1
|
Yang Y, Xie S, Kang Y, Li X, Chen H, Shan X, Cai Z. Analysis of changes in bone mineral density and cortical bone thickness after reconstruction of the mandible with fibula, is condyle preservation a critical influence factor? Clin Oral Investig 2024; 28:622. [PMID: 39482400 DOI: 10.1007/s00784-024-06027-0] [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: 04/20/2024] [Accepted: 10/28/2024] [Indexed: 11/03/2024]
Abstract
OBJECTIVE This study aimed to investigate the features of bone mineral density (BMD) and cortical bone thickness in grafted fibula. MATERIALS AND METHODS Eighty-six patients who underwent mandibular reconstruction using vascularized fibula flaps were enrolled, all of whom were followed up at 3, 6, and 12 months after surgery. The patients were grouped according to whether the condyle was preserved. BMD and cortical bone thickness were also measured. RESULTS Condyle-preserved group consisted of 65 patients and condyle-unpreserved group consisted of 21 patients. There was a significant correlation between thickness and BMD, which was significantly correlated with follow-up time. One year after surgery, the BMD of the condyle-preserved group decreased from 1029.61 ± 156.01 mg/cm3 to 978.6 ± 141.90 mg/cm3, and thickness decreased from 3.29 ± 0.65 mm to 2.72 ± 0.72 mm. BMD of the condyle-unpreserved group decreased from 1062.21 ± 126.01 mg/cm3 to 851.26 ± 144.38 mg/cm3, and thickness decreased from 3.46 ± 0.89 mm to 2.56 ± 0.73 mm. In the condyle-preserved and unpreserved groups, the absorption rates of BMD were 3.29 ± 11.97% and 17.09 ± 12.42% at 12 months, respectively, and the rate of thickness was 20.7 ± 11.45% and 26.39 ± 12.23% at 12 months, respectively. CONCLUSION BMD and thickness showed a decreasing trend over time. Preserving the condyle can slow bone resorption of the fibula. Regarding implant restoration, we recommend doctors to perform the treatment within 6-12 months after surgery in order to effectively manage bone resorption. CLINICAL RELEVANCE Our study found that condylar preservation can decrease the absorption rate of BMD and cortical bone thickness, helping doctors make better clinical decisions. TRIAL REGISTRATION NUMBER ChiCTR2300069661 (March 22, 2023).
Collapse
Affiliation(s)
- Yihui Yang
- Department of Oral and Maxillofacial Surgery, National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Peking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Shang Xie
- Department of Oral and Maxillofacial Surgery, National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Peking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Yifan Kang
- Department of Oral and Maxillofacial Surgery, National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Peking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Xiangyu Li
- Department of Oral and Maxillofacial Surgery, National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Peking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Huimin Chen
- Department of Oral and Maxillofacial Surgery, National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Peking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
| | - Xiaofeng Shan
- Department of Oral and Maxillofacial Surgery, National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Peking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
| | - Zhigang Cai
- Department of Oral and Maxillofacial Surgery, National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials, Peking University School and Hospital of Stomatology, No.22, Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| |
Collapse
|
2
|
Restier-Verlet J, Ferlazzo ML, Granzotto A, Al-Choboq J, Bellemou C, Estavoyer M, Lecomte F, Bourguignon M, Pujo-Menjouet L, Foray N. Accelerated Aging Effects Observed In Vitro after an Exposure to Gamma-Rays Delivered at Very Low and Continuous Dose-Rate Equivalent to 1-5 Weeks in International Space Station. Cells 2024; 13:1703. [PMID: 39451221 PMCID: PMC11506070 DOI: 10.3390/cells13201703] [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: 09/26/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/26/2024] Open
Abstract
Radiation impacting astronauts in their spacecraft come from a "bath" of high-energy rays (0.1-0.5 mGy per mission day) that reaches deep tissues like the heart and bones and a "stochastic rain" of low-energy particles from the shielding and impacting surface tissues like skin and lenses. However, these two components cannot be reproduced on Earth together. The MarsSimulator facility (Toulouse University, France) emits, thanks to a bag containing thorium salts, a continuous exposure of 120 mSv/y, corresponding to that prevailing in the International Space Station (ISS). By using immunofluorescence, we assessed DNA double-strand breaks (DSB) induced by 1-5 weeks exposure in ISS of human tissues evoked above, identified at risk for space exploration. All the tissues tested elicited DSBs that accumulated proportionally to the dose at a tissue-dependent rate (about 40 DSB/Gy for skin, 3 times more for lens). For the lens, bones, and radiosensitive skin cells tested, perinuclear localization of phosphorylated forms of ataxia telangiectasia mutated protein (pATM) was observed during the 1st to 3rd week of exposure. Since pATM crowns were shown to reflect accelerated aging, these findings suggest that a low dose rate of 120 mSv/y may accelerate the senescence process of the tested tissues. A mathematical model of pATM crown formation and disappearance has been proposed. Further investigations are needed to document these results in order to better evaluate the risks related to space exploration.
Collapse
Affiliation(s)
- Juliette Restier-Verlet
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, 28 Rue Laennec, 69008 Lyon, France; juliette.restier-- (J.R.-V.); (M.L.F.); (A.G.); (J.A.-C.); (C.B.); (M.B.)
| | - Mélanie L. Ferlazzo
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, 28 Rue Laennec, 69008 Lyon, France; juliette.restier-- (J.R.-V.); (M.L.F.); (A.G.); (J.A.-C.); (C.B.); (M.B.)
| | - Adeline Granzotto
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, 28 Rue Laennec, 69008 Lyon, France; juliette.restier-- (J.R.-V.); (M.L.F.); (A.G.); (J.A.-C.); (C.B.); (M.B.)
| | - Joëlle Al-Choboq
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, 28 Rue Laennec, 69008 Lyon, France; juliette.restier-- (J.R.-V.); (M.L.F.); (A.G.); (J.A.-C.); (C.B.); (M.B.)
| | - Camélia Bellemou
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, 28 Rue Laennec, 69008 Lyon, France; juliette.restier-- (J.R.-V.); (M.L.F.); (A.G.); (J.A.-C.); (C.B.); (M.B.)
| | - Maxime Estavoyer
- Universite Claude Bernard Lyon 1, CNRS, Ecole Centrale de Lyon, INSA Lyon, Université Jean Monnet, ICJ UMR5208, Inria, 69622 Villeurbanne, France; (M.E.); (F.L.); (L.P.-M.)
| | - Florentin Lecomte
- Universite Claude Bernard Lyon 1, CNRS, Ecole Centrale de Lyon, INSA Lyon, Université Jean Monnet, ICJ UMR5208, Inria, 69622 Villeurbanne, France; (M.E.); (F.L.); (L.P.-M.)
| | - Michel Bourguignon
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, 28 Rue Laennec, 69008 Lyon, France; juliette.restier-- (J.R.-V.); (M.L.F.); (A.G.); (J.A.-C.); (C.B.); (M.B.)
- Département de Biophysique et Médecine Nucléaire, Université Paris Saclay, Versailles St. Quentin-en-Yvelines, 78035 Versailles, France
| | - Laurent Pujo-Menjouet
- Universite Claude Bernard Lyon 1, CNRS, Ecole Centrale de Lyon, INSA Lyon, Université Jean Monnet, ICJ UMR5208, Inria, 69622 Villeurbanne, France; (M.E.); (F.L.); (L.P.-M.)
| | - Nicolas Foray
- INSERM U1296 Unit “Radiation: Defense, Health, Environment”, 28 Rue Laennec, 69008 Lyon, France; juliette.restier-- (J.R.-V.); (M.L.F.); (A.G.); (J.A.-C.); (C.B.); (M.B.)
| |
Collapse
|
3
|
Oommen AM, Stafford P, Joshi L. Profiling muscle transcriptome in mice exposed to microgravity using gene set enrichment analysis. NPJ Microgravity 2024; 10:94. [PMID: 39367013 PMCID: PMC11452717 DOI: 10.1038/s41526-024-00434-z] [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: 02/09/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024] Open
Abstract
Space exploration's advancement toward long-duration missions prompts intensified research on physiological effects. Despite adaptive physiological stability in some variables, persistent changes affect genome integrity, immune response, and cognitive function. Our study, utilizing multi-omics data from GeneLab, provides crucial insights investigating muscle atrophy during space mission. Leveraging NASA GeneLab's data resources, we apply systems biology-based analyses, facilitating comprehensive understanding and enabling meta-analysis. Through transcriptomics, we establish a reference profile of biological processes underlying muscle atrophy, crucial for intervention development. We emphasize the often-overlooked role of glycosylation in muscle atrophy. Our research sheds light on fundamental molecular mechanisms, bridging gaps between space research and terrestrial conditions. This study underscores the importance of interdisciplinary collaboration and data-sharing initiatives like GeneLab in advancing space medicine research.
Collapse
Affiliation(s)
- Anup Mammen Oommen
- Advanced Glycoscience Research Cluster (AGRC), University of Galway, Galway, Ireland
| | - Phillip Stafford
- Arizona State University, School of Life Sciences, Biodesign Institute, Arizona, USA
| | - Lokesh Joshi
- Advanced Glycoscience Research Cluster (AGRC), University of Galway, Galway, Ireland.
- Aquila Bioscience, University of Galway, Galway, Ireland.
| |
Collapse
|
4
|
Gogichaeva KK, Ogneva IV. Administration of Essential Phospholipids Prevents Drosophila Melanogaster Oocytes from Responding to Change in Gravity. Cells 2024; 13:1593. [PMID: 39329774 PMCID: PMC11430006 DOI: 10.3390/cells13181593] [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: 08/28/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024] Open
Abstract
The aim of this study was to prevent initial changes in Drosophila melanogaster oocytes under simulated weightlessness and hypergravity at the 2 g level. Phospholipids with polyunsaturated fatty acids in the tail groups (essential phospholipids) at a concentration of 500 mg/kg of nutrient medium were used as a protective agent. Cell stiffness was determined using atomic force microscopy, the change in the oocytes' area was assessed as a mark of deformation, and the contents of cholesterol and neutral lipids were determined using fluorescence microscopy. The results indicate that the administration of essential phospholipids leads to a decrease in the cholesterol content in the oocytes' membranes by 13% (p < 0.05). The stiffness of oocytes from flies that received essential phospholipids was 14% higher (p < 0.05) and did not change during 6 h of simulated weightlessness or hypergravity, and neither did the area, which indicates their resistance to deformation. Moreover, the exposure to simulated weightlessness and hypergravity of oocytes from flies that received a standard nutrient medium led to a more intense loss of cholesterol from cell membranes after 30 min by 13% and 18% (p < 0.05), respectively, compared to the control, but essential phospholipids prevented this effect.
Collapse
Affiliation(s)
- Ksenia K Gogichaeva
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 76 a, Khoroshevskoyoe Shosse, 123007 Moscow, Russia
| | - Irina V Ogneva
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 76 a, Khoroshevskoyoe Shosse, 123007 Moscow, Russia
- Medical and Biological Physics Department, I.M. Sechenov First Moscow State Medical University, 8-2 Trubetskaya Street, 119991 Moscow, Russia
| |
Collapse
|
5
|
Zhang J, Huang Y, Bai N, Sun Y, Li K, Ruan H, Yan B, Hu J, Zhang N, Zhang H, Chen W, Fan D. Spirulina platensis components mitigate bone density loss induced by simulated microgravity: A mechanistic insight. Food Chem 2024; 463:141361. [PMID: 39340915 DOI: 10.1016/j.foodchem.2024.141361] [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: 06/13/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024]
Abstract
In microgravity conditions, the consumption of Spirulina platensis (SP) as a renewable food source shows promise in mitigating osteoporosis due to its high nutritional content photosynthetic efficiency, environmental adaptability and positive effects on bone density, though the exact bioactive components and mechanisms remain unclear. Using a hindlimb suspension (HLS) model, this study investigated SP components: proteins (SPP), polysaccharides (SPS), lipids (SPL), and residue (SPR) on bone density and metabolism. Findings revealed that SPP and SPS significantly enhanced bone density and reduced oxidative stress. Activation of the FoxO3/Wnt/β-catenin pathway reduced FoxO3a expression and increased Wnt signaling molecules and β-catenin protein, boosting bone formation. Moreover, these components promoted beneficial gut bacteria like Turicibacter, reduced the Firmicutes-to-Bacteroidetes ratio, and enhanced SCFAs production, crucial for bone health. This study emphasized the potential of Spirulina nutrients in addressing space-induced osteoporosis and developing functional foods for long-term space missions.
Collapse
Affiliation(s)
- Jian Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yaxin Huang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Ning Bai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; Department of Endocrinology, Affiliated Hospital of Jiangnan University, Wuxi 214062, Jiangsu Province, China
| | - Yuying Sun
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Ke Li
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Huan Ruan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bowen Yan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jian Hu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; Key Laboratory of Chinese Cuisine Intangible Cultural Heritage Technology Inheritance, Ministry of Culture and Tourism, College of Tourism and Culinary Science, Yangzhou University, Yangzhou 225127, China
| | - Nana Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Daming Fan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
6
|
Wang N, Zuo Z, Meng T, Liu Y, Zheng X, Ma Y. Salidroside alleviates simulated microgravity-induced bone loss by activating the Nrf2/HO-1 pathway. J Orthop Surg Res 2024; 19:531. [PMID: 39218922 PMCID: PMC11367893 DOI: 10.1186/s13018-024-05030-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Bone loss caused by microgravity exposure presents a serious threat to the health of astronauts, but existing treatment strategies have specific restrictions. This research aimed to investigate whether salidroside (SAL) can mitigate microgravity-induced bone loss and its underlying mechanism. METHODS In this research, we used hindlimb unloading (HLU) and the Rotary Cell Culture System (RCCS) to imitate microgravity in vivo and in vitro. RESULTS The results showed that salidroside primarily enhances bone density, microstructure, and biomechanical properties by stimulating bone formation and suppressing bone resorption, thereby preserving bone mass in HLU rats. In MC3T3-E1 cells cultured under simulated microgravity in rotary wall vessel bioreactors, the expression of osteogenic genes significantly increased after salidroside administration, indicating that salidroside can promote osteoblast differentiation under microgravity conditions. Furthermore, the Nrf2 inhibitor ML385 diminished the therapeutic impact of salidroside on microgravity-induced bone loss. Overall, this research provides the first evidence that salidroside can mitigate bone loss induced by microgravity exposure through stimulating the Nrf2/HO-1 pathway. CONCLUSION These findings indicate that salidroside has great potential for treating space-related bone loss in astronauts and suggest that Nrf2/HO-1 is a viable target for counteracting microgravity-induced bone damage.
