrum Tower Hospital affiliated to Medical School of Nanjing UniversityNanjingChina
| | - Wayne Yuk‐wai Lee
- SH Ho Scoliosis Research LaboratoryDepartment of Orthopaedics and TraumatologyThe Chinese University of Hong KongHong Kong SARChina
- Joint Scoliosis Research Center of the Chinese University of Hong Kong and Nanjing UniversityThe Chinese University of Hong KongHong Kong SARChina
- Li Ka Shing Institute of Health SciencesThe Chinese University of Hong KongHong Kong SARChina
| |
Collapse
|
38
|
Hua R, Zhang J, Riquelme MA, Jiang JX. Connexin Gap Junctions and Hemichannels Link Oxidative Stress to Skeletal Physiology and Pathology. Curr Osteoporos Rep 2021; 19:66-74. [PMID: 33403446 PMCID: PMC8174533 DOI: 10.1007/s11914-020-00645-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW The goal of this review is to provide an overview of the impact and underlying mechanism of oxidative stress on connexin channel function, and their roles in skeletal aging, estrogen deficiency, and glucocorticoid excess associated bone loss. RECENT FINDINGS Connexin hemichannel opening is increased under oxidative stress conditions, which confers a cell protective role against oxidative stress-induced cell death. Oxidative stress acts as a key contributor to aging, estrogen deficiency, and glucocorticoid excess-induced osteoporosis and impairs osteocytic network and connexin gap junction communication. This paper reviews the current knowledge for the role of oxidative stress and connexin channels in the pathogenesis of osteoporosis and physiological and pathological responses of connexin channels to oxidative stress. Oxidative stress decreases osteocyte viability and impairs the balance of anabolic and catabolic responses. Connexin 43 (Cx43) channels play a critical role in bone remodeling, mechanotransduction, and survival of osteocytes. Under oxidative stress conditions, there is a consistent reduction of Cx43 expression, while the opening of Cx43 hemichannels protects osteocytes against cell injury caused by oxidative stress.
Collapse
Affiliation(s)
- Rui Hua
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA
| | - Jingruo Zhang
- 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
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX, USA.
| |
Collapse
|
39
|
Gatti M, Beretti F, Zavatti M, Bertucci E, Ribeiro Luz S, Palumbo C, Maraldi T. Amniotic Fluid Stem Cell-Derived Extracellular Vesicles Counteract Steroid-Induced Osteoporosis In Vitro. Int J Mol Sci 2020; 22:ijms22010038. [PMID: 33375177 PMCID: PMC7792960 DOI: 10.3390/ijms22010038] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 12/19/2022] Open
Abstract
Background—Osteoporosis is characterized by defects in both quality and quantity of bone tissue, which imply high susceptibility to fractures with limitations of autonomy. Current therapies for osteoporosis are mostly concentrated on how to inhibit bone resorption but give serious adverse effects. Therefore, more effective and safer therapies are needed that even encourage bone formation. Here we examined the effect of extracellular vesicles secreted by human amniotic fluid stem cells (AFSC) (AFSC-EV) on a model of osteoporosis in vitro. Methods—human AFSC-EV were added to the culture medium of a human pre-osteoblast cell line (HOB) induced to differentiate, and then treated with dexamethasone as osteoporosis inducer. Aspects of differentiation and viability were assessed by immunofluorescence, Western blot, mass spectrometry, and histological assays. Since steroids induce oxidative stress, the levels of reactive oxygen species and of redox related proteins were evaluated. Results—AFSC-EV were able to ameliorate the differentiation ability of HOB both in the case of pre-osteoblasts and when the differentiation process was affected by dexamethasone. Moreover, the viability was increased and parallelly apoptotic markers were reduced. The presence of EV positively modulated the redox unbalance due to dexamethasone. Conclusion—these findings demonstrated that EV from hAFSC have the ability to recover precursor cell potential and delay local bone loss in steroid-related osteoporosis.
