1
|
Ding Y, Xu Y, Wang X, Zhang M, Zeng Q, Yan C, Zhou G, Hu W. Microgravity alleviates low-dose radiation-induced non-targeted carcinogenic effects. NPJ Microgravity 2025; 11:26. [PMID: 40527880 DOI: 10.1038/s41526-025-00484-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 06/06/2025] [Indexed: 06/20/2025] Open
Abstract
The main hazards astronauts face in space collectively affect their health, especially increasing the carcinogenesis risk. However, it is still unclear how these hazards, especially microgravity and space radiation, induce the carcinogenic transformation of normal cells. In the simulated microgravity (SMG) environment, although radiation could inhibit SMG-accentuated target cell proliferation, increase genomic instability (GI) and carcinogenic transformation rate dose-dependently, we found that for bystander cells, radiation-induced damage could be reduced, GI and the probability of carcinogenic transformation could also be decreased at lower doses (below 0.1 Gy for X-rays and 0.3 Gy for carbon ions). After filtration and KEGG analysis, five differentially expressed genes (DEGs) relating to carcinogenesis were screened out from the transcriptomic sequencing results. Based on the Cancer Genome Atlas (TCGA) from NCI, we found that AREG was closely related to the occurrence and development of lung cancer. Using AREG knockdown or overexpression cell lines, we further validated the significant correlation between abnormal expression of AREG and GI. Our findings indicate that AREG plays a substantial role in GI and carcinogenic transformation following exposure to SMG and radiation.
Collapse
Affiliation(s)
- Yunan Ding
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Ying Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Xiaofei Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Miaomiao Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Qi Zeng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Congchong Yan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China.
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China.
| | - Wentao Hu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Key Laboratory of Radiation Damage and Treatment of Jiangsu Provincial Universities and Colleges, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China.
| |
Collapse
|
2
|
Wang K, Zhao J, Wang Y, Liu M. Exercise benefits yourself and your offspring: a mini-review. Front Cell Dev Biol 2025; 13:1606790. [PMID: 40519261 PMCID: PMC12162635 DOI: 10.3389/fcell.2025.1606790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Accepted: 05/22/2025] [Indexed: 06/18/2025] Open
Abstract
Regular physical activity is widely recognized for its systemic health benefits, extending beyond physical fitness to influence metabolism, immunity, and neurophysiology. Pregnancy is a physiologically unique period characterized by dynamic immunometabolic changes that are crucial for maternal and fetal health. Maternal exercise during this window offers a non-pharmacological strategy to enhance maternal wellbeing and optimize offspring development. This review summarizes recent advances in understanding the effects of maternal exercise on both pregnant women and their offspring. In mothers, exercise improves metabolic profiles, modulates inflammatory responses, supports neuroplasticity, and promotes skeletal health. In offspring, maternal exercise confers long-term benefits including improved glucose metabolism, enhanced neurogenesis, cognitive development, and immune resilience. Mechanistically, these effects are mediated through molecular pathways such as placental superoxide dismutase 3 (SOD3) upregulation, adenosine 5'-monophosphate-activated protein kinase/ten-eleven translocation (AMPK/TET) signaling in the fetal liver, and exercise-induced circulating factors like Apelin and SERPINA3C, which contribute to epigenetic remodeling and tissue-specific programming. Despite growing evidence, gaps remain in understanding the optimal intensity, timing, and molecular mediators of maternal exercise, particularly regarding long-term immune and neurodevelopmental outcomes in offspring. Future studies leveraging multi-omics approaches are needed to elucidate cross-organ signaling mechanisms and identify therapeutic targets to mimic exercise-induced benefits. Overall, maternal exercise emerges as a safe, accessible intervention with significant potential to improve maternal-fetal health and reduce offspring disease risk across the lifespan.