Collapse
Affiliation(s)
- Nan Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhuan Zuo
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tong Meng
- Department of Orthopedic Surgery, Xi'an City First Hospital, Xi'an, China
| | - Yuliang Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiwei Zheng
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yongsheng Ma
- Department of Orthopedic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
7
|
Swain P, Caplan N, Hughes L. Blood flow restriction: The acute effects of body tilting and reduced gravity analogues on limb occlusion pressure. Exp Physiol 2024. [PMID: 39153209 DOI: 10.1113/ep091874] [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: 03/11/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
Blood flow restriction (BFR) has been identified as a potential countermeasure to mitigate physiological deconditioning during spaceflight. Guidelines recommend that tourniquet pressure be prescribed relative to limb occlusion pressure (LOP); however, it is unclear whether body tilting or reduced gravity analogues influence LOP. We examined LOP at the leg and arm during supine bedrest and bodyweight suspension (BWS) at 6° head-down tilt (HDT), horizontal (0°), and 9.5° head-up tilt (HUT) positions. Twenty-seven adults (age, 26 ± 5 years; height, 1.75 ± 0.08 m; body mass, 73 ± 12 kg) completed all tilts during bedrest. A subgroup (n = 15) additionally completed the tilts during BWS. In each position, LOP was measured twice in the leg and arm using the Delfi Personalized Tourniquet System after 5 min of rest and again after a further 5 min. The LOP at the leg increased significantly from 6° HDT to 9.5° HUT in bedrest and BWS by 9-15 mmHg (Cohen's d = 0.7-1.0). Leg LOP was significantly higher during BWS at horizontal and 9.5° HUT postures relative to the same angles during bedrest by 8 mmHg (Cohen's d = 0.6). Arm LOP remained unchanged between body tilts and analogues. Intraclass correlation coefficients for LOP measurements taken after an initial and subsequent 5 min rest period in all conditions ranged between 0.91-0.95 (leg) and 0.83-0.96 (arm). It is advised that LOP be measured before the application of a vascular occlusion in the same body tilt/setting to which it is applied to minimize discrepancies between the actual and prescribed tourniquet pressure.
Collapse
Affiliation(s)
- Patrick Swain
- Aerospace Medicine and Rehabilitation Laboratory, Department of Sport, Exercise and Rehabilitation, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Nick Caplan
- Aerospace Medicine and Rehabilitation Laboratory, Department of Sport, Exercise and Rehabilitation, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Luke Hughes
- Aerospace Medicine and Rehabilitation Laboratory, Department of Sport, Exercise and Rehabilitation, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| |
Collapse
|
8
|
Wei Y, Wu B, Liu M, Cui CP. The Discovery of a Specific CKIP-1 Ligand for the Potential Treatment of Disuse Osteoporosis. Int J Mol Sci 2024; 25:8870. [PMID: 39201556 PMCID: PMC11354310 DOI: 10.3390/ijms25168870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 09/02/2024] Open
Abstract
Bone homeostasis relies on the delicate balance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption. The casein kinase 2 interacting protein-1 (CKIP-1), a specific CK2α subunit-interacting protein, has been documented as one of the crucial negative regulators of bone formation. CKIP-1 siRNA therapy has constraints that limit its use in clinical applications. Therefore, it is necessary to explore effective targeting strategies for CKIP-1. In this study, we observed an upregulation of CKIP-1 protein expression in the microgravity environment, while its ubiquitination levels decreased. We further investigated the interaction between CKIP-1 and VHL and found that VHL enhanced CKIP-1 degradation through the ubiquitylation-proteasome system (UPS). Additionally, we discovered a small molecule ligand, named C77, through DNA-encoded library (DEL) screening, which binds to CKIP-1 both in vivo and in vitro, as confirmed by Surface Plasmon Resonance (SPR) and the Cellular Thermal shift assay (CETSA), respectively. Our findings demonstrated the potential of VHL and C77 as guiding factors in the development of CKIP-1-based Proteolysis-Targeting Chimeras (PROTACs), which could be future therapeutic interventions in disuse osteoporosis.
Collapse
Affiliation(s)
| | | | | | - Chun-Ping Cui
- State Key Laboratory of Medical Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China; (Y.W.); (B.W.); (M.L.)
| |
Collapse
|
9
|
Paladugu P, Ong J, Kumar R, Waisberg E, Zaman N, Kamran SA, Tavakkoli A, Rivolta MC, Nelson N, Yoo T, Douglas VP, Douglas K, Song A, Tso H, Lee AG. Lower body negative pressure as a research tool and countermeasure for the physiological effects of spaceflight: A comprehensive review. LIFE SCIENCES IN SPACE RESEARCH 2024; 42:8-16. [PMID: 39067995 DOI: 10.1016/j.lssr.2024.03.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/07/2024] [Accepted: 03/31/2024] [Indexed: 07/30/2024]
Abstract
Lower Body Negative Pressure (LBNP) redistributes blood from the upper body to the lower body. LBNP may prove to be a countermeasure for the multifaceted physiological changes endured by astronauts during spaceflight related to cephalad fluid shift. Over more than five decades, beginning with the era of Skylab, advancements in LBNP technology have expanded our understanding of neurological, ophthalmological, cardiovascular, and musculoskeletal adaptations in space, with particular emphasis on mitigating issues such as bone loss. To date however, no comprehensive review has been conducted that chronicles the evolution of this technology or elucidates the broad-spectrum potential of LBNP in managing the diverse physiological challenges encountered in the microgravity environment. Our study takes a chronological perspective, systematically reviewing the historical development and application of LBNP technology in relation to the various pathophysiological impacts of spaceflight. The primary objective is to illustrate how this technology, as it has evolved, offers an increasingly sophisticated lens through which to interpret the systemic effects of space travel on human physiology. We contend that the insights gained from LBNP studies can significantly aid in formulating targeted and effective countermeasures to ensure the health and safety of astronauts. Ultimately, this paper aspires to promote a more cohesive understanding of the broad applicability of LBNP as a countermeasure against multiple bodily effects of space travel, thereby contributing to a safer and more scientifically informed approach to human space exploration.
Collapse
Affiliation(s)
- Phani Paladugu
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States; Sidney Kimmel Medical College, Philadelphia, PA, United States
| | - Joshua Ong
- Michigan Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Rahul Kumar
- University of Miami, Coral Gables, FL, United States
| | - Ethan Waisberg
- University College Dublin School of Medicine, Belfield, Dublin, Ireland
| | - Nasif Zaman
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, United States
| | - Sharif Amit Kamran
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, United States
| | - Alireza Tavakkoli
- Human-Machine Perception Laboratory, Department of Computer Science and Engineering, University of Nevada, Reno, Reno, NV, United States
| | | | - Nicolas Nelson
- Sidney Kimmel Medical College, Philadelphia, PA, United States
| | - Taehwan Yoo
- Sidney Kimmel Medical College, Philadelphia, PA, United States
| | | | - Konstantinos Douglas
- First Department of Ophthalmology, Gennimatas General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| | - Amy Song
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Hanna Tso
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, United States
| | - Andrew G Lee
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, United States; Department of Ophthalmology, Blanton Eye Institute, Houston Methodist Hospital, Houston, TX, United States; The Houston Methodist Research Institute, Houston Methodist Hospital, Houston, TX, United States; Departments of Ophthalmology, Neurology, and Neurosurgery, Weill Cornell Medicine, New York, NY, United States; Department of Ophthalmology, University of Texas Medical Branch, Galveston, TX, United States; University of Texas MD Anderson Cancer Center, Houston, TX, United States; Texas A&M College of Medicine, TX, United States; Department of Ophthalmology, The University of Iowa Hospitals and Clinics, Iowa City, IA, United States.
| |
Collapse
|
10
|
Jullienne A, Malo M, Shaw K, Zheng Y, Johnston JD, Kontulainen S, Chilibeck PD, Dadachova E, Obenaus A, Sarty GE. Musculoskeletal perturbations of deep space radiation: Assessment using a Gateway MRI. LIFE SCIENCES IN SPACE RESEARCH 2024; 42:74-83. [PMID: 39067994 DOI: 10.1016/j.lssr.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 07/30/2024]
Abstract
Human space exploration expansion from Low-Earth Orbit to deep space is accelerating the need to monitor and address the known health concerns related to deep space radiation. The human musculoskeletal system is vulnerable to these risks (alongside microgravity) and its health reflects the well-being of other body systems. Multiparametric magnetic resonance imaging (MRI) is an important approach for assessing temporal physiological changes in the musculoskeletal system. We propose that ultra-low-field MRI provides an optimal low Size Weight and Power (SwaP) solution for non-invasively monitoring muscle and bone changes on the planned Gateway lunar space station. Our proposed ultra-low-field Gateway MRI meets low SWaP design specifications mandated by limited room in the lunar space station. This review summarizes the current state of our knowledge on musculoskeletal consequences of spaceflight, especially with respect to radiation, and then elaborates how MRI can be used to monitor the deleterious effects of space travel and the efficacy of putative countermeasures. We argue that an ultra-low-field MRI in cis-lunar space on the Gateway can provide valuable research and medical insights into the effects of deep space radiation exposure on astronauts. Such an MRI would also allow the development of imaging protocols that would facilitate Earth-bound teams to monitor space personnel musculoskeletal changes during future interplanetary spaceflight. It will especially have a role in monitoring countermeasures, such as the use of melanin, in protecting space explorers.
Collapse
Affiliation(s)
- Amandine Jullienne
- School of Medicine, University of California Irvine, 1001 Health Sciences Rd, Irvine, CA 92617, United States
| | - Mackenzie Malo
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK S7N 5E5, Canada
| | - Keely Shaw
- College of Kinesiology, University of Saskatchewan, 87 Campus Dr, Saskatoon, SK S7N 5B2, Canada
| | - Yuwen Zheng
- College of Kinesiology, University of Saskatchewan, 87 Campus Dr, Saskatoon, SK S7N 5B2, Canada
| | - James D Johnston
- College of Engineering, University of Saskatchewan, 57 Campus Dr, Saskatoon, SK S7N 5A9, Canada
| | - Saija Kontulainen
- College of Kinesiology, University of Saskatchewan, 87 Campus Dr, Saskatoon, SK S7N 5B2, Canada
| | - Philip D Chilibeck
- College of Kinesiology, University of Saskatchewan, 87 Campus Dr, Saskatoon, SK S7N 5B2, Canada
| | - Ekaterina Dadachova
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK S7N 5E5, Canada
| | - Andre Obenaus
- School of Medicine, University of California Irvine, 1001 Health Sciences Rd, Irvine, CA 92617, United States; School of Medicine, University of California Riverside, United States
| | - Gordon E Sarty
- Space MRI Lab, University of Saskatchewan, QuanTA Centre, 9 Campus Dr, Saskatoon, SK S7N 5A5, Canada.
| |
Collapse
|
11
|
van Loon JJWA, Berezovska OP, Bervoets TJM, Montufar-Solis D, Semeins CM, Zandieh-Doulabi B, Rodionova PNV, Duke J, Veldhuijzen JP. Growth and mineralization of fetal mouse long bones under microgravity and daily 1 g gravity exposure. NPJ Microgravity 2024; 10:80. [PMID: 39060264 PMCID: PMC11282293 DOI: 10.1038/s41526-024-00421-4] [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: 12/22/2023] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
In a previous Space Shuttle/Spacelab experiment (STS-42), we observed direct responses of isolated fetal mouse long bones to near weightlessness. This paper aimed to verify those results and study the effects of daily 1×g exposure during microgravity on the growth and mineralization of these bones. Two experiments were conducted: one on an American Space Shuttle mission (IML-2 on STS-65) and another on a Russian Bio-Cosmos flight (Bion-10 on Cosmos-2229). Despite differences in hardware, both used 17-day-old fetal mouse metatarsals cultured for 4 days. Results showed reduced proteoglycan content under microgravity compared to 1×g conditions, with no main differences in other cellular structures. While the overall metatarsal length was unaffected, the length increase of the mineralized diaphysis was significantly reduced under microgravity. Daily 1×g exposure for at least 6 h abolished the microgravity-induced reduction in cartilage mineralization, indicating the need for long-duration exposure to 1×g as an in-flight countermeasure using artificial gravity.
Collapse
Affiliation(s)
- Jack J W A van Loon
- Department of Oral Biology, Section Oral Cell Biology, ACTA-Vrije Universiteit, Amsterdam, The Netherlands.
| | - Olga P Berezovska
- Department of Radiobiology and Radioecology, Institute for Nuclear Research of National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Theodorus J M Bervoets
- Department of Oral Biology, Section Oral Cell Biology, ACTA-Vrije Universiteit, Amsterdam, The Netherlands
| | - Dina Montufar-Solis
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Cor M Semeins
- Department of Oral Biology, Section Oral Cell Biology, ACTA-Vrije Universiteit, Amsterdam, The Netherlands
| | - Behrouz Zandieh-Doulabi
- Department of Oral Biology, Section Oral Cell Biology, ACTA-Vrije Universiteit, Amsterdam, The Netherlands
| | - P Natalia V Rodionova
- Schmalhausen Institute for Zoology, National Academy of Sciences Ukraine, Kiev, Ukraine
| | - Jackie Duke
- Department of Orthodontics & Dentofacial Orthopedics, University of Texas Health Science Center, Houston, TX, USA
| | - J Paul Veldhuijzen
- Department of Oral Biology, Section Oral Cell Biology, ACTA-Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Beckett LJ, Williams PM, Toh LS, Hessel V, Gerstweiler L, Fisk I, Toronjo-Urquiza L, Chauhan VM. Advancing insights into microgravity induced muscle changes using Caenorhabditis elegans as a model organism. NPJ Microgravity 2024; 10:79. [PMID: 39060303 PMCID: PMC11282318 DOI: 10.1038/s41526-024-00418-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Spaceflight presents significant challenges to the physiological state of living organisms. This can be due to the microgravity environment experienced during long-term space missions, resulting in alterations in muscle structure and function, such as atrophy. However, a comprehensive understanding of the adaptive mechanisms of biological systems is required to devise potential solutions and therapeutic approaches for adapting to spaceflight conditions. This review examines the current understanding of the challenges posed by spaceflight on physiological changes, alterations in metabolism, dysregulation of pathways and the suitability and advantages of using the model organism Caenorhabditis elegans nematodes to study the effects of spaceflight. Research has shown that changes in the gene and protein composition of nematodes significantly occur across various larval stages and rearing environments, including both microgravity and Earth gravity settings, often mirroring changes observed in astronauts. Additionally, the review explores significant insights into the fundamental metabolic changes associated with muscle atrophy and growth, which could lead to the development of diagnostic biomarkers and innovative techniques to prevent and counteract muscle atrophy. These insights not only advance our understanding of microgravity-induced muscle atrophy but also lay the groundwork for the development of targeted interventions to mitigate its effects in the future.
Collapse
Affiliation(s)
- Laura J Beckett
- School of Pharmacy, University of Nottingham, Nottingham, UK
- School of Chemical Engineering, North Terrace Campus, The University of Adelaide, Adelaide, SA, Australia
| | | | - Li Shean Toh
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Volker Hessel
- School of Chemical Engineering, North Terrace Campus, The University of Adelaide, Adelaide, SA, Australia
| | - Lukas Gerstweiler
- School of Chemical Engineering, North Terrace Campus, The University of Adelaide, Adelaide, SA, Australia
| | - Ian Fisk
- International Flavour Research Centre, Division of Food, Nutrition and Dietetics, University of Nottingham, Sutton Bonington Campus, Loughborough, UK
- International Flavour Research Centre (Adelaide), School of Agriculture, Food and Wine and Waite Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Luis Toronjo-Urquiza
- School of Chemical Engineering, North Terrace Campus, The University of Adelaide, Adelaide, SA, Australia
| | | |
Collapse
|
13
|
Xie L, Cheng Y, Hu B, Chen X, An Y, Xia Z, Cai G, Li C, Peng H. PCLAF induces bone marrow adipocyte senescence and contributes to skeletal aging. Bone Res 2024; 12:38. [PMID: 38961077 PMCID: PMC11222446 DOI: 10.1038/s41413-024-00337-5] [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: 10/14/2023] [Revised: 03/30/2024] [Accepted: 04/11/2024] [Indexed: 07/05/2024] Open
Abstract
Bone marrow adipocytes (BMAds) affect bone homeostasis, but the mechanism remains unclear. Here, we showed that exercise inhibited PCNA clamp-associated factor (PCLAF) secretion from the bone marrow macrophages to inhibit BMAds senescence and thus alleviated skeletal aging. The genetic deletion of PCLAF in macrophages inhibited BMAds senescence and delayed skeletal aging. In contrast, the transplantation of PCLAF-mediated senescent BMAds into the bone marrow of healthy mice suppressed bone turnover. Mechanistically, PCLAF bound to the ADGRL2 receptor to inhibit AKT/mTOR signaling that triggered BMAds senescence and subsequently spread senescence among osteogenic and osteoclastic cells. Of note, we developed a PCLAF-neutralizing antibody and showed its therapeutic effects on skeletal health in old mice. Together, these findings identify PCLAF as an inducer of BMAds senescence and provide a promising way to treat age-related osteoporosis.