Collapse
Affiliation(s)
- Martina Gatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (M.G.); (F.B.); (M.Z.); (S.R.L.); (C.P.)
| | - Francesca Beretti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (M.G.); (F.B.); (M.Z.); (S.R.L.); (C.P.)
| | - Manuela Zavatti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (M.G.); (F.B.); (M.Z.); (S.R.L.); (C.P.)
| | - Emma Bertucci
- Department of Medical and Surgical Sciences for Mothers, Children and Adults, University of Modena and Reggio Emilia, Azienda Ospedaliero Universitaria Policlinico, Via Del Pozzo 71, 41124 Modena, Italy;
| | - Soraia Ribeiro Luz
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (M.G.); (F.B.); (M.Z.); (S.R.L.); (C.P.)
| | - Carla Palumbo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (M.G.); (F.B.); (M.Z.); (S.R.L.); (C.P.)
| | - Tullia Maraldi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy; (M.G.); (F.B.); (M.Z.); (S.R.L.); (C.P.)
- Correspondence: ; Tel.: +39-05-9422-3178; Fax: +39-05-9422-4859
| |
Collapse
|
40
|
Abstract
Osteocytes are an ancient cell, appearing in fossilized skeletal remains of early fish and dinosaurs. Despite its relative high abundance, even in the context of nonskeletal cells, the osteocyte is perhaps among the least studied cells in all of vertebrate biology. Osteocytes are cells embedded in bone, able to modify their surrounding extracellular matrix via specialized molecular remodeling mechanisms that are independent of the bone forming osteoblasts and bone-resorbing osteoclasts. Osteocytes communicate with osteoclasts and osteoblasts via distinct signaling molecules that include the RankL/OPG axis and the Sost/Dkk1/Wnt axis, among others. Osteocytes also extend their influence beyond the local bone environment by functioning as an endocrine cell that controls phosphate reabsorption in the kidney, insulin secretion in the pancreas, and skeletal muscle function. These cells are also finely tuned sensors of mechanical stimulation to coordinate with effector cells to adjust bone mass, size, and shape to conform to mechanical demands.
Collapse
Affiliation(s)
- Alexander G Robling
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA;
| | - Lynda F Bonewald
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA;
| |
Collapse
|
41
|
Osteocyte apoptosis: the roles and key molecular mechanisms in resorption-related bone diseases. Cell Death Dis 2020; 11:846. [PMID: 33046704 PMCID: PMC7552426 DOI: 10.1038/s41419-020-03059-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 01/18/2023]
Abstract
Vital osteocytes have been well known to function as an important orchestrator in the preservation of robustness and fidelity of the bone remodeling process. Nevertheless, some key pathological factors, such as sex steroid deficiency and excess glucocorticoids, and so on, are implicated in inducing a bulk of apoptotic osteocytes, subsequently resulting in resorption-related bone loss. As much, osteocyte apoptosis, under homeostatic conditions, is in an optimal state of balance tightly controlled by pro- and anti-apoptotic mechanism pathways. Importantly, there exist many essential signaling proteins in the process of osteocyte apoptosis, which has a crucial role in maintaining a homeostatic environment. While increasing in vitro and in vivo studies have established, in part, key signaling pathways and cross-talk mechanism on osteocyte apoptosis, intrinsic and complex mechanism underlying osteocyte apoptosis occurs in various states of pathologies remains ill-defined. In this review, we discuss not only essential pro- and anti-apoptotic signaling pathways and key biomarkers involved in these key mechanisms under different pathological agents, but also the pivotal role of apoptotic osteocytes in osteoclastogenesis-triggered bone loss, hopefully shedding new light on the attractive and proper actions of pharmacotherapeutics of targeting apoptosis and ensuing resorption-related bone diseases such as osteoporosis and fragility fractures.