Collapse
Affiliation(s)
- Kun Wang
- Faculty of Physical Education, China West Normal University, Nanchong, China
| | - Jiajia Zhao
- School of Physical Education, Central China Normal University, Wuhan, China
| | - Yanqiu Wang
- School of Physical Education, Central China Normal University, Wuhan, China
| | - Mairu Liu
- School of Physical Education, Central China Normal University, Wuhan, China
| |
Collapse
|
3
|
Liu J, Meng Z, Song J, Yu J, Guo Q, Zhang J, Wang S, Wang Y, Qiu Z, Zhang X, He J, Wang W. Yoda1-Loaded Microfibrous Scaffolds Accelerate Osteogenesis through Piezo1-F-Actin Pathway-Mediated YAP Nuclear Localization and Functionalization. ACS APPLIED MATERIALS & INTERFACES 2025; 17:30559-30572. [PMID: 40377908 PMCID: PMC12123560 DOI: 10.1021/acsami.5c03093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/30/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025]
Abstract
Yoda1 has been recognized as an effective pharmacological intervention for the treatment of critical bone defects. However, the local delivery strategy of Yoda1 is uncommon, and the underlying mechanism through which Yoda1 enhances osteogenesis has been poorly investigated. Here, we propose utilizing electrohydrodynamic (EHD)-printed microfibrous scaffolds as a drug carrier for loading Yoda1 through a polydopamine (PDA) coating, and the synthetic mechanisms for enhancing bone regeneration are explored. Yoda1 was successfully loaded on the surface of the EHD-printed microfibrous scaffolds with the assistance of PDA. The results of in vitro experiments demonstrated that the Yoda1-loaded microfibrous scaffold group exhibited a more than 2-fold increase in COL-I protein levels compared to the control group. Additionally, the expression levels of osteogenic indicators such as ALP, Runx2, and OCN genes were significantly increased by 2-4-fold compared to those in the control group. We revealed that Yoda1 can effectively activate the Piezo1-F-actin pathway, thereby facilitating YAP nucleation and promoting lysine histone acetylation. Consequently, this mechanism enhanced the functionality of YAP nucleation and upregulated the expression of COL-I. Moreover, when implanted in vivo, the Yoda1-loaded microfibrous scaffold group could promote macrophage M2 polarization, thereby enhancing bone regeneration at defect sites. It is believed that the localized release of Yoda1 via EHD-printed PCL scaffolds might represent a promising strategy for the clinically precise treatment of bone defects.
Collapse
Affiliation(s)
- Junzheng Liu
- Comprehensive
Orthopedics Department, the Second Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi710004, P. R. China
- State
Key
Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an710049, P. R. China
| | - Zijie Meng
- Frontier
Institute of Science and Technology, Xi’an
Jiaotong University, Xi’an710049, P. R. China
- State
Key
Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an710049, P. R. China
- National
Medical Products Administration (NMPA) Key Laboratory for Research
and Evaluation of Additive Manufacturing Medical Devices, Xi’an Jiaotong University, Xi’an710049, P. R. China
- State Industry-Education
Integration Center for Medical Innovations, Xi’an Jiaotong University, Xi’an710049, P. R. China
| | - Jidong Song
- Comprehensive
Orthopedics Department, the Second Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi710004, P. R. China
| | - Jiaming Yu
- Shaanxi
University of Chinese Medicine, Xianyang, Shaanxi712046, P. R. China
| | - Qin Guo
- Comprehensive
Orthopedics Department, the Second Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi710004, P. R. China
| | - Jiahao Zhang
- Comprehensive
Orthopedics Department, the Second Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi710004, P. R. China
| | - Shuo Wang
- Shaanxi
University of Chinese Medicine, Xianyang, Shaanxi712046, P. R. China
| | - Yulin Wang
- Comprehensive
Orthopedics Department, the Second Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi710004, P. R. China
| | - Zhennan Qiu
- State
Key
Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an710049, P. R. China
- National
Medical Products Administration (NMPA) Key Laboratory for Research
and Evaluation of Additive Manufacturing Medical Devices, Xi’an Jiaotong University, Xi’an710049, P. R. China
- State Industry-Education
Integration Center for Medical Innovations, Xi’an Jiaotong University, Xi’an710049, P. R. China
| | - Xinyi Zhang
- Comprehensive