Collapse
Affiliation(s)
- Lingqi Xie
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yalun Cheng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Xin Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Yuze An
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Zhuying Xia
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Guangping Cai
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, 410008, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, Hunan, 410008, China
| | - Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
14
|
Huang M, Zhou J, Li X, Liu R, Jiang Y, Chen K, Jiao Y, Yin X, Liu L, Sun Y, Wang W, Xiao Y, Su T, Guo Q, Huang Y, Yang M, Wei J, Darryl Quarles L, Xiao Z, Zeng C, Luo X, Lei G, Li C. Mechanical protein polycystin-1 directly regulates osteoclastogenesis and bone resorption. Sci Bull (Beijing) 2024; 69:1964-1979. [PMID: 38760248 PMCID: PMC11462616 DOI: 10.1016/j.scib.2024.04.044] [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: 10/03/2023] [Revised: 03/25/2024] [Accepted: 03/26/2024] [Indexed: 05/19/2024]
Abstract
Mechanical loading is required for bone homeostasis, but the underlying mechanism is still unclear. Our previous studies revealed that the mechanical protein polycystin-1 (PC1, encoded by Pkd1) is critical for bone formation. However, the role of PC1 in bone resorption is unknown. Here, we found that PC1 directly regulates osteoclastogenesis and bone resorption. The conditional deletion of Pkd1 in the osteoclast lineage resulted in a reduced number of osteoclasts, decreased bone resorption, and increased bone mass. A cohort study of 32,500 patients further revealed that autosomal dominant polycystic kidney disease, which is mainly caused by loss-of-function mutation of the PKD1 gene, is associated with a lower risk of hip fracture than those with other chronic kidney diseases. Moreover, mice with osteoclast-specific knockout of Pkd1 showed complete resistance to unloading-induced bone loss. A mechanistic study revealed that PC1 facilitated TAZ nuclear translocation via the C-terminal tail-TAZ complex and that conditional deletion of Taz in the osteoclast lineage resulted in reduced osteoclastogenesis and increased bone mass. Pharmacological regulation of the PC1-TAZ axis alleviated unloading- and estrogen deficiency- induced bone loss. Thus, the PC1-TAZ axis may be a potential therapeutic target for osteoclast-related osteoporosis.
Collapse
Affiliation(s)
- Mei Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jingxuan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiaoxiao Li
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China; Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ran Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yangzi Jiang
- School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong 999077, China; Center for Neuromusculoskeletal Restorative Medicine (CNRM), The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Kaixuan Chen
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yurui Jiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xin Yin
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ling Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yuchen Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Weishan Wang
- Department of Orthopaedics, The First Affiliated Hospital of Shihezi University, Shihezi 832061, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jie Wei
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China; Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Epidemiology and Health Statistics, Xiangya School of Public Health, Central South University, Changsha 410008, China
| | - L Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis 38163, USA
| | - Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis 38163, USA
| | - Chao Zeng
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China; Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Guanghua Lei
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China; Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha 410008, China; Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Changjun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital, Central South University, Changsha 410008, China; Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China; Laboratory Animal Center, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
15
|
Yang Y, Dai Q, Gao X, Zhu Y, Chung MR, Jin A, Liu Y, Wang X, Huang X, Sun S, Xu H, Liu J, Jiang L. Occlusal force orchestrates alveolar bone homeostasis via Piezo1 in female mice. J Bone Miner Res 2024; 39:580-594. [PMID: 38477783 DOI: 10.1093/jbmr/zjae032] [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: 10/19/2023] [Revised: 02/07/2024] [Accepted: 02/15/2024] [Indexed: 03/14/2024]
Abstract
Healthy alveolar bone is the cornerstone of oral function and oral treatment. Alveolar bone is highly dynamic during the entire lifespan and is affected by both systemic and local factors. Importantly, alveolar bone is subjected to unique occlusal force in daily life, and mechanical force is a powerful trigger of bone remodeling, but the effect of occlusal force in maintaining alveolar bone mass remains ambiguous. In this study, the Piezo1 channel is identified as an occlusal force sensor. Activation of Piezo1 rescues alveolar bone loss caused by a loss of occlusal force. Moreover, we identify Piezo1 as the mediator of occlusal force in osteoblasts, maintaining alveolar bone homeostasis by directly promoting osteogenesis and by sequentially regulating catabolic metabolism through Fas ligand (FasL)-induced osteoclastic apoptosis. Interestingly, Piezo1 activation also exhibits remarkable efficacy in the treatment of alveolar bone osteoporosis caused by estrogen deficiency, which is highly prevalent among middle-aged and elderly women. Promisingly, Piezo1 may serve not only as a treatment target for occlusal force loss-induced alveolar bone loss but also as a potential target for metabolic bone loss, especially in older patients.
Collapse
Affiliation(s)
- Yiling Yang
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Qinggang Dai
- Shanghai Key Laboratory of Stomatology, The 2 nd Dental Center, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Xin Gao
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Yanfei Zhu
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Mi Ri Chung
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Anting Jin
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Yuanqi Liu
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Xijun Wang
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Xiangru Huang
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Siyuan Sun
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Hongyuan Xu
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Jingyi Liu
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| | - Lingyong Jiang
- Shanghai Key Laboratory of Stomatology, Department of Oral and Cranio-Maxillofacial Science, Center of Craniofacial Orthodontics, National Clinical Research Center of Stomatology, Shanghai Research Institute of Stomatology, Shanghai Jiaotong University School of Medicine, , Ninth People's Hospital, Shanghai, Shanghai 200011, China
| |
Collapse
|
16
|
Liang W, Wei T, Hu L, Chen M, Tong L, Zhou W, Duan X, Zhao X, Zhou W, Jiang Q, Xiao G, Zou W, Chen D, Zou Z, Bai X. An integrated multi-omics analysis reveals osteokines involved in global regulation. Cell Metab 2024; 36:1144-1163.e7. [PMID: 38574738 DOI: 10.1016/j.cmet.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/22/2024] [Accepted: 03/10/2024] [Indexed: 04/06/2024]
Abstract
Bone secretory proteins, termed osteokines, regulate bone metabolism and whole-body homeostasis. However, fundamental questions as to what the bona fide osteokines and their cellular sources are and how they are regulated remain unclear. In this study, we analyzed bone and extraskeletal tissues, osteoblast (OB) conditioned media, bone marrow supernatant (BMS), and serum, for basal osteokines and those responsive to aging and mechanical loading/unloading. We identified 375 candidate osteokines and their changes in response to aging and mechanical dynamics by integrating data from RNA-seq, scRNA-seq, and proteomic approaches. Furthermore, we analyzed their cellular sources in the bone and inter-organ communication facilitated by them (bone-brain, liver, and aorta). Notably, we discovered that senescent OBs secrete fatty-acid-binding protein 3 to propagate senescence toward vascular smooth muscle cells (VSMCs). Taken together, we identified previously unknown candidate osteokines and established a dynamic regulatory network among them, thus providing valuable resources to further investigate their systemic roles.
Collapse
Affiliation(s)
- Wenquan Liang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tiantian Wei
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Le Hu
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Meijun Chen
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Liping Tong
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wu Zhou
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xingwei Duan
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoyang Zhao
- Department of Developmental Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Weijie Zhou
- Department of Pathology, Nanfang Hospital, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Guozhi Xiao
- Department of Biochemistry, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen 518055, China
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Di Chen
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Shenzhen, China.
| | - Zhipeng Zou
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Xiaochun Bai
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Academy of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province 510630, China.
| |
Collapse
|
17
|
Gao F, Hu Q, Chen W, Li J, Qi C, Yan Y, Qian C, Wan M, Ficke J, Zheng J, Cao X. Brain regulates weight bearing bone through PGE2 skeletal interoception: implication of ankle osteoarthritis and pain. Bone Res 2024; 12:16. [PMID: 38443372 PMCID: PMC10914853 DOI: 10.1038/s41413-024-00316-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: 09/04/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 03/07/2024] Open
Abstract
Bone is a mechanosensitive tissue and undergoes constant remodeling to adapt to the mechanical loading environment. However, it is unclear whether the signals of bone cells in response to mechanical stress are processed and interpreted in the brain. In this study, we found that the hypothalamus of the brain regulates bone remodeling and structure by perceiving bone prostaglandin E2 (PGE2) concentration in response to mechanical loading. Bone PGE2 levels are in proportion to their weight bearing. When weight bearing changes in the tail-suspension mice, the PGE2 concentrations in bones change in line with their weight bearing changes. Deletion of cyclooxygenase-2 (COX2) in the osteoblast lineage cells or knockout of receptor 4 (EP4) in sensory nerve blunts bone formation in response to mechanical loading. Moreover, knockout of TrkA in sensory nerve also significantly reduces mechanical load-induced bone formation. Moreover, mechanical loading induces cAMP-response element binding protein (CREB) phosphorylation in the hypothalamic arcuate nucleus (ARC) to inhibit sympathetic tyrosine hydroxylase (TH) expression in the paraventricular nucleus (PVN) for osteogenesis. Finally, we show that elevated PGE2 is associated with ankle osteoarthritis (AOA) and pain. Together, our data demonstrate that in response to mechanical loading, skeletal interoception occurs in the form of hypothalamic processing of PGE2-driven peripheral signaling to maintain physiologic bone homeostasis, while chronically elevated PGE2 can be sensed as pain during AOA and implication of potential treatment.
Collapse
Affiliation(s)
- Feng Gao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Qimiao Hu
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Wenwei Chen
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Jilong Li
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Cheng Qi
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Yiwen Yan
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Cheng Qian
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Mei Wan
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - James Ficke
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Junying Zheng
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21205, USA.
| |
Collapse
|
18
|
Camy C, Grünewald T, Lamy E, Roseren F, Caumes M, Fovet T, Brioche T, Genovesio C, Chopard A, Pithioux M, Roffino S. Characterization of the mechanical properties of the mouse Achilles tendon enthesis by microindentation. Effects of unloading and subsequent reloading. Bone Rep 2024; 20:101734. [PMID: 38292933 PMCID: PMC10825371 DOI: 10.1016/j.bonr.2024.101734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 12/14/2023] [Accepted: 01/03/2024] [Indexed: 02/01/2024] Open
Abstract
The fibrocartilaginous tendon enthesis, i.e. the site where a tendon is attached to bone through a fibrocartilaginous tissue, is considered as a functionally graded interface. However, at local scale, a very limited number of studies have characterized micromechanical properties of this transitional tissue. The first goal of this work was to characterize the micromechanical properties of the mineralized part of the healthy Achilles tendon enthesis (ATE) through microindentation testing and to assess the degree of mineralization and of carbonation of mineral crystals by Raman spectroscopy. Since little is known about enthesis biological plasticity, our second objective was to examine the effects of unloading and reloading, using a mouse hindlimb-unloading model, on both the micromechanical properties and the mineral phase of the ATE. Elastic modulus, hardness, degree of mineralization, and degree of carbonation were assessed after 14 days of hindlimb suspension and again after a subsequent 6 days of reloading. The elastic modulus gradually increased along the mineralized part of the ATE from the tidemark to the subchondral bone, with the same trend being found for hardness. Whereas the degree of carbonation did not differ according to zone of measurement, the degree of mineralization increased by >70 % from tidemark to subchondral bone. Thus, the gradient in micromechanical properties is in part explained by a mineralization gradient. A 14-day unloading period did not appear to affect the gradient of micromechanical properties of the ATE, nor the degree of mineralization or carbonation. However, contrary to a short period of unloading, early return to normal mechanical load reduced the micromechanical properties gradient, regardless of carbonate-to-phosphate ratios, likely due to the more homogeneous degree of mineralization. These findings provide valuable data not only for tissue bioengineering, but also for musculoskeletal clinical studies and microgravity studies focusing on long-term space travel by astronauts.
Collapse
Affiliation(s)
- Claire Camy
- Aix Marseille Univ, CNRS, ISM, 13009 Marseille, France
| | - Tilman Grünewald
- Aix Marseille Univ, CNRS, Centrale Marseille, Institut Fresnel, Marseille, France
| | - Edouard Lamy
- Aix Marseille Univ, CNRS, ISM, 13009 Marseille, France
| | - Flavy Roseren
- Aix Marseille Univ, CNRS, ISM, 13009 Marseille, France
- Aix Marseille Univ, APHM, CNRS, ISM, Mecabio Platform, Department of Orthopaedics and Traumatology, 13009 Marseille, France
| | | | - Théo Fovet
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | - Thomas Brioche
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | | | - Angèle Chopard
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | - Martine Pithioux
- Aix Marseille Univ, CNRS, ISM, 13009 Marseille, France
- Aix Marseille Univ, APHM, CNRS, ISM, Mecabio Platform, Department of Orthopaedics and Traumatology, 13009 Marseille, France
- Aix Marseille Univ, APHM, CNRS, ISM, Sainte-Marguerite Hospital, Institute for Locomotion, Department of Orthopaedics and Traumatology, 13009 Marseille, France
| | | |
Collapse
|
19
|
Gao L, Chen R, Liu J, Tan Y, Gao Q, Zhang C, Lv L. Hypergravity stimulates mechanical behavior and micro-architecture of tibia in rats. J Bone Miner Metab 2024; 42:17-26. [PMID: 38062272 DOI: 10.1007/s00774-023-01481-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/02/2023] [Indexed: 02/10/2024]
Abstract
INTRODUCTION The bone tissue is susceptible to hypergravity (+ G) environment. It is necessary to discuss the extent to which specific + G values are beneficial or detrimental to bone tissue. The objective of this study was to characterize the effects of high + G values on mechanical properties, microstructures, and cellular metabolism of bone. MATERIALS AND METHODS 30 male Wistar rats aged 12 weeks were randomly divided into 5 groups, and bore different + G (namely + 1G, + 4G, + 8G, + 10G and + 12G) environments respectively for 4 weeks, 5 days each week, and 3 minutes each day. The macro-mechanical parameters, microstructure parameters, and mRNA transcription levels of the tibia were determined through the three-point bending method, micro-CT detection, and q-PCR analysis, respectively. RESULTS As the + G value increases, hypergravity becomes increasingly detrimental to the macro-mechanical performance of rat tibia. Concerning the microstructure of cancellous bone, there appears to be a favorable trend at + 4G, followed by a progressively detrimental trend at higher G values. In addition, the mRNA transcription levels of OPG and RANKL show an initial tendency of enhanced bone absorption at +4G, followed by an increase in bone remodeling capacity as G value increases. CONCLUSION The higher G values correspond to poorer macro-mechanical properties of the tibia, and a + 4G environment benefits the microstructure of the tibia. At the cellular level, bone resorption is enhanced in the + 4G group, but the bone remodeling capability gradually increases with further increments in G values.
Collapse
Affiliation(s)
- Lilan Gao
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300384, People's Republic of China
| | - Ruiqi Chen
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300384, People's Republic of China
| | - Jin Liu
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300384, People's Republic of China
| | - Yansong Tan
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China.