Collapse
|
42
|
Schreurs AS, Torres S, Truong T, Moyer EL, Kumar A, Tahimic CGT, Alwood JS, Globus RK. Skeletal tissue regulation by catalase overexpression in mitochondria. Am J Physiol Cell Physiol 2020; 319:C734-C745. [PMID: 32783660 DOI: 10.1152/ajpcell.00068.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Accumulation of oxidative damage from excess reactive oxygen species (ROS) may contribute to skeletal aging and mediate adverse responses to physiological challenges. Wild-type (WT) mice and transgenic mice (male, 16 wk of age) with human catalase targeted to the mitochondria (mCAT) were analyzed for skeletal responses to the remodeling stimuli of combined hind-limb unloading and exposure to ionizing radiation (137Cs, 2 Gy). Treatment for 2 wk caused lipid peroxidation in the bones WT but not mCAT mice, showing that transgene expression mitigated oxidative stress. Ex vivo osteoblast colony growth rate was 95% greater in mCAT than WT mice and correlated with catalase activity levels (P < 0.005, r = 0.67), although terminal osteoblast and osteoclast differentiation were unaffected. mCAT mice had lower cancellous bone volume and cortical size than WT mice. Ambulatory control mCAT animals also displayed reduced cancellous and cortical structural properties compared with control WT mice. In mCAT but not WT mice, treatment caused an unexpectedly rapid radial expansion (+8% cortical area, +22% moment of inertia), reminiscent of compensatory bone growth during advancing age. In contrast, treatment caused similar structural deficits in cancellous tissue of mCAT and WT mice. In sum, mitochondrial ROS signaling via H2O2 was important for the acquisition of adult bone structure and catalase overexpression failed to protect cancellous tissue from treatment. In contrast, catabolic stimuli caused radial expansion in mCAT not WT mice, suggesting that mitochondrial ROS in skeletal cells act to suppress tissue turnover in response to remodeling challenges.
Collapse
Affiliation(s)
- Ann-Sofie Schreurs
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Universities Space Research Association, Moffett Field, California
| | - Samantha Torres
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Tiffany Truong
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Eric L Moyer
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Blue Marble Space Institute of Science, Seattle, Washington
| | - Akhhilesh Kumar
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,Universities Space Research Association, Moffett Field, California
| | - Candice G T Tahimic
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California.,KBR, Moffett Field, California
| | - Joshua S Alwood
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California
| | - Ruth K Globus
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, California
| |
Collapse
|
43
|
Mitochondrial dysfunction impairs osteogenesis, increases osteoclast activity, and accelerates age related bone loss. Sci Rep 2020; 10:11643. [PMID: 32669663 PMCID: PMC7363892 DOI: 10.1038/s41598-020-68566-2] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 06/24/2020] [Indexed: 02/07/2023] Open
Abstract
The pathogenesis of declining bone mineral density, a universal feature of ageing, is not fully understood. Somatic mitochondrial DNA (mtDNA) mutations accumulate with age in human tissues and mounting evidence suggests that they may be integral to the ageing process. To explore the potential effects of mtDNA mutations on bone biology, we compared bone microarchitecture and turnover in an ageing series of wild type mice with that of the PolgAmut/mut mitochondrial DNA ‘mutator’ mouse. In vivo analyses showed an age-related loss of bone in both groups of mice; however, it was significantly accelerated in the PolgAmut/mut mice. This accelerated rate of bone loss is associated with significantly reduced bone formation rate, reduced osteoblast population densities, increased osteoclast population densities, and mitochondrial respiratory chain deficiency in osteoblasts and osteoclasts in PolgAmut/mut mice compared with wild-type mice. In vitro assays demonstrated severely impaired mineralised matrix formation and increased osteoclast resorption by PolgAmut/mut cells. Finally, application of an exercise intervention to a subset of PolgAmut/mut mice showed no effect on bone mass or mineralised matrix formation in vitro. Our data demonstrate that mitochondrial dysfunction, a universal feature of human ageing, impairs osteogenesis and is associated with accelerated bone loss.
Collapse
|
44
|
Osteonecrosis of Femoral Head, An Overlooked Long-Term Complication after Paraquat Intoxication: A Retrospective Cohort Study. Sci Rep 2020; 10:8827. [PMID: 32483235 PMCID: PMC7264308 DOI: 10.1038/s41598-020-65756-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/29/2020] [Indexed: 01/04/2023] Open
Abstract
With increasing numbers of patients surviving acute intoxication phase, long-term complication after paraquat intoxication is a topic worth exploring, such as osteonecrosis (ON) of femoral head. We reviewed 86 paraquat-intoxicated survivors between 2000 and 2012 in Chang Gung Memorial Hospital, a 3700-bed tertiary hospital in Taiwan. With all the patients underwent same detoxification protocol in the acute stage, 17.4% of paraquat poisoning survivors developed ON of femoral head requiring surgery during follow up. Most of ON episodes occurred within 2 to 4 years after paraquat intoxication and then plateau after 6 years. ON patients exhibited higher SOFA scores than non-ON patients (2.80 ± 2.14 vs. 1.76 ± 1.52, p = 0.028). Furthermore, AKIN scores are also higher in the ON patients than non-ON patients (0.87 ± 1.13 vs. 0.38 ± 0.74, p = 0.040). Multivariate logistic regression showed higher AKIN score and higher partial pressure of carbon dioxide in the blood 48 hours after admission significantly predicted ON of femoral head after paraquat intoxication (p = 0.002 and p = 0.006 respectively). Larger studies with longer follow-up durations are warranted to confirm our finding.
Collapse
|
45
|
How Physical Activity across the Lifespan Can Reduce the Impact of Bone Ageing: A Literature Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17061862. [PMID: 32183049 PMCID: PMC7143872 DOI: 10.3390/ijerph17061862] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/05/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Bone remodeling is a lifelong process, due to the balanced activity of the osteoblasts (OBs), the bone-forming cells, and osteoclasts (OCs), the bone-resorbing cells. This equilibrium is mainly regulated by the WNT-ß-cathenin pathway and the RANK-RANKL/OPG system, respectively. Bone ageing is a process which normally occurs during life due to the imbalance between bone formation and bone resorption, potentially leading to osteoporosis. Bone loss associated with bone ageing is determined by oxidative stress, the result of the increasing production of reactive oxygen species (ROS). The promotion of physical exercise during growth increases the chances of accruing bone and delaying the onset of osteoporosis. Several studies demonstrate that physical exercise is associated with higher bone mineral density and lower fracture incidence, and the resulting bone mineral gain is maintained with ageing, despite a reduction of physical activity in adulthood. The benefits of exercise are widely recognized, thus physical activity is considered the best non-pharmacologic treatment for pathologies such as osteoporosis, obesity, diabetes and cardiovascular disease. We reviewed the physiological mechanisms which control bone remodeling, the effects of physical activity on bone health, and studies on the impact of exercise in reducing bone ageing.
Collapse
|
46
|
Targeting heme-oxidized soluble guanylate cyclase to promote osteoblast function. Drug Discov Today 2019; 25:422-429. [PMID: 31846712 DOI: 10.1016/j.drudis.2019.12.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/25/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022]
Abstract
The enzyme soluble guanylate cyclase (sGC) plays an essential part in the nitric oxide (NO) signaling pathway by binding to the prosthetic heme group; thereby catalyzing the synthesis of cyclic guanosine monophosphate (cGMP)-dependent protein kinases. Impaired NO-sGC-cGMP signaling could lead to osteoblast apoptosis by mechanisms involving the oxidative-stress-induced shift of the redox state of the reduced heme to oxidized sGC, leading to diminished heme binding to the enzyme and rendering the sGC unresponsive to NO. Targeting oxidized sGC to enhance cGMP production could restore proliferation and differentiation of osteoblasts into osteocytes. Here, the potential role of sGC activators of an oxidized or heme-free sGC as a target for promoting osteoblast function is reviewed and strategies for delivering drugs to bone are identified.
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW In perilacunar/canalicular remodeling (PLR), osteocytes dynamically resorb, and then replace, the organic and mineral components of the pericellular extracellular matrix. Given the enormous surface area of the osteocyte lacuna-canalicular network (LCN), PLR is important for maintaining homeostasis of the skeleton. The goal of this review is to examine the motivations and critical considerations for the analysis of PLR, in both in vitro and in vivo systems. RECENT FINDINGS Morphological approaches alone are insufficient to elucidate the complex mechanisms regulating PLR in the healthy skeleton and in disease. Understanding the role and regulation of PLR will require the incorporation of standardized PLR outcomes as a routine part of skeletal phenotyping, as well as the development of improved molecular and cellular outcomes. Current PLR outcomes assess PLR enzyme expression, the LCN, and bone matrix composition and organization, among others. Here, we discuss current PLR outcomes and how they have been applied to study PLR induction and suppression in vitro and in vivo. Given the role of PLR in skeletal health and disease, integrated analysis of PLR has potential to elucidate new mechanisms by which osteocytes participate in skeletal health and disease.