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300384, People's Republic of China.
| | - Qijun Gao
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China.
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300384, People's Republic of China.
| | - Chunqiu Zhang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300384, People's Republic of China
| | - Linwei Lv
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, People's Republic of China
- National Demonstration Center for Experimental Mechanical and Electrical Engineering Education, Tianjin University of Technology, Tianjin, 300384, People's Republic of China
| |
Collapse
|
20
|
Ogura H, Nakamura T, Ishii T, Saito A, Onodera S, Yamaguchi A, Nishii Y, Azuma T. Mechanical stress-induced FGF-2 promotes proliferation and consequently induces osteoblast differentiation in mesenchymal stem cells. Biochem Biophys Res Commun 2023; 684:149145. [PMID: 37913587 DOI: 10.1016/j.bbrc.2023.149145] [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: 08/29/2023] [Accepted: 10/23/2023] [Indexed: 11/03/2023]
Abstract
Mechanical stimuli serve as crucial regulators of bone mass, promoting bone formation. However, the molecular mechanisms governing how mesenchymal stem cells (MSCs) respond to mechanical cues during their differentiation into osteogenic cells remain elusive. In this study, we found that cyclic stretching enhances MSC proliferation but does not increase the expression of osteoblast-related genes. We further revealed that this proliferative effect is mediated by fibroblast growth factor 2 (FGF-2), synthesized by MSCs in response to mechanical stress. Cell proliferation induced by cyclic stretching was inhibited upon the addition of either U0126, an inhibitor of mitogen-activated protein kinase kinase (MEK), or early growth response 1 (EGR1)-targeting small-hairpin RNA (shRNA), indicating the involvement of the extracellular signal-regulated kinase (ERK)/EGR1 signaling pathway. Osteoblast differentiation, evaluated through ALP activity, osteoblast-related gene expression, and mineralization, was stimulated by recombinant human FGF-2 (rhFGF-2) when applied during the proliferation phase, but not when applied during the differentiation stage alone. Our results suggest that FGF-2 indirectly promotes osteoblast differentiation as a downstream effect of stimulating cell proliferation. For the first time, we demonstrate that cyclic stretching induces MSCs to produce FGF-2, which in turn encourages cell proliferation through an autocrine/paracrine mechanism, consequently leading to osteoblast differentiation.
Collapse
Affiliation(s)
- Hiroyuki Ogura
- Department of Orthodontics, Tokyo Dental College, Tokyo, 101-0061, Japan
| | - Takashi Nakamura
- Department of Biochemistry, Tokyo Dental College, Tokyo, 101-0061, Japan; Oral Health Science Center, Tokyo Dental College, Tokyo, 101-0061, Japan.
| | - Takenobu Ishii
- Department of Orthodontics, Tokyo Dental College, Tokyo, 101-0061, Japan
| | - Akiko Saito
- Department of Biochemistry, Tokyo Dental College, Tokyo, 101-0061, Japan
| | - Shoko Onodera
- Department of Biochemistry, Tokyo Dental College, Tokyo, 101-0061, Japan
| | - Akira Yamaguchi
- Oral Health Science Center, Tokyo Dental College, Tokyo, 101-0061, Japan
| | - Yasushi Nishii
- Department of Orthodontics, Tokyo Dental College, Tokyo, 101-0061, Japan
| | - Toshifumi Azuma
- Department of Biochemistry, Tokyo Dental College, Tokyo, 101-0061, Japan; Oral Health Science Center, Tokyo Dental College, Tokyo, 101-0061, Japan
| |
Collapse
|
21
|
Fonte C, Jacob P, Vanet A, Ghislin S, Frippiat JP. Hindlimb unloading, a physiological model of microgravity, modifies the murine bone marrow IgM repertoire in a similar manner as aging but less strongly. Immun Ageing 2023; 20:64. [PMID: 37986079 PMCID: PMC10659048 DOI: 10.1186/s12979-023-00393-1] [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: 06/05/2023] [Accepted: 11/12/2023] [Indexed: 11/22/2023]
Abstract
BACKGROUND The spaceflight environment is an extreme environment that affects the immune system of approximately 50% of astronauts. With planned long-duration missions, such as the deployment of the Lunar Gateway and possible interplanetary missions, it is mandatory to determine how all components of the immune system are affected, which will allow the establishment of countermeasures to preserve astronaut health. However, despite being an important component of the immune system, antibody-mediated humoral immunity has rarely been investigated in the context of the effects of the space environment. It has previously been demonstrated that 30 days aboard the BION-M1 satellite and 21 days of hindlimb unloading (HU), a model classically used to mimic the effects of microgravity, decrease murine B lymphopoiesis. Furthermore, modifications in B lymphopoiesis reported in young mice subjected to 21 days of HU were shown to be similar to those observed in aged mice (18-22 months). Since the primary antibody repertoire composed of IgM is created by V(D) J recombination during B lymphopoiesis, the objective of this study was to assess the degree of similarity between changes in the bone marrow IgM repertoire and in the V(D)J recombination process in 2.5-month-old mice subjected to 21 days of HU and aged (18 months) mice. RESULTS We found that in 21 days, HU induced changes in the IgM repertoire that were approximately 3-fold less than those in aged mice, which is a rapid effect. Bone remodeling and epigenetics likely mediate these changes. Indeed, we previously demonstrated a significant decrease in tibial morphometric parameters from day 6 of HU and a progressive reduction in these parameters until day 21 of HU, and it has been shown that age and microgravity induce epigenetic changes. CONCLUSION These data reveal novel immune changes that are akin to advanced aging and underline the importance of studying the effects of spaceflight on antibody-mediated humoral immunity.
Collapse
Affiliation(s)
- Coralie Fonte
- Stress Immunity Pathogens Laboratory, UR 7300 SIMPA, Faculty of Medicine, Lorraine University, Vandoeuvre-lès, Nancy, France
| | - Pauline Jacob
- Stress Immunity Pathogens Laboratory, UR 7300 SIMPA, Faculty of Medicine, Lorraine University, Vandoeuvre-lès, Nancy, France
| | - Anne Vanet
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013, Paris, France
| | - Stéphanie Ghislin
- Stress Immunity Pathogens Laboratory, UR 7300 SIMPA, Faculty of Medicine, Lorraine University, Vandoeuvre-lès, Nancy, France
| | - Jean-Pol Frippiat
- Stress Immunity Pathogens Laboratory, UR 7300 SIMPA, Faculty of Medicine, Lorraine University, Vandoeuvre-lès, Nancy, France.
| |
Collapse
|
22
|
Meas SJ, Daire GM, Friedman MA, DeNapoli R, Ghosh P, Farr JN, Donahue HJ. Hindlimb Unloading Induces Bone Microarchitectural and Transcriptomic Changes in Murine Long Bones in an Age-Dependent Manner. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561510. [PMID: 37873408 PMCID: PMC10592678 DOI: 10.1101/2023.10.09.561510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Age and disuse-related bone loss both result in decreases in bone mineral density, cortical thickness, and trabecular thickness and connectivity. Disuse induces physiological changes in bone like those seen with aging. Bone microarchitecture and biomechanical properties were compared between 6- and 22-month-old C57BL/6J male control mice and 6-month-old mice that were hindlimb unloaded (HLU) for 3 weeks. Epiphyseal trabecular bone was the compartment most affected by HLU and demonstrated an intermediate bone phenotype between age-matched controls and aged controls. RNA extracted from whole-bone marrow-flushed tibiae was sequenced and analyzed. Differential gene expression analysis additionally included 4-month-old male mice unloaded for 3 weeks compared to age-matched controls. Gene ontology analysis demonstrated that there were age-dependent differences in differentially expressed genes. Genes related to downregulation of cellular processes were most affected in 4-month-old mice after disuse whereas those related to mitochondrial function were most affected in 6- month-old mice. Cell-cycle transition was downregulated with aging. A publicly available dataset (GSE169292) from 3-month female C57BL/6N mice unloaded for 7 days was included in ingenuity pathway analysis with the other datasets. IPA was used to identify the leading canonical pathways and upstream regulators in each HLU age group. IPA identified "Senescence Pathway" as the second leading canonical pathway enriched in mice exposed to HLU. HLU induced activation of the senescence pathway in 3- month and 4-month-old mice but inhibited it in 6-month-old mice. In conclusion, we demonstrate that hindlimb unloading and aging initiate similar changes in bone microarchitecture and gene expression. However, aging is responsible for more significant transcriptome and tissue-level changes compared to hindlimb unloading. Highlights Epiphyseal trabecular bone is most susceptible to hindlimb unloading.Hindlimb unloaded limbs resemble an intermediate phenotype between age-matched and aged controls.Hindlimb unloading induces gene expression changes that are age dependent and may lead to inflammation and/or mitochondrial dysfunction depending on context.Younger mice (3-4 months) activate the senescence pathway upon hindlimb unloading, whereas skeletally mature (6 months) mice inhibit it.
Collapse
|
23
|
Bukreeva I, Gulimova VI, Krivonosov YS, Buzmakov AV, Junemann O, Cedola A, Fratini M, Maugeri L, Begani Provinciali G, Palermo F, Sanna A, Pieroni N, Asadchikov VE, Saveliev SV. The Study of the Caudal Vertebrae of Thick-Toed Geckos after a Prolonged Space Flight by X-ray Phase-Contrast Micro-CT. Cells 2023; 12:2415. [PMID: 37830629 PMCID: PMC10572532 DOI: 10.3390/cells12192415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 10/14/2023] Open
Abstract
The proximal caudal vertebrae and notochord in thick-toed geckos (TG) (Chondrodactylus turneri, Gray, 1864) were investigated after a 30-day space flight onboard the biosatellite Bion-M1. This region has not been explored in previous studies. Our research focused on finding sites most affected by demineralization caused by microgravity (G0). We used X-ray phase-contrast tomography to study TG samples without invasive prior preparation to clarify our previous findings on the resistance of TG's bones to demineralization in G0. The results of the present study confirmed that geckos are capable of preserving bone mass after flight, as neither cortical nor trabecular bone volume fraction showed statistically significant changes after flight. On the other hand, we observed a clear decrease in the mineralization of the notochordal septum and a substantial rise in intercentrum volume following the flight. To monitor TG's mineral metabolism in G0, we propose to measure the volume of mineralized tissue in the notochordal septum. This technique holds promise as a sensitive approach to track the demineralization process in G0, given that the volume of calcification within the septum is limited, making it easy to detect even slight changes in mineral content.
Collapse
Affiliation(s)
- Inna Bukreeva
- Institute of Nanotechnology, CNR, Rome Unit, Piazzale Aldo Moro 5, 00185 Rome, Italy; (I.B.); (O.J.); (A.C.); (M.F.)
- P.N. Lebedev Physical Institute Russian Academy of Sciences, Leninskiy Prospekt 53, 119991 Moscow, Russia
| | - Victoria I. Gulimova
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution, “Petrovsky National Research Centre of Surgery”, Tsyurupy Str. 3, 117418 Moscow, Russia;
| | - Yuri S. Krivonosov
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Leninskiy Prospekt 59, 119333 Moscow, Russia (V.E.A.)
| | - Alexey V. Buzmakov
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Leninskiy Prospekt 59, 119333 Moscow, Russia (V.E.A.)
| | - Olga Junemann
- Institute of Nanotechnology, CNR, Rome Unit, Piazzale Aldo Moro 5, 00185 Rome, Italy; (I.B.); (O.J.); (A.C.); (M.F.)
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution, “Petrovsky National Research Centre of Surgery”, Tsyurupy Str. 3, 117418 Moscow, Russia;
| | - Alessia Cedola
- Institute of Nanotechnology, CNR, Rome Unit, Piazzale Aldo Moro 5, 00185 Rome, Italy; (I.B.); (O.J.); (A.C.); (M.F.)
| | - Michela Fratini
- Institute of Nanotechnology, CNR, Rome Unit, Piazzale Aldo Moro 5, 00185 Rome, Italy; (I.B.); (O.J.); (A.C.); (M.F.)
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306/354, 00142 Roma, Italy
| | - Laura Maugeri
- IRCCS Fondazione Santa Lucia, Via Ardeatina 306/354, 00142 Roma, Italy
| | - Ginevra Begani Provinciali
- Institute of Nanotechnology, CNR, Rome Unit, Piazzale Aldo Moro 5, 00185 Rome, Italy; (I.B.); (O.J.); (A.C.); (M.F.)
- Physics Department, ‘Sapienza’ University, Piazzale Aldo Moro 2, 00185 Rome, Italy
- Laboratoire d’Optique Appliquée, CNRS, ENSTA Paris, Ecole Polytechnique IP Paris, 91120 Palaiseau, France
| | - Francesca Palermo
- Institute of Nanotechnology, CNR, Rome Unit, Piazzale Aldo Moro 5, 00185 Rome, Italy; (I.B.); (O.J.); (A.C.); (M.F.)
| | - Alessia Sanna
- Institute of Nanotechnology, CNR, Rome Unit, Piazzale Aldo Moro 5, 00185 Rome, Italy; (I.B.); (O.J.); (A.C.); (M.F.)
| | - Nicola Pieroni
- Institute of Nanotechnology, CNR, Rome Unit, Piazzale Aldo Moro 5, 00185 Rome, Italy; (I.B.); (O.J.); (A.C.); (M.F.)
| | - Victor E. Asadchikov
- Federal Scientific Research Centre “Crystallography and Photonics”, Russian Academy of Sciences, Leninskiy Prospekt 59, 119333 Moscow, Russia (V.E.A.)
| | - Sergey V. Saveliev
- Avtsyn Research Institute of Human Morphology of Federal State Budgetary Scientific Institution, “Petrovsky National Research Centre of Surgery”, Tsyurupy Str. 3, 117418 Moscow, Russia;
| |
Collapse
|
24
|
Ha P, Kwak JH, Zhang Y, Shi J, Tran L, Liu TP, Pan HC, Lee S, Kim JK, Chen E, Shirazi-Fard Y, Stodieck LS, Lin A, Zheng Z, Dong SN, Zhang X, Wu BM, Ting K, Soo C. Bisphosphonate conjugation enhances the bone-specificity of NELL-1-based systemic therapy for spaceflight-induced bone loss in mice. NPJ Microgravity 2023; 9:75. [PMID: 37723136 PMCID: PMC10507033 DOI: 10.1038/s41526-023-00319-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/18/2023] [Indexed: 09/20/2023] Open
Abstract
Microgravity-induced bone loss results in a 1% bone mineral density loss monthly and can be a mission critical factor in long-duration spaceflight. Biomolecular therapies with dual osteogenic and anti-resorptive functions are promising for treating extreme osteoporosis. We previously confirmed that NELL-like molecule-1 (NELL-1) is crucial for bone density maintenance. We further PEGylated NELL-1 (NELL-polyethylene glycol, or NELL-PEG) to increase systemic delivery half-life from 5.5 to 15.5 h. In this study, we used a bio-inert bisphosphonate (BP) moiety to chemically engineer NELL-PEG into BP-NELL-PEG and specifically target bone tissues. We found conjugation with BP improved hydroxyapatite (HA) binding and protein stability of NELL-PEG while preserving NELL-1's osteogenicity in vitro. Furthermore, BP-NELL-PEG showed superior in vivo bone specificity without observable pathology in liver, spleen, lungs, brain, heart, muscles, or ovaries of mice. Finally, we tested BP-NELL-PEG through spaceflight exposure onboard the International Space Station (ISS) at maximal animal capacity (n = 40) in a long-term (9 week) osteoporosis therapeutic study and found that BP-NELL-PEG significantly increased bone formation in flight and ground control mice without obvious adverse health effects. Our results highlight BP-NELL-PEG as a promising therapeutic to mitigate extreme bone loss from long-duration microgravity exposure and musculoskeletal degeneration on Earth, especially when resistance training is not possible due to incapacity (e.g., bone fracture, stroke).
Collapse
Affiliation(s)
- Pin Ha
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jin Hee Kwak
- Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, 90089, USA
| | - Yulong Zhang
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Forsyth Institute, Cambridge, MA, 02142, USA
| | - Jiayu Shi
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Luan Tran
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Timothy Pan Liu
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hsin-Chuan Pan
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Samantha Lee
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jong Kil Kim
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Eric Chen
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yasaman Shirazi-Fard
- Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA, 94035, USA
| | - Louis S Stodieck
- BioServe Space Technologies and Aerospace Engineering Sciences, University of Colorado, Boulder, CO, 80303, USA
| | - Andy Lin
- Office of Advanced Research Computing, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhong Zheng
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Stella Nuo Dong
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinli Zhang
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Benjamin M Wu
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Forsyth Institute, Cambridge, MA, 02142, USA.
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Kang Ting
- Forsyth Institute, Cambridge, MA, 02142, USA.
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Orthopaedic Surgery and the Orthopaedic Hospital Research Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Science, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
25
|
Hajj-Boutros G, Sonjak V, Faust A, Hedge E, Mastrandrea C, Lagacé JC, St-Martin P, Naz Divsalar D, Sadeghian F, Chevalier S, Liu-Ambrose T, Blaber AP, Dionne IJ, Duchesne S, Hughson R, Kontulainen S, Theou O, Morais JA. Impact of 14 Days of Bed Rest in Older Adults and an Exercise Countermeasure on Body Composition, Muscle Strength, and Cardiovascular Function: Canadian Space Agency Standard Measures. Gerontology 2023; 69:1284-1294. [PMID: 37717560 PMCID: PMC10634275 DOI: 10.1159/000534063] [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] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023] Open
Abstract
INTRODUCTION Head-down bed rest (HDBR) has long been used as an analog to microgravity, and it also enables studying the changes occurring with aging. Exercise is the most effective countermeasure for the deleterious effects of inactivity. The aim of this study was to investigate the efficacy of an exercise countermeasure in healthy older participants on attenuating musculoskeletal deconditioning, cardiovascular fitness level, and muscle strength during 14 days of HDBR as part of the standard measures of the Canadian Space Agency. METHODS Twenty-three participants (12 males and 11 females), aged 55-65 years, were admitted for a 26-day inpatient stay at the McGill University Health Centre. After 5 days of baseline assessment tests, they underwent 14 days of continuous HDBR followed by 7 days of recovery with repeated tests. Participants were randomized to passive physiotherapy or an exercise countermeasure during the HDBR period consisting of 3 sessions per day of either high-intensity interval training (HIIT) or low-intensity cycling or strength exercises for the lower and upper body. Peak aerobic power (V̇O2peak) was determined using indirect calorimetry. Body composition was assessed by dual-energy X-ray absorptiometry, and several muscle group strengths were evaluated using an adjustable chair dynamometer. A vertical jump was used to assess whole-body power output, and a tilt test was used to measure cardiovascular and orthostatic challenges. Additionally, changes in various blood parameters were measured as well as the effects of exercise countermeasure on these measurements. RESULTS There were no differences at baseline in main characteristics between the control and exercise groups. The exercise group maintained V̇O2peak levels similar to baseline, whereas it decreased in the control group following 14 days of HDBR. Body weight significantly decreased in both groups. Total and leg lean masses decreased in both groups. However, total body fat mass decreased only in the exercise group. Isometric and isokinetic knee extension muscle strength were significantly reduced in both groups. Peak velocity, flight height, and flight time were significantly reduced in both groups with HDBR. CONCLUSION In this first Canadian HDBR study in older adults, an exercise countermeasure helped maintain aerobic fitness and lean body mass without affecting the reduction of knee extension strength. However, it was ineffective in protecting against orthostatic intolerance. These results support HIIT as a promising approach to preserve astronaut health and functioning during space missions, and to prevent deconditioning as a result of hospitalization in older adults.
Collapse
Affiliation(s)
- Guy Hajj-Boutros
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada,
| | - Vita Sonjak
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Andréa Faust
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Eric Hedge
- Department of Kinesiology, Schlegel-University of Waterloo Research Institute for Aging, Waterloo, Ontario, Canada
| | - Carmelo Mastrandrea
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Jean-Christophe Lagacé
- Faculté des Sciences de l'activité Physique, Centre de Recherche sur le Vieillissement, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Philippe St-Martin
- Faculté des Sciences de l'activité Physique, Centre de Recherche sur le Vieillissement, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Donya Naz Divsalar
- Department of Biomedical Physiology and Kinesiology, Aerospace Physiology Laboratory, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Farshid Sadeghian
- Department of Biomedical Physiology and Kinesiology, Aerospace Physiology Laboratory, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Stéphanie Chevalier
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
- School of Human Nutrition, McGill University, Montreal, Québec, Canada
| | - Teresa Liu-Ambrose
- Aging, Mobility and Cognitive Neuroscience Laboratory, Department of Physical Therapy, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew P Blaber
- Department of Biomedical Physiology and Kinesiology, Aerospace Physiology Laboratory, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Isabelle J Dionne
- Faculté des Sciences de l'activité Physique, Centre de Recherche sur le Vieillissement, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Simon Duchesne
- Department of Radiology and Nuclear Medicine, Université Laval, Quebec City, Québec, Canada
- CERVO Brain Research Center, Quebec City, Québec, Canada
| | - Richard Hughson
- Department of Kinesiology, Schlegel-University of Waterloo Research Institute for Aging, Waterloo, Ontario, Canada
| | - Saija Kontulainen
- College of Kinesiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Olga Theou
- Division of Geriatric Medicine, Queen Elizabeth II Health Sciences Centre, Nova Scotia Health, Halifax, Nova Scotia, Canada
| | - José A Morais
- Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
- Division of Geriatric Medicine, Faculty of Medicine, McGill University, Montreal, Québec, Canada
| |
Collapse
|
26
|
Song C, Kang T, Gao K, Shi X, Zhang M, Zhao L, Zhou L, Guo J. Preparation for mice spaceflight: Indications for training C57BL/6J mice to adapt to microgravity effect with three-dimensional clinostat on the ground. Heliyon 2023; 9:e19355. [PMID: 37662714 PMCID: PMC10472007 DOI: 10.1016/j.heliyon.2023.e19355] [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: 02/23/2023] [Revised: 08/09/2023] [Accepted: 08/20/2023] [Indexed: 09/05/2023] Open
Abstract
Like astronauts, animals need to undergo training and screening before entering space. At present, pre-launch training for mice mainly focuses on adaptation to habitat system. Training for the weightless environment of space in mice has not received much attention. Three-dimensional (3D) clinostat is a method to simulate the effects of microgravity on Earth. However, few studies have used a 3D clinostat apparatus to simulate the effects of microgravity on animal models. Therefore, we conducted a study to evaluate the feasibility and effects of long-term treatment with three-dimensional clinostat in C57BL/6 J mice. Thirty 8-week-old male C57BL/6 J mice were randomly assigned to three groups: mice in individually ventilated cages (MC group, n = 6), mice in survival boxes (SB group, n = 12), and mice in survival boxes receiving 3D clinostat treatment (CS group, n = 12). The mice showed good tolerance after 12 weeks of alternate day training. To evaluate the biological effects of simulated microgravity, the changes in serum metabolites were monitored using untargeted metabolomics, whereas bone loss was assessed using microcomputed tomography of the left femur. Compared with the metabolome of the SB group, the metabolome of the CS group showed significant differences during the first three weeks and the last three weeks. The KEGG pathways in the late stages were mainly related to the nervous system, indicating the influence of long-term microgravity on the central nervous system. Besides, a marked reduction in the trabecular number (P < 0.05) and an increasing trend of trabecular spacing (P < 0.1) were observed to occur in a time-dependent manner in the CS group compared with the SB group. These results showed that mice tolerated well in a 3D clinostat and may provide a new strategy in pre-launch training for mice and conducting relevant ground-based modeling experiments.
Collapse
Affiliation(s)
- Chenchen Song
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Taisheng Kang
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Kai Gao
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Xudong Shi
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Meng Zhang
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Lianlian Zhao
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Li Zhou
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| | - Jianguo Guo
- Key Laboratory of Human Disease Comparative Medicine, Chinese Ministry of Health, Key Laboratory for Animal Models of Emerging and Reemerging Infectious Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences and Comparative Medicine Center, Peking Union Medical College, Beijing, China
| |
Collapse
|
27
|
Hedge ET, Vico L, Hughson RL, Mastrandrea CJ. Effects of Posture and Walking on Tibial Vascular Hemodynamics Before and After 14 Days of Head-Down Bed Rest. JBMR Plus 2023; 7:e10756. [PMID: 37457881 PMCID: PMC10339089 DOI: 10.1002/jbm4.10756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 04/17/2023] [Indexed: 07/18/2023] Open
Abstract
Human skeletal hemodynamics remain understudied. Neither assessments in weight-bearing bones during walking nor following periods of immobility exist, despite knowledge of altered nutrient-artery characteristics after short-duration unloading in rodents. We studied 12 older adults (8 females, aged 59 ± 3 years) who participated in ambulatory near-infrared spectroscopy (NIRS) assessments of tibial hemodynamics before (PRE) and after (POST) 14 days of head-down bed rest (HDBR), with most performing daily resistance and aerobic exercise countermeasures during HDBR. Continual simultaneous NIRS recordings were acquired over the proximal anteromedial tibial prominence of the right lower leg and ipsilateral lateral head of the gastrocnemius muscle during supine rest, walking, and standing. During 10 minutes of walking, desaturation kinetics in the tibia were slower (time to 95% nadir values 125.4 ± 56.8 s versus 55.0 ± 30.1 s, p = 0.0014). Tibial tissue saturation index (TSI) immediately fell (-9.9 ± 4.55) and did not completely recover by the end of 10 minutes of walking (-7.4 ± 6.7%, p = 0.027). Upon standing, total hemoglobin (tHb) kinetics were faster in the tibia (p < 0.0001), whereas HDBR resulted in faster oxygenated hemoglogin (O2Hb) kinetics in both tissues (p = 0.039). After the walk-to-stand transition, changes in O2Hb (p = 0.0022) and tHb (p = 0.0047) were attenuated in the tibia alone after bed rest. Comparisons of NIRS-derived variables during ambulation and changes in posture revealed potentially deleterious adaptations of feed vessels after HDBR. We identify important and novel tibial hemodynamics in humans during ambulation before and after bed rest, necessitating further investigation. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Eric T Hedge
- Schlegel‐UW Research Institute for AgingWaterlooCanada
- Department of Kinesiology and Health SciencesUniversity of WaterlooWaterlooCanada
| | - Laurence Vico
- U1059 INSERM—SAINBIOSE (Santé Ingéniérie Biologie St‐Etienne) Campus Santé InnovationUniversité Jean MonnetSaint‐Priest‐en‐JarezFrance
| | | | | |
Collapse
|
28
|
Nottmeier C, Lavicky J, Gonzalez Lopez M, Knauth S, Kahl-Nieke B, Amling M, Schinke T, Helms J, Krivanek J, Koehne T, Petersen J. Mechanical-induced bone remodeling does not depend on Piezo1 in dentoalveolar hard tissue. Sci Rep 2023; 13:9563. [PMID: 37308580 DOI: 10.1038/s41598-023-36699-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/08/2023] [Indexed: 06/14/2023] Open
Abstract
Mechanosensory ion channels are proteins that are sensitive to mechanical forces. They are found in tissues throughout the body and play an important role in bone remodeling by sensing changes in mechanical stress and transmitting signals to bone-forming cells. Orthodontic tooth movement (OTM) is a prime example of mechanically induced bone remodeling. However, the cell-specific role of the ion channels Piezo1 and Piezo2 in OTM has not been investigated yet. Here we first identify the expression of PIEZO1/2 in the dentoalveolar hard tissues. Results showed that PIEZO1 was expressed in odontoblasts, osteoblasts, and osteocytes, while PIEZO2 was localized in odontoblasts and cementoblasts. We therefore used a Piezo1floxed/floxed mouse model in combination with Dmp1cre to inactivate Piezo1 in mature osteoblasts/cementoblasts, osteocytes/cementocytes, and odontoblasts. Inactivation of Piezo1 in these cells did not affect the overall morphology of the skull but caused significant bone loss in the craniofacial skeleton. Histological analysis revealed a significantly increased number of osteoclasts in Piezo1floxed/floxed;Dmp1cre mice, while osteoblasts were not affected. Despite this increased number of osteoclasts, orthodontic tooth movement was not altered in these mice. Our results suggest that despite Piezo1 being crucial for osteoclast function, it may be dispensable for mechanical sensing of bone remodeling.
Collapse
Affiliation(s)
- Cita Nottmeier
- Department of Orthodontics, University of Leipzig Medical Center, Saxony, Germany
| | - Josef Lavicky
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Marcos Gonzalez Lopez
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sarah Knauth
- Department of Orthodontics, University of Leipzig Medical Center, Saxony, Germany
| | - Bärbel Kahl-Nieke
- Department of Orthodontics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Institute of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Institute of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jill Helms
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Jan Krivanek
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Till Koehne
- Department of Orthodontics, University of Leipzig Medical Center, Saxony, Germany.
| | - Julian Petersen
- Department of Orthodontics, University of Leipzig Medical Center, Saxony, Germany.
| |
Collapse
|
29
|
Bedree JK, Kerns K, Chen T, Lima BP, Liu G, Ha P, Shi J, Pan HC, Kim JK, Tran L, Minot SS, Hendrickson EL, Lamont EI, Schulte F, Hardt M, Stephens D, Patel M, Kokaras A, Stodieck L, Shirazi-Fard Y, Wu B, Kwak JH, Ting K, Soo C, McLean JS, He X, Shi W. Specific host metabolite and gut microbiome alterations are associated with bone loss during spaceflight. Cell Rep 2023; 42:112299. [PMID: 37080202 PMCID: PMC10344367 DOI: 10.1016/j.celrep.2023.112299] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 10/30/2022] [Accepted: 03/07/2023] [Indexed: 04/22/2023] Open
Abstract
Understanding the axis of the human microbiome and physiological homeostasis is an essential task in managing deep-space-travel-associated health risks. The NASA-led Rodent Research 5 mission enabled an ancillary investigation of the gut microbiome, varying exposure to microgravity (flight) relative to ground controls in the context of previously shown bone mineral density (BMD) loss that was observed in these flight groups. We demonstrate elevated abundance of Lactobacillus murinus and Dorea sp. during microgravity exposure relative to ground control through whole-genome sequencing and 16S rRNA analyses. Specific functionally assigned gene clusters of L. murinus and Dorea sp. capable of producing metabolites, lactic acid, leucine/isoleucine, and glutathione are enriched. These metabolites are elevated in the microgravity-exposed host serum as shown by liquid chromatography-tandem mass spectrometry (LC-MS/MS) metabolomic analysis. Along with BMD loss, ELISA reveals increases in osteocalcin and reductions in tartrate-resistant acid phosphatase 5b signifying additional loss of bone homeostasis in flight.