Collapse
Affiliation(s)
- Cristal S Yee
- Department of Orthopaedic Surgery, University of California, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Charles A Schurman
- Department of Orthopaedic Surgery, University of California, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA, 94143, USA
| | - Carter R White
- Department of Orthopaedic Surgery, University of California, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, 513 Parnassus Avenue, San Francisco, CA, 94143, USA.
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA, 94143, USA.
| |
Collapse
|
48
|
Werner SL, Sharma R, Woodruff K, Horn D, Harris SE, Gorin Y, Lee DY, Hua R, Gu S, Fajardo RJ, Habib SL, Jiang JX. CSF-1 in Osteocytes Inhibits Nox4-mediated Oxidative Stress and Promotes Normal Bone Homeostasis. JBMR Plus 2019; 4:e10080. [PMID: 32666016 DOI: 10.1002/jbm4.10080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/28/2018] [Accepted: 08/30/2018] [Indexed: 12/12/2022] Open
Abstract
CSF-1 is a key factor in regulating bone remodeling; osteocytes express CSF-1 and its receptor. Viable osteocytes are essential for bone remodeling through cell-cell contact and secretion of factors that regulate osteoblasts and osteoclasts. Increased oxidative stress contributes to osteocyte death and correlates with bone loss during aging. The NADPH oxidase Nox4 is a major source of ROS in bone. CSF-1 decreases Nox4, suggesting that CSF-1 protects against oxidative stress. Here, we show that osteocyte apoptosis previously reported in our global CSF-1KO mice is associated with increased Nox4, as well as 4-HNE expression in osteocytes. Osteocytes isolated from CSF-1KO mice were less viable and showed increased intracellular ROS, elevated NADPH oxidase activity/Nox4 protein, activation of mTOR/S6K, and downstream apoptosis signals compared with WT osteocytes. Nox4 expression was also increased in CSF-1KO osteocytes and colocalized with MitoTracker Red in mitochondria. Notably, CSF-1 inhibited Nox4 expression and apoptosis cascade signals. In additional studies, shNox4 decreased these signals in CSF-1KO osteocytes, whereas overexpression of Nox4 in WT osteocytes activated the apoptosis pathway. To determine the role of CSF-1 in osteocytes, DMP1Cre-CSF-1cKO (CSF-1cKO) mice that lack CSF-1 in osteocytes/late osteoblasts were developed. Osteocyte defects in CSF-1cKO mice overlapped with those in CSF-1KO mice, including increased apoptosis, Nox4, and 4-HNE-expressing osteocytes. CSF-1cKO mice showed unbalanced cancellous bone remodeling with decreased bone formation and resorption. Continued exposure to high Nox4/ROS levels may further compromise bone formation and predispose to bone loss and skeletal fragility. Taken together, our findings suggest a novel link between CSF-1, Nox4-derived ROS, and osteocyte survival/function that is crucial for osteocyte-mediated bone remodeling. Results reveal new mechanisms by which CSF-1/oxidative stress regulate osteocyte homeostasis, which may lead to therapeutic strategies to improve skeletal health in aging. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Sherry L Werner
- Department of Pathology University of Texas Health Science Center at San Antonio TX USA
| | - Ramaswamy Sharma
- Department of Cell Systems and Anatomy University of Texas Health Science Center at San Antonio TX USA
| | - Kathleen Woodruff
- Department of Pathology University of Texas Health Science Center at San Antonio TX USA
| | - Diane Horn
- Department of Pathology University of Texas Health Science Center at San Antonio TX USA
| | - Stephen E Harris
- Department of Periodontics University of Texas Health Science Center at San Antonio TX USA
| | - Yves Gorin
- Department of Medicine/Nephrology University of Texas Health Science Center at San Antonio TX USA
| | - Doug-Yoon Lee
- Department of Medicine/Nephrology University of Texas Health Science Center at San Antonio TX USA
| | - Rui Hua
- Department of Biochemistry and Structural Biology University of Texas Health Science Center at San Antonio TX USA
| | - Sumin Gu
- Department of Biochemistry and Structural Biology University of Texas Health Science Center at San Antonio TX USA
| | - Roberto J Fajardo
- Department of Orthopedics University of Texas Health Science Center at San Antonio TX USA
| | - Samy L Habib
- South Texas Veterans Health Care and Department of Cell Systems and Anatomy University of Texas Health Science Center at San Antonio TX USA
| | - Jean X Jiang
- Department of Biochemistry and Structural Biology University of Texas Health Science Center at San Antonio TX USA
| |
Collapse
|
49
|
Tiede-Lewis LM, Dallas SL. Changes in the osteocyte lacunocanalicular network with aging. Bone 2019; 122:101-113. [PMID: 30743014 PMCID: PMC6638547 DOI: 10.1016/j.bone.2019.01.025] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 01/28/2019] [Indexed: 12/22/2022]
Abstract
Osteoporosis is an aging-related disease of reduced bone mass that is particularly prevalent in post-menopausal women, but also affects the aged male population and is associated with increased fracture risk. Osteoporosis is the result of an imbalance whereby bone formation by osteoblasts no longer keeps pace with resorption of bone by osteoclasts. Osteocytes are the most abundant cells in bone and, although previously thought to be quiescent, they are now known to be active, multifunctional cells that play a key role in the maintenance of bone mass by regulating both osteoblast and osteoclast activity. They are also thought to regulate bone mass through their role as mechanoresponsive cells in bone that coordinate adaptive responses to mechanical loading. Osteocytes form an extensive interconnected network throughout the mineralized bone matrix and receive their nutrients as well as hormones and signaling factors through the lacunocanalicular system. Several studies have shown that the extent and connectivity of the lacunocanalicular system and osteocyte networks degenerates in aged humans as well as in animal models of aging. It is also known that the bone anabolic response to loading is decreased with aging. This review summarizes recent research on the degenerative changes that occur in osteocytes and their lacunocanalicular system as a result of aging and discusses the implications for skeletal health and homeostasis as well as potential mechanisms that may underlie these degenerative changes. Since osteocytes are such key regulators of skeletal homeostasis, maintaining the health of the osteocyte network would seem critical for maintenance of bone health. Therefore, a more complete understanding of the structure and function of the osteocyte network, its lacunocanalicular system, and the degenerative changes that occur with aging should lead to advances in our understanding of age related bone loss and potentially lead to improved therapies.
Collapse
Affiliation(s)
- LeAnn M Tiede-Lewis
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO 64108, United States of America
| | - Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO 64108, United States of America.
| |
Collapse
|
50
|
Sherk VD, Rosen CJ. Senescent and apoptotic osteocytes and aging: Exercise to the rescue? Bone 2019; 121:255-258. [PMID: 30735796 PMCID: PMC6459182 DOI: 10.1016/j.bone.2019.02.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 02/04/2019] [Accepted: 02/05/2019] [Indexed: 12/11/2022]
Abstract
Osteocytes are the most prevalent cell in the skeleton and are the master regulator of bone remodeling. Despite the understanding that osteocytes have a multiyear lifespan, and some factors induce apoptosis in osteocytes, much less is understood about the induction and consequences of osteocyte senescence. Filling these gaps in knowledge will provide novel approaches to slowing age-related bone loss and preventing fragility fractures. The purpose of this review is to examine the roles of senescence and apoptosis in osteocytes in age-related bone loss. Based on evidence that exercise can prevent senescence in skeletal muscle, we provide a novel hypothesis by which exercise can prolong skeletal health.
Collapse
Affiliation(s)
- Vanessa D Sherk
- Department of Medicine, Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America.
| | - Clifford J Rosen
- Tufts University School of Medicine, Maine Medical Center Research Institute Scarborough, ME, United States of America
| |
Collapse
|