Collapse
Affiliation(s)
- Joseph K Bedree
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA.
| | - Kristopher Kerns
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Tsute Chen
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Bruno P Lima
- Department of Diagnostic and Biological Sciences, University of Minnesota, Minneapolis, MN 55455, USA
| | - Guo Liu
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Pin Ha
- Section of Orthodontics, Division of Growth & Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Plastic and Reconstructive Surgery, School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jiayu Shi
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthodontics and Pediatric Dentistry, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Hsin Chuan Pan
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jong Kil Kim
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Luan Tran
- Section of Oral Biology, Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Samuel S Minot
- Microbiome Research Initiative, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Erik L Hendrickson
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Eleanor I Lamont
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Fabian Schulte
- Forsyth Center for Salivary Diagnostics, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA; Harvard School of Dental Medicine, Department of Developmental Biology, Boston, MA 02115, USA
| | - Markus Hardt
- Forsyth Center for Salivary Diagnostics, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA; Harvard School of Dental Medicine, Department of Developmental Biology, Boston, MA 02115, USA
| | - Danielle Stephens
- Multiplex Core, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Michele Patel
- Multiplex Core, Department of Applied Oral Sciences, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Alexis Kokaras
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA
| | - Louis Stodieck
- BioServe Space Technologies, Department of Aerospace Engineering Sciences, University of Colorado, Boulder, CO 80303, USA
| | - Yasaman Shirazi-Fard
- Bone and Signaling Laboratory, Space Biosciences Division, NASA Ames Research Center, Mail Stop 288-2, Moffett Field, CA 94035, USA
| | - Benjamin Wu
- Department of Bioengineering, School of Engineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Advanced Prosthodontics, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jin Hee Kwak
- Section of Orthodontics, Division of Growth & Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kang Ting
- Section of Orthodontics, Division of Growth & Development, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Chia Soo
- Division of Plastic and Reconstructive Surgery, School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Orthopedic Surgery, School of Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jeffrey S McLean
- Department of Periodontics, School of Dentistry, University of Washington, Seattle, WA 98195, USA
| | - Xuesong He
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA; Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Wenyuan Shi
- Department of Microbiology, The Forsyth Institute, Cambridge, MA 02142, USA.
| |
Collapse
|
30
|
Sun W, Li Y, Li J, Tan Y, Yuan X, Meng H, Ye J, Zhong G, Jin X, Liu Z, Du R, Xing W, Zhao D, Song J, Li Y, Pan J, Zhao Y, Li Q, Wang A, Ling S, Dai R, Li Y. Mechanical stimulation controls osteoclast function through the regulation of Ca 2+-activated Cl - channel Anoctamin 1. Commun Biol 2023; 6:407. [PMID: 37055517 PMCID: PMC10102170 DOI: 10.1038/s42003-023-04806-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 04/04/2023] [Indexed: 04/15/2023] Open
Abstract
Mechanical force loading is essential for maintaining bone homeostasis, and unloading exposure can lead to bone loss. Osteoclasts are the only bone resorbing cells and play a crucial role in bone remodeling. The molecular mechanisms underlying mechanical stimulation-induced changes in osteoclast function remain to be fully elucidated. Our previous research found Ca2+-activated Cl- channel Anoctamin 1 (Ano1) was an essential regulator for osteoclast function. Here, we report that Ano1 mediates osteoclast responses to mechanical stimulation. In vitro, osteoclast activities are obviously affected by mechanical stress, which is accompanied by the changes of Ano1 levels, intracellular Cl- concentration and Ca2+ downstream signaling. Ano1 knockout or calcium binding mutants blunts the response of osteoclast to mechanical stimulation. In vivo, Ano1 knockout in osteoclast blunts loading induced osteoclast inhibition and unloading induced bone loss and. These results demonstrate that Ano1 plays an important role in mechanical stimulation induced osteoclast activity changes.
Collapse
Affiliation(s)
- Weijia Sun
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing, China
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yuheng Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Jianwei Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yingjun Tan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Xinxin Yuan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Haoye Meng
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing, China
| | - Jianting Ye
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing, China
| | - Guohui Zhong
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - XiaoYan Jin
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Zizhong Liu
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Ruikai Du
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Wenjuan Xing
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Dingsheng Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Jinping Song
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Youyou Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Junjie Pan
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Yunzhang Zhao
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Qi Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China
| | - Aiyuan Wang
- Institute of Orthopaedics, Chinese PLA General Hospital, Beijing, China
| | - Shukuan Ling
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, China.
| | - Rongji Dai
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, Beijing Institute of Technology, Beijing, China.
| | - Yingxian Li
- State Key Laboratory of Space Medicine Fundamentals and Application, China Astronaut Research and Training Center, Beijing, China.
| |
Collapse
|
31
|
Peurière L, Mastrandrea C, Vanden-Bossche A, Linossier MT, Thomas M, Normand M, Lafage-Proust MH, Vico L. Hindlimb unloading in C57BL/6J mice induces bone loss at thermoneutrality without change in osteocyte and lacuno-canalicular network. Bone 2023; 169:116640. [PMID: 36526262 DOI: 10.1016/j.bone.2022.116640] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/13/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022]
Abstract
Impaired mechanical stimuli during hindlimb unloading (HLU) are believed to exacerbate osteocyte paracrine regulation of osteoclasts. We hypothesized that bone loss and deterioration of the osteocyte lacuno-canalicular network are attenuated in HLU mice housed at thermoneutrality (28 °C) compared with those housed at ambient temperature (22 °C). Following acclimatization, 20-week-old male C57BL/6J mice were submitted to HLU or kept in pair-fed control cages (CONT), for 5 days (5d) or 14d, at 22 °C or 28 °C. In the femur distal metaphysis, thermoneutral CONT mice had higher bone volume (p = 0.0007, BV/TV, in vivo μCT, vs. 14dCONT22) whilst osteoclastic surfaces of CONT and HLU were greater at 22 °C (5dCONT22 + 53 %, 5dHLU22 + 50 %, 14dCONT22 + 186 %, 14dHLU22 + 104 %, vs matching 28 °C group). In the femur diaphysis and at both temperatures, 14dHLU exhibited thinner cortices distally or proximally compared to controls; the mid-diaphysis being thicker at 28 °C than at 22 °C in all groups. Expression of cortical genes for proteolytic enzyme (Mmp13), markers for osteoclastogenic differentiation (MCSF, RANKL), and activity (TRAP, Ctsk) were increased following 22 °C HLU, whereas only Ctsk expression was increased following 28 °C HLU. Expression of cortical genes for apoptosis, senescence, and autophagy were not elevated following HLU at any temperature. Osteocyte density at the posterior mid-diaphysis was similar between groups, as was the proportion of empty lacunae (<0.5 %). However, analysis of the lacuno-canalicular network (LCN, fluorescein staining) revealed unstained areas in the 14dHLU22 group only, suggesting disrupted LCN flow in this group alone. In conclusion, 28 °C housing influences the HLU bone response but does not prevent bone loss. Furthermore, our results do not show osteocyte senescence or death, and at thermoneutrality, HLU-induced bone resorption is not triggered by osteoclastic activators RANKL and MCSF.
Collapse
Affiliation(s)
- Laura Peurière
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F-42023, Saint-Étienne, France.
| | - Carmelo Mastrandrea
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F-42023, Saint-Étienne, France
| | - Arnaud Vanden-Bossche
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F-42023, Saint-Étienne, France
| | - Marie-Thérèse Linossier
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F-42023, Saint-Étienne, France
| | - Mireille Thomas
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F-42023, Saint-Étienne, France
| | - Myriam Normand
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F-42023, Saint-Étienne, France
| | - Marie-Hélène Lafage-Proust
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F-42023, Saint-Étienne, France
| | - Laurence Vico
- Université Jean Monnet Saint-Étienne, Mines Saint-Étienne, INSERM, SAINBIOSE U1059, F-42023, Saint-Étienne, France
| |
Collapse
|
32
|
Iordachescu A, Eisenstein N, Appleby-Thomas G. Space habitats for bioengineering and surgical repair: addressing the requirement for reconstructive and research tissues during deep-space missions. NPJ Microgravity 2023; 9:23. [PMID: 36966158 PMCID: PMC10039948 DOI: 10.1038/s41526-023-00266-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 03/13/2023] [Indexed: 03/27/2023] Open
Abstract
Numerous technical scenarios have been developed to facilitate a human return to the Moon, and as a testbed for a subsequent mission to Mars. Crews appointed with constructing and establishing planetary bases will require a superior level of physical ability to cope with the operational demands. However, the challenging environments of nearby planets (e.g. geological, atmospheric, gravitational conditions) as well as the lengthy journeys through microgravity, will lead to progressive tissue degradation and an increased susceptibility to injury. The isolation, distance and inability to evacuate in an emergency will require autonomous medical support, as well as a range of facilities and specialised equipment to repair tissue damage on-site. Here, we discuss the design requirements of such a facility, in the form of a habitat that would concomitantly allow tissue substitute production, maintenance and surgical implantation, with an emphasis on connective tissues. The requirements for the individual modules and their operation are identified. Several concepts are assessed, including the presence of adjacent wet lab and medical modules supporting the gradual implementation of regenerative biomaterials and acellular tissue substitutes, leading to eventual tissue grafts and, in subsequent decades, potential tissues/organ-like structures. The latter, currently in early phases of development, are assessed particularly for researching the effects of extreme conditions on representative analogues for astronaut health support. Technical solutions are discussed for bioengineering in an isolated planetary environment with hypogravity, from fluid-gel bath suspended manufacture to cryostorage, cell sourcing and on-site resource utilisation for laboratory infrastructure. Surgical considerations are also discussed.
Collapse
Affiliation(s)
- Alexandra Iordachescu
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
- Consortium for organotypic research on ageing and microgravity, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
- Healthcare Technologies Institute, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
- Cranfield Defence and Security, Cranfield University, Defence Academy of the United Kingdom, Shrivenham, SN6 8LA, United Kingdom.
| | - Neil Eisenstein
- Healthcare Technologies Institute, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
| | - Gareth Appleby-Thomas
- Consortium for organotypic research on ageing and microgravity, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom
- Cranfield Defence and Security, Cranfield University, Defence Academy of the United Kingdom, Shrivenham, SN6 8LA, United Kingdom
| |
Collapse
|
33
|
Capri M, Conte M, Ciurca E, Pirazzini C, Garagnani P, Santoro A, Longo F, Salvioli S, Lau P, Moeller R, Jordan J, Illig T, Villanueva MM, Gruber M, Bürkle A, Franceschi C, Rittweger J. Long-term human spaceflight and inflammaging: Does it promote aging? Ageing Res Rev 2023; 87:101909. [PMID: 36918115 DOI: 10.1016/j.arr.2023.101909] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Spaceflight and its associated stressors, such as microgravity, radiation exposure, confinement, circadian derailment and disruptive workloads represent an unprecedented type of exposome that is entirely novel from an evolutionary stand point. Within this perspective, we aimed to review the effects of prolonged spaceflight on immune-neuroendocrine systems, brain and brain-gut axis, cardiovascular system and musculoskeletal apparatus, highlighting in particular the similarities with an accelerated aging process. In particular, spaceflight-induced muscle atrophy/sarcopenia and bone loss, vascular and metabolic changes, hyper and hypo reaction of innate and adaptive immune system appear to be modifications shared with the aging process. Most of these modifications are mediated by molecular events that include oxidative and mitochondrial stress, autophagy, DNA damage repair and telomere length alteration, among others, which directly or indirectly converge on the activation of an inflammatory response. According to the inflammaging theory of aging, such an inflammatory response could be a driver of an acceleration of the normal, physiological rate of aging and it is likely that all the systemic modifications in turn lead to an increase of inflammaging in a sort of vicious cycle. The most updated countermeasures to fight these modifications will be also discussed in the light of their possible application not only for astronauts' benefit, but also for older adults on the ground.
Collapse
Affiliation(s)
- Miriam Capri
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | - Maria Conte
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy.
| | - Erika Ciurca
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Chiara Pirazzini
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Paolo Garagnani
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy; Clinical Chemistry Department of Laboratory Medicine, Karolinska Institutet at Huddinge University Hospital, Stockholm, Sweden; CNR Institute of Molecular Genetics, Unit of Bologna, Bologna, Italy; Center for Applied Biomedical Research (CRBA), St. Orsola-Malpighi University Hospital, Bologna, Italy
| | - Aurelia Santoro
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate), University of Bologna, Bologna, Italy
| | - Federica Longo
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy
| | - Stefano Salvioli
- Department of Medical and Surgical Science, University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Patrick Lau
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Ralf Moeller
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Medical Faculty, University of Cologne, Cologne, Germany
| | - Thomas Illig
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Maria-Moreno Villanueva
- Human Performance Research Centre, Department of Sport Science, University of Konstanz, Konstanz, Germany
| | - Markus Gruber
- Human Performance Research Centre, Department of Sport Science, University of Konstanz, Konstanz, Germany
| | - Alexander Bürkle
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Claudio Franceschi
- Department of Applied Mathematics of the Institute of ITMM, National Research Lobachevsky State University of Nizhny Novgorod, the Russian Federation
| | - Jörn Rittweger
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
34
|
Wang J, Shang P. Static magnetic field: A potential tool of controlling stem cells fates for stem cell therapy in osteoporosis. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 178:91-102. [PMID: 36596343 DOI: 10.1016/j.pbiomolbio.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/10/2022] [Accepted: 12/29/2022] [Indexed: 01/01/2023]
Abstract
Osteoporosis is a kind of bone diseases characterized by dynamic imbalance of bone formation and bone absorption, which is prone to fracture, and seriously endangers human health. At present, there is a lack of highly effective drugs for it, and the existing measures all have some side effects. In recent years, mesenchymal stem cell therapy has brought a certain hope for osteoporosis, while shortcomings such as homing difficulty and unstable therapeutic effects limit its application widely. Therefore, it is extremely urgent to find effective and reliable means/drugs for adjuvant stem cell therapy or develop new research techniques. It has been reported that static magnetic fields(SMFs) has a certain alleviating and therapeutic effect on varieties of bone diseases, also promotes the proliferation and osteogenic differentiation of mesenchymal stem cells derived from different tissues to a certain extent. Basing on the above background, this article focuses on the key words "static/constant magnetic field, mesenchymal stem cell, osteoporosis", combined literature and relevant contents were studied to look forward that SMFs has unique advantages in the treatment of osteoporosis with mesenchymal stem cells, which can be used as an application tool to promote the progress of stem cell therapy in clinical application.
Collapse
Affiliation(s)
- Jianping Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China
| | - Peng Shang
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen, 518057, China; School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China; Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, China.
| |
Collapse
|
35
|
Sakharkar A, Yang J. Designing a Novel Monitoring Approach for the Effects of Space Travel on Astronauts' Health. Life (Basel) 2023; 13:life13020576. [PMID: 36836933 PMCID: PMC9964234 DOI: 10.3390/life13020576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/22/2023] Open
Abstract
Space exploration and extraterrestrial civilization have fascinated humankind since the earliest days of human history. It was only in the last century that humankind finally began taking significant steps towards these goals by sending astronauts into space, landing on the moon, and building the International Space Station. However, space voyage is very challenging and dangerous, and astronauts are under constant space radiation and microgravity. It has been shown that astronauts are at a high risk of developing a broad range of diseases/disorders. Thus, it is critical to develop a rapid and effective assay to monitor astronauts' health in space. In this study, gene expression and correlation patterns were analyzed for 10 astronauts (8 male and 2 female) using the publicly available microarray dataset E-GEOD-74708. We identified 218 differentially expressed genes between In-flight and Pre-flight and noticed that space travel decreased genome regulation and gene correlations across the entire genome, as well as individual signaling pathways. Furthermore, we systematically developed a shortlist of 32 genes that could be used to monitor astronauts' health during space travel. Further studies, including microgravity experiments, are warranted to optimize and validate the proposed assay.
Collapse
Affiliation(s)
- Anurag Sakharkar
- College of Arts and Science, University of Saskatchewan, 9 Campus Drive, Saskatoon, SK S7N 5A5, Canada
| | - Jian Yang
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada
- Correspondence:
| |
Collapse
|
36
|
Wang J, Zhao B, Che J, Shang P. Hypoxia Pathway in Osteoporosis: Laboratory Data for Clinical Prospects. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:3129. [PMID: 36833823 PMCID: PMC9963321 DOI: 10.3390/ijerph20043129] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/02/2023] [Accepted: 02/04/2023] [Indexed: 05/29/2023]
Abstract
The hypoxia pathway not only regulates the organism to adapt to the special environment, such as short-term hypoxia in the plateau under normal physiological conditions, but also plays an important role in the occurrence and development of various diseases such as cancer, cardiovascular diseases, osteoporosis. Bone, as a special organ of the body, is in a relatively low oxygen environment, in which the expression of hypoxia-inducible factor (HIF)-related molecules maintains the necessary conditions for bone development. Osteoporosis disease with iron overload endangers individuals, families and society, and bone homeostasis disorder is linked to some extent with hypoxia pathway abnormality, so it is urgent to clarify the hypoxia pathway in osteoporosis to guide clinical medication efficiently. Based on this background, using the keywords "hypoxia/HIF, osteoporosis, osteoblasts, osteoclasts, osteocytes, iron/iron metabolism", a matching search was carried out through the Pubmed and Web Of Science databases, then the papers related to this review were screened, summarized and sorted. This review summarizes the relationship and regulation between the hypoxia pathway and osteoporosis (also including osteoblasts, osteoclasts, osteocytes) by arranging the references on the latest research progress, introduces briefly the application of hyperbaric oxygen therapy in osteoporosis symptoms (mechanical stimulation induces skeletal response to hypoxic signal activation), hypoxic-related drugs used in iron accumulation/osteoporosis model study, and also puts forward the prospects of future research.
Collapse
Affiliation(s)
- Jianping Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Bin Zhao
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Jingmin Che
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Peng Shang
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
- Research & Development Institute in Shenzhen, Northwestern Polytechnical University, Shenzhen 518057, China
| |
Collapse
|
37
|
Liu X, Yan Z, Cai J, Wang D, Yang Y, Ding Y, Shao X, Hao X, Luo E, Guo XE, Luo P, Shen L, Jing D. Glucose- and glutamine-dependent bioenergetics sensitize bone mechanoresponse after unloading by modulating osteocyte calcium dynamics. J Clin Invest 2023; 133:164508. [PMID: 36512405 PMCID: PMC9888392 DOI: 10.1172/jci164508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Disuse osteoporosis is a metabolic bone disease resulting from skeletal unloading (e.g., during extended bed rest, limb immobilization, and spaceflight), and the slow and insufficient bone recovery during reambulation remains an unresolved medical challenge. Here, we demonstrated that loading-induced increase in bone architecture/strength was suppressed in skeletons previously exposed to unloading. This reduction in bone mechanosensitivity was directly associated with attenuated osteocytic Ca2+ oscillatory dynamics. The unloading-induced compromised osteocytic Ca2+ response to reloading resulted from the HIF-1α/PDK1 axis-mediated increase in glycolysis, and a subsequent reduction in ATP synthesis. HIF-1α also transcriptionally induced substantial glutaminase 2 expression and thereby glutamine addiction in osteocytes. Inhibition of glycolysis by blockade of PDK1 or glutamine supplementation restored the mechanosensitivity in those skeletons with previous unloading by fueling the tricarboxylic acid cycle and rescuing subsequent Ca2+ oscillations in osteocytes. Thus, we provide mechanistic insight into disuse-induced deterioration of bone mechanosensitivity and a promising therapeutic approach to accelerate bone recovery after long-duration disuse.
Collapse
Affiliation(s)
- Xiyu Liu
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Zedong Yan
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Jing Cai
- College of Basic Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Dan Wang
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Yongqing Yang
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Yuanjun Ding
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Xi Shao
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Xiaoxia Hao
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - Erping Luo
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China
| | - X. Edward Guo
- Bone Bioengineering Laboratory, Department of Biomedical Engineering, Columbia University, New York, New York, USA
| | - Peng Luo
- Department of Neurosurgery, Xijing Hospital
| | - Liangliang Shen
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology
| | - Da Jing
- Department of Biomedical Engineering, Fourth Military Medical University, Xi’an, China.,Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, and,Shaanxi Provincial Key Laboratory of Bioelectromagnetic Detection and Intelligent Perception, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
38
|
Hirayama J, Hattori A, Takahashi A, Furusawa Y, Tabuchi Y, Shibata M, Nagamatsu A, Yano S, Maruyama Y, Matsubara H, Sekiguchi T, Suzuki N. Physiological consequences of space flight, including abnormal bone metabolism, space radiation injury, and circadian clock dysregulation: Implications of melatonin use and regulation as a countermeasure. J Pineal Res 2023; 74:e12834. [PMID: 36203395 DOI: 10.1111/jpi.12834] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 12/15/2022]
Abstract
Exposure to the space environment induces a number of pathophysiological outcomes in astronauts, including bone demineralization, sleep disorders, circadian clock dysregulation, cardiovascular and metabolic dysfunction, and reduced immune system function. A recent report describing experiments aboard the Space Shuttle mission, STS-132, showed that the level of melatonin, a hormone that provides the biochemical signal of darkness, was decreased during microgravity in an in vitro culture model. Additionally, abnormal lighting conditions in outer space, such as low light intensity in orbital spacecraft and the altered 24-h light-dark cycles, may result in the dysregulation of melatonin rhythms and the misalignment of the circadian clock from sleep and work schedules in astronauts. Studies on Earth have demonstrated that melatonin regulates various physiological functions including bone metabolism. These data suggest that the abnormal regulation of melatonin in outer space may contribute to pathophysiological conditions of astronauts. In addition, experiments with high-linear energy transfer radiation, a ground-based model of space radiation, showed that melatonin may serve as a protectant against space radiation. Gene expression profiling using an in vitro culture model exposed to space flight during the STS-132 mission, showed that space radiation alters the expression of DNA repair and oxidative stress response genes, indicating that melatonin counteracts the expression of these genes responsive to space radiation to promote cell survival. These findings implicate the use of exogenous melatonin and the regulation of endogenous melatonin as countermeasures for the physiological consequences of space flight.
Collapse
Affiliation(s)
- Jun Hirayama
- Department of Clinical Engineering, Faculty of Health Sciences & Division of Health Sciences, Graduate School of Sustainable Systems Science, Komatsu University, Komatsu, Japan
| | - Atsuhiko Hattori
- Department of Biology, College of Liberal Arts and Sciences, Tokyo Medical and Dental University, Ichikawa, Japan
| | | | - Yukihiro Furusawa
- Department of Pharmaceutical Engineering, Faculty of Engineering, Toyama Prefectural University, Toyama, Japan
| | - Yoshiaki Tabuchi
- Life Science Research Center, University of Toyama, Toyama, Japan
| | - Masahiro Shibata
- Department of Biology, College of Liberal Arts and Sciences, Tokyo Medical and Dental University, Ichikawa, Japan
| | | | - Sachiko Yano
- Japan Aerospace Exploration Agency, Tsukuba, Japan
| | - Yusuke Maruyama
- Department of Biology, College of Liberal Arts and Sciences, Tokyo Medical and Dental University, Ichikawa, Japan
| | - Hajime Matsubara
- Noto Center for Fisheries Science and Technology, Kanazawa University, Noto-cho, Ishikawa, Japan
| | - Toshio Sekiguchi
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Noto-cho, Japan
| | - Nobuo Suzuki
- Noto Marine Laboratory, Institute of Nature and Environmental Technology, Kanazawa University, Noto-cho, Japan
| |
Collapse
|
39
|
Zhang G, Zhen C, Yang J, Zhang Z, Wu Y, Che J, Shang P. 1–2 T static magnetic field combined with Ferumoxytol prevent unloading-induced bone loss by regulating iron metabolism in osteoclastogenesis. J Orthop Translat 2023; 38:126-140. [DOI: 10.1016/j.jot.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
|
40
|
Murphy P, Rolfe RA. Building a Co-ordinated Musculoskeletal System: The Plasticity of the Developing Skeleton in Response to Muscle Contractions. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2023; 236:81-110. [PMID: 37955772 DOI: 10.1007/978-3-031-38215-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
The skeletal musculature and the cartilage, bone and other connective tissues of the skeleton are intimately co-ordinated. The shape, size and structure of each bone in the body is sculpted through dynamic physical stimuli generated by muscle contraction, from early development, with onset of the first embryo movements, and through repair and remodelling in later life. The importance of muscle movement during development is shown by congenital abnormalities where infants that experience reduced movement in the uterus present a sequence of skeletal issues including temporary brittle bones and joint dysplasia. A variety of animal models, utilising different immobilisation scenarios, have demonstrated the precise timing and events that are dependent on mechanical stimulation from movement. This chapter lays out the evidence for skeletal system dependence on muscle movement, gleaned largely from mouse and chick immobilised embryos, showing the many aspects of skeletal development affected. Effects are seen in joint development, ossification, the size and shape of skeletal rudiments and tendons, including compromised mechanical function. The enormous plasticity of the skeletal system in response to muscle contraction is a key factor in building a responsive, functional system. Insights from this work have implications for our understanding of morphological evolution, particularly the challenging concept of emergence of new structures. It is also providing insight for the potential of physical therapy for infants suffering the effects of reduced uterine movement and is enhancing our understanding of the cellular and molecular mechanisms involved in skeletal tissue differentiation, with potential for informing regenerative therapies.
Collapse
Affiliation(s)
- Paula Murphy
- School of Natural Sciences, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland.
| | - Rebecca A Rolfe
- School of Natural Sciences, Trinity College Dublin, The University of Dublin, Dublin 2, Ireland
| |
Collapse
|
41
|
Yang J, Feng Y, Li Q, Zeng Y. Evidence of the static magnetic field effects on bone-related diseases and bone cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:168-180. [PMID: 36462638 DOI: 10.1016/j.pbiomolbio.2022.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/28/2022] [Accepted: 11/30/2022] [Indexed: 12/03/2022]
Abstract
Static magnetic fields (SMFs), magnetic fields with constant intensity and orientation, have been extensively studied in the field of bone biology both fundamentally and clinically as a non-invasive physical factor. A large number of animal experiments and clinical studies have shown that SMFs have effective therapeutic effects on bone-related diseases such as non-healing fractures, bone non-union of bone implants, osteoporosis and osteoarthritis. The maintenance of bone health in adults depends on the basic functions of bone cells, such as bone formation by osteoblasts and bone resorption by osteoclasts. Numerous studies have revealed that SMFs can regulate the proliferation, differentiation, and function of bone tissue cells, including bone marrow mesenchymal stem cells (BMSCs), osteoblasts, bone marrow monocytes (BMMs), osteoclasts, and osteocytes. In this paper, the effects of SMFs on bone-related diseases and bone tissue cells are reviewed from both in vivo studies and in vitro studies, and the possible mechanisms are analyzed. In addition, some challenges that need to be further addressed in the research of SMF and bone are also discussed.
Collapse
Affiliation(s)
- Jiancheng Yang
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yan Feng
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qingmei Li
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yuhong Zeng
- Department of Osteoporosis, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
42
|
Bezerra A, Freitas L, Maciel L, Fonseca H. Bone Tissue Responsiveness To Mechanical Loading-Possible Long-Term Implications of Swimming on Bone Health and Bone Development. Curr Osteoporos Rep 2022; 20:453-468. [PMID: 36401774 DOI: 10.1007/s11914-022-00758-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/29/2022] [Indexed: 11/21/2022]
Abstract
PURPOSE OF REVIEW To revisit the bone tissue mechanotransduction mechanisms behind the bone tissue response to mechanical loading and, within this context, explore the possible negative influence of regular swimming practice on bone health, particularly during the growth and development period. RECENT FINDINGS Bone is a dynamic tissue, responsive to mechanical loading and unloading, being these adaptative responses more intense during the growth and development period. Cross-sectional studies usually report a lower bone mass in swimmers compared to athletes engaged in weigh-bearing sports. However, studies with animal models show contradictory findings about the effect of swimming on bone health, highlighting the need for longitudinal studies. Due to its microgravity characteristics, swimming seems to impair bone mass, but mostly at the lower limbs. It is unkown if there is a causal relationship between swimming and low BMD or if other confounding factors, such as a natural selection whithin the sport, are the cause.
Collapse
Affiliation(s)
- Andréa Bezerra
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto (FADE/UP), 4200-450, Porto, Portugal.
- Laboratory for Integrative and Translational Research in Population Health (ITR), 4050-600, Porto, Portugal.
| | - Laura Freitas
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto (FADE/UP), 4200-450, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), 4050-600, Porto, Portugal
| | - Leonardo Maciel
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto (FADE/UP), 4200-450, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), 4050-600, Porto, Portugal
- Department of Physiotherapy, Federal University of Sergipe, Campus Lagarto, Lagarto, Brazil
| | - Hélder Fonseca
- Research Centre in Physical Activity, Health and Leisure (CIAFEL), Faculty of Sport, University of Porto (FADE/UP), 4200-450, Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), 4050-600, Porto, Portugal
| |
Collapse
|
43
|
Arbeille P, Greaves D, Guillon L, Hughson RL. 4 days in dry immersion increases arterial wall response to ultrasound wave as measured using radio-frequency signal, comparison with spaceflight data. Front Physiol 2022; 13:983837. [DOI: 10.3389/fphys.2022.983837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
Recent studies have reported a significant increase in common carotid artery (CCA) intima media thickness, wall stiffness and reflectivity to ultrasound, in astronauts, after six months of spaceflight. The hypothesis was that 4 days in dry immersion (subjects under bags of water) will be sufficient to change the CCA wall reflectivity to ultrasound similar to what observed after spaceflight. Such response would be quantified using the amplitude of the ultrasound signal returned to the probe by the target concerned. [coefficient of signal return (Rs)]. The Rs for anterior and posterior CCA wall, sternocleidomastoid muscle, intima layer and CCA lumen were calculated from the ultrasound radio frequency (RF) data displayed along each echographic line. After four days of DI, Rs increased in the CCA posterior wall (+15% +/- 10 from pre DI, p < 0.05), while no significant change was observed in the other targets. The observed increase in Rs with DI was approximately half compared to what was observed after six months of space flight (+34% +/- 14). This difference may be explained by dose response (dry immersion only four days in duration). As a marker of tissue-level physical changes, Rs provide complimentary information alongside previously observed CCA wall thickness and stiffness.
Collapse
|
44
|
Ogneva IV. Single Cell in a Gravity Field. Life (Basel) 2022; 12:1601. [PMID: 36295035 PMCID: PMC9604728 DOI: 10.3390/life12101601] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/09/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023] Open
Abstract
The exploration of deep space or other bodies of the solar system, associated with a long stay in microgravity or altered gravity, requires the development of fundamentally new methods of protecting the human body. Most of the negative changes in micro- or hypergravity occur at the cellular level; however, the mechanism of reception of the altered gravity and transduction of this signal, leading to the formation of an adaptive pattern of the cell, is still poorly understood. At the same time, most of the negative changes that occur in early embryos when the force of gravity changes almost disappear by the time the new organism is born. This review is devoted to the responses of early embryos and stem cells, as well as terminally differentiated germ cells, to changes in gravity. An attempt was made to generalize the data presented in the literature and propose a possible unified mechanism for the reception by a single cell of an increase and decrease in gravity based on various deformations of the cortical cytoskeleton.
Collapse
Affiliation(s)
- Irina V Ogneva
- Cell Biophysics Laboratory, State Scientific Center of the Russian Federation Institute of Biomedical Problems of the Russian Academy of Sciences, 76a, Khoroshevskoyoe Shosse, 123007 Moscow, Russia
| |
Collapse
|
45
|
Plehuna A, Green DA, Amirova LE, Tomilovskaya ES, Rukavishnikov IV, Kozlovskaya IB. Dry immersion induced acute low back pain and its relationship with trunk myofascial viscoelastic changes. Front Physiol 2022; 13:1039924. [PMID: 36311233 PMCID: PMC9606241 DOI: 10.3389/fphys.2022.1039924] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 09/26/2022] [Indexed: 08/12/2023] Open
Abstract
Microgravity induces spinal elongation and Low Back Pain (LBP) but the pathophysiology is unknown. Changes in paraspinal muscle viscoelastic properties may play a role. Dry Immersion (DI) is a ground-based microgravity analogue that induces changes in m. erector spinae superficial myofascial tissue tone within 2 h. This study sought to determine whether bilateral m. erector spinae tone, creep, and stiffness persist beyond 2 h; and if such changes correlate with DI-induced spinal elongation and/or LBP. Ten healthy males lay in the DI bath at the Institute of Biomedical Problems (Moscow, Russia) for 6 h. Bilateral lumbar (L1, L4) and thoracic (T11, T9) trunk myofascial tone, stiffness and creep (MyotonPRO), and subjective LBP (0-10 NRS) were recorded before DI, after 1h, 6 h of DI, and 30min post. The non-standing spinal length was evaluated on the bath lifting platform using a bespoke stadiometer before and following DI. DI significantly modulated m. erector spinae viscoelastic properties at L4, L1, T11, and T9 with no effect of laterality. Bilateral tissue tone was significantly reduced after 1 and 6 h DI at L4, L1, T11, and T9 to a similar extent. Stiffness was also reduced by DI at 1 h but partially recovered at 6 h for L4, L1, and T11. Creep was increased by DI at 1 h, with partial recovery at 6 h, although only T11 was significant. All properties returned to baseline 30 min following DI. Significant spinal elongation (1.17 ± 0.20 cm) with mild (at 1 h) to moderate (at 6 h) LBP was induced, mainly in the upper lumbar and lower thoracic regions. Spinal length increases positively correlated (Rho = 0.847, p = 0.024) with middle thoracic (T9) tone reduction, but with no other stiffness or creep changes. Spinal length positively correlated (Rho = 0.557, p = 0.039) with Max LBP; LBP failed to correlate with any m. erector spinae measured parameters. The DI-induced bilateral m. erector spinae tone, creep, and stiffness changes persist beyond 2 h. Evidence of spinal elongation and LBP allows suggesting that the trunk myofascial tissue changes could play a role in LBP pathogenesis observed in real and simulated microgravity. Further study is warranted with longer duration DI, assessment of IVD geometry, and vertebral column stability.
Collapse
Affiliation(s)
- Anastasija Plehuna
- King’s College London, Centre of Human & Applied Physiological Sciences, London, United Kingdom
- Laboratory of Gravitational Physiology of the Sensorimotor System, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - David Andrew Green
- King’s College London, Centre of Human & Applied Physiological Sciences, London, United Kingdom
- Space Medicine Team, HRE-OM, European Astronaut Centre, European Space Agency, Cologne, Germany
- KBRwyle Laboratories GmbH, Cologne, Germany
| | - Liubov E. Amirova
- Laboratory of Gravitational Physiology of the Sensorimotor System, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Elena S. Tomilovskaya
- Laboratory of Gravitational Physiology of the Sensorimotor System, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Ilya V. Rukavishnikov
- Laboratory of Gravitational Physiology of the Sensorimotor System, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Inessa B. Kozlovskaya
- Laboratory of Gravitational Physiology of the Sensorimotor System, Institute of Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
46
|
Sharma N, Weivoda MM, Søe K. Functional Heterogeneity Within Osteoclast Populations-a Critical Review of Four Key Publications that May Change the Paradigm of Osteoclasts. Curr Osteoporos Rep 2022; 20:344-355. [PMID: 35838878 DOI: 10.1007/s11914-022-00738-7] [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] [Accepted: 06/26/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW In this review, we critically evaluate the literature for osteoclast heterogeneity, including heterogeneity in osteoclast behavior, which has hitherto been unstudied and has only recently come to attention. We give a critical review centered on four recent high-impact papers on this topic and aim to shed light on the elusive biology of osteoclasts and focus on the variant features of osteoclasts that diverge from the classical viewpoint. RECENT FINDINGS Osteoclasts originate from the myeloid lineage and are best known for their unique ability to resorb bone. For decades, osteoclasts have been defined simply as multinucleated cells positive for tartrate-resistant acid phosphatase activity and quantified relative to the bone perimeter or surface in histomorphometric analyses. However, several recent, high-profile studies have demonstrated the existence of heterogeneous osteoclast populations, with variable origins and functions depending on the microenvironment. This includes long-term persisting osteoclasts, inflammatory osteoclasts, recycling osteoclasts (osteomorphs), and bone resorption modes. Most of these findings have been revealed through murine studies and have helped identify new targets for human studies. These studies have also uncovered distinct sets of behavioral patterns in heterogeneous osteoclast cultures. The underlying osteoclast heterogeneity likely drives differences in bone remodeling, altering patient risk for osteoporosis and fracture. Thus, identifying the underlying key features of osteoclast heterogeneity may help in better targeting bone diseases.
Collapse
Affiliation(s)
- Neha Sharma
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark
- Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 1. Floor, 5000, Odense C, Denmark
| | | | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark.
- Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
- Department of Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 1. Floor, 5000, Odense C, Denmark.
| |
Collapse
|
47
|
Camy C, Brioche T, Senni K, Bertaud A, Genovesio C, Lamy E, Fovet T, Chopard A, Pithioux M, Roffino S. Effects of hindlimb unloading and subsequent reloading on the structure and mechanical properties of Achilles tendon-to-bone attachment. FASEB J 2022; 36:e22548. [PMID: 36121701 DOI: 10.1096/fj.202200713r] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/10/2022] [Accepted: 09/02/2022] [Indexed: 11/11/2022]
Abstract
While muscle and bone adaptations to deconditioning have been widely described, few studies have focused on the tendon enthesis. Our study examined the effects of mechanical loading on the structure and mechanical properties of the Achilles tendon enthesis. We assessed the fibrocartilage surface area, the organization of collagen, the expression of collagen II, the presence of osteoclasts, and the tensile properties of the mouse enthesis both after 14 days of hindlimb suspension (HU) and after a subsequent 6 days of reloading. Although soleus atrophy was severe after HU, calcified fibrocartilage (CFc) was a little affected. In contrast, we observed a decrease in non-calcified fibrocartilage (UFc) surface area, collagen fiber disorganization, modification of morphological characteristics of the fibrocartilage cells, and altered collagen II distribution. Compared to the control group, restoring normal loads increased both UFc surface area and expression of collagen II, and led to a crimp pattern in collagen. Reloading induced an increase in CFc surface area, probably due to the mineralization front advancing toward the tendon. Functionally, unloading resulted in decreased enthesis stiffness and a shift in site of failure from the osteochondral interface to the bone, whereas 6 days of reloading restored the original elastic properties and site of failure. In the context of spaceflight, our results suggest that care must be taken when performing countermeasure exercises both during missions and during the return to Earth.
Collapse
Affiliation(s)
- Claire Camy
- Aix Marseille University, CNRS, ISM, Institute of Movement Sciences, Marseille, France
| | - Thomas Brioche
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | - Karim Senni
- Laboratoire EBInnov, Ecole de Biologie Industrielle-EBI, Cergy, France
| | - Alexandrine Bertaud
- Aix Marseille Univ, INSERM, INRAE, C2VN, Marseille, France.,Laboratoire de Biochimie, Faculté de Pharmacie, Marseille, France
| | - Cécile Genovesio
- Laboratoire de Biochimie, Faculté de Pharmacie, Marseille, France
| | - Edouard Lamy
- Aix Marseille University, CNRS, ISM, Institute of Movement Sciences, Marseille, France.,Laboratoire de Biochimie, Faculté de Pharmacie, Marseille, France
| | - Théo Fovet
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | - Angèle Chopard
- DMEM, Montpellier University, INRAE, UMR 866, Montpellier, France
| | - Martine Pithioux
- Aix Marseille University, CNRS, ISM, Institute of Movement Sciences, Marseille, France.,Department of Orthopaedics and Traumatology, Aix Marseille Univ, APHM, CNRS, ISM, Sainte-Marguerite Hospital, Institute for Locomotion, Marseille, France.,Aix Marseille Univ, APHM, CNRS, Centrale Marseille, ISM, Mecabio Platform, Anatomy Laboratory, Timone, Marseille, France
| | - Sandrine Roffino
- Aix Marseille University, CNRS, ISM, Institute of Movement Sciences, Marseille, France.,Aix Marseille Univ, APHM, CNRS, Centrale Marseille, ISM, Mecabio Platform, Anatomy Laboratory, Timone, Marseille, France
| |
Collapse
|
48
|
Multi-system responses to altered gravity and spaceflight: Insights from Drosophila melanogaster. Neurosci Biobehav Rev 2022; 142:104880. [PMID: 36126744 DOI: 10.1016/j.neubiorev.2022.104880] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 09/12/2022] [Accepted: 09/14/2022] [Indexed: 11/21/2022]
Abstract
NASA is planning to resume human-crewed lunar missions and lay the foundation for human exploration to Mars. However, our knowledge of the overall effects of long-duration spaceflight on human physiology is limited. During spaceflight, astronauts are exposed to multiple risk factors, including gravitational changes, ionizing radiation, physiological stress, and altered circadian lighting. These factors contribute to pathophysiological responses that target different organ systems in the body. This review discusses the advancements in gravitational biology using Drosophila melanogaster, one of the first organisms to be launched into space. As a well-established spaceflight model organism, fruit flies have yielded significant information, including neurobehavioral, aging, immune, cardiovascular, developmental, and multi-omics changes across tissues and developmental stages, as detailed in this review.
Collapse
|
49
|
Horeau M, Ropert M, Mulder E, Tank J, Frings-Meuthen P, Armbrecht G, Loréal O, Derbré F. Iron Metabolism Regulation in Females and Males Exposed to Simulated Microgravity: results from the Randomized Trial AGBRESA. Am J Clin Nutr 2022; 116:1430-1440. [PMID: 36026525 DOI: 10.1093/ajcn/nqac205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/23/2022] [Accepted: 08/14/2022] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Iron metabolism imbalance could contribute to physical deconditioning experienced by astronauts due to its essential role in energy metabolism, cellular respiration, and oxygen transport. OBJECTIVES In this clinical exploratory study, we wanted to determine whether artificial gravity (AG) training modulated iron metabolism, red blood cell indices, and body lean mass in male and female healthy participants exposed to head-down tilt (HDT) bed rest, the reference ground-based model of microgravity. METHODS We recruited 8 female and 16 male healthy participants who were all exposed to HDT bed rest for 60 days. In addition, they were assigned to three experimental groups (n = 8/each): controls, continuous AG training in a short-arm centrifuge (1×30 min/day), and intermittent AG training (6 × 5 min/day). RESULTS The iron metabolism responses to simulated microgravity of AG training groups do not significantly differ from the responses of controls. Independently from AG, we found that both serum iron (+31.3%, P = 0.027) and transferrin saturation levels (+28.4%, P = 0.009) increased in males after 6 days of HDT bed rest, as well as serum hepcidin levels (+36.9% P = 0.005). The increase of transferrin saturation levels persisted after 57 days of HDT bed rest (+13.5%, P = 0.026), suggesting that long-term exposure to microgravity sustainably increases serum iron availability in males, and consequently the risk of iron excess or misdistribution. In females, 6 and 57 days of HDT bed rest did not significantly change serum iron, transferrin saturation, and hepcidin levels. CONCLUSIONS The data from this exploratory study suggest that 1) AG training does not influence the iron metabolism responses to microgravity; 2) iron metabolism parameters, especially iron availability for cells, are significantly increased in males, but not in females, exposed to long-term simulated microgravity. Due to the small sample size of females, we nevertheless must be cautious before concluding that iron metabolism could differently respond to microgravity in females. Clinical trial registry number: DRKS00015677.
Collapse
Affiliation(s)
- Mathieu Horeau
- Laboratory "Movement Sport and Health Sciences" EA7470, University of Rennes/ENS Rennes, France.,INSERM, University of Rennes, INRAE, UMR 1241, AEM2 platform, Nutrition Metabolisms and Cancer (NuMeCan) institute, Rennes, France
| | - Martine Ropert
- INSERM, University of Rennes, INRAE, UMR 1241, AEM2 platform, Nutrition Metabolisms and Cancer (NuMeCan) institute, Rennes, France.,Department of Biochemistry, CHU Rennes, France
| | - Edwin Mulder
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Jens Tank
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Petra Frings-Meuthen
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Gabriele Armbrecht
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Radiology, Berlin, Germany
| | - Olivier Loréal
- INSERM, University of Rennes, INRAE, UMR 1241, AEM2 platform, Nutrition Metabolisms and Cancer (NuMeCan) institute, Rennes, France
| | - Frédéric Derbré
- Laboratory "Movement Sport and Health Sciences" EA7470, University of Rennes/ENS Rennes, France
| |
Collapse
|
50
|
Peng H, Hu B, Xie LQ, Su T, Li CJ, Liu Y, Yang M, Xiao Y, Feng X, Zhou R, Guo Q, Zhou HY, Huang Y, Jiang TJ, Luo XH. A mechanosensitive lipolytic factor in the bone marrow promotes osteogenesis and lymphopoiesis. Cell Metab 2022; 34:1168-1182.e6. [PMID: 35705079 DOI: 10.1016/j.cmet.2022.05.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 04/29/2022] [Accepted: 05/20/2022] [Indexed: 12/19/2022]
Abstract
Exercise can prevent osteoporosis and improve immune function, but the mechanism remains unclear. Here, we show that exercise promotes reticulocalbin-2 secretion from the bone marrow macrophages to initiate bone marrow fat lipolysis. Given the crucial role of lipolysis in exercise-stimulated osteogenesis and lymphopoiesis, these findings suggest that reticulocalbin-2 is a pivotal regulator of a local adipose-osteogenic/immune axis. Mechanistically, reticulocalbin-2 binds to a functional receptor complex, which is composed of neuronilin-2 and integrin beta-1, to activate a cAMP-PKA signaling pathway that mobilizes bone marrow fat via lipolysis to fuel the differentiation and function of mesenchymal and hematopoietic stem cells. Notably, the administration of recombinant reticulocalbin-2 in tail-suspended and old mice remarkably decreases bone marrow fat accumulation and promotes osteogenesis and lymphopoiesis. These findings identify reticulocalbin-2 as a novel mechanosensitive lipolytic factor in maintaining energy homeostasis in bone resident cells, and it provides a promising target for skeletal and immune health.
Collapse
Affiliation(s)
- Hui Peng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ling-Qi Xie
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Tian Su
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Chang-Jun Li
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ya Liu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Mi Yang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Ye Xiao
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xu Feng
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Rui Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Qi Guo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Hai-Yan Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Yan Huang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Tie-Jian Jiang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xiang-Hang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China; Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Hunan 410008, China.
| |
Collapse
|