Orthopedics Department, the Second Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi710004, P. R. China
| | - Jiankang He
- State
Key
Laboratory for Manufacturing Systems Engineering, Xi’an Jiaotong University, Xi’an710049, P. R. China
- National
Medical Products Administration (NMPA) Key Laboratory for Research
and Evaluation of Additive Manufacturing Medical Devices, Xi’an Jiaotong University, Xi’an710049, P. R. China
- State Industry-Education
Integration Center for Medical Innovations, Xi’an Jiaotong University, Xi’an710049, P. R. China
| | - Wei Wang
- Comprehensive
Orthopedics Department, the Second Affiliated
Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi710004, P. R. China
| |
Collapse
|
4
|
Ely EV, Lenz KL, Paradi SG, Ack S, Behrmann A, Dunivan S, Braxton L, Liedtke W, Chen Y, Collins KH, Guilak F. Chondrocyte-Specific Knockout of Piezo1 and Piezo2 Protects Against Post-Traumatic Osteoarthritis Structural Damage and Pain in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.22.655585. [PMID: 40501713 PMCID: PMC12154791 DOI: 10.1101/2025.05.22.655585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/22/2025]
Abstract
Background Osteoarthritis (OA) is a debilitating joint disease characterized by cartilage degeneration, synovial inflammation, and bone remodeling, with limited therapeutic options targeting the underlying pathophysiology. Mechanosensitive ion channels Piezo1 and Piezo2 play crucial roles in chondrocyte responses to mechanical stress, mediating mechanotransduction pathways that influence chondrocyte survival, matrix production, and inflammatory signaling, but their distinct contributions to OA pathogenesis remain unclear. Methods Using inducible, chondrocyte-specific Aggrecan-Cre ( Acan ) mice, we investigated Piezo1 , Piezo2 , and combined Piezo1 / 2 conditional knockouts (cKOs) using the destabilization of the medial meniscus (DMM) model of post-traumatic OA in male and female mice. Pain and behavioral assessments were conducted at four time points to evaluate OA progression, while cartilage damage, bone remodeling, and synovial inflammation were assessed at the final endpoint of 28 weeks. Statistical analyses included one-way and two-way ANOVA with Tukey's multiple comparisons test. Results Piezo1 cKO delayed pain onset but ultimately exacerbated cartilage degradation and synovitis, emphasizing its dual role in protective and pathogenic mechanotransduction. While the Piezo2 cKO reduced pain and preserved activity, it failed to protect cartilage. Notably, Piezo1/2 cKO provided the greatest protection against cartilage degeneration, synovitis, and pain. Micro-computed tomography analyses revealed that Piezo2 is critical for maintaining trabecular bone integrity, with a Piezo2 cKO leading to decreased bone volume, thickness, and density, independent of injury. Piezo2 cKO also reduced normal meniscal ossification that occurs with age in mice. In contrast, a Piezo1/2 cKO normalized most bone remodeling parameters observed in Piezo2 cKO mice but did not restore medial tibial plateau thickness, highlighting Piezo2 's essential role in bone structure. Conclusions These findings demonstrate the overlapping and compensatory roles of Piezo1 and Piezo2 in OA pathogenesis. Dual inhibition of Piezo1 and Piezo2 may offer a novel, effective therapeutic strategy targeting both structural and symptomatic aspects of the disease.
Collapse
Affiliation(s)
- Erica V. Ely
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Kristin L. Lenz
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Sophie G. Paradi
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri
| | - Seth Ack
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri
| | - Abraham Behrmann
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Sarah Dunivan
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
| | - Lauryn Braxton
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| | - Wolfgang Liedtke
- Department of Neurology, Duke University, Durham, North Carolina
| | - Yong Chen
- Department of Neurology, Duke University, Durham, North Carolina
| | - Kelsey H. Collins
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in Saint Louis, Saint Louis, Missouri
- Shriners Hospitals for Children–St. Louis, St. Louis, Missouri
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, Missouri
- Center of Regenerative Medicine, Washington University in Saint Louis, Saint Louis, Missouri
| |
Collapse
|
5
|
Meslier QA, Oehrlein R, Shefelbine SJ. Combined Effects of Mechanical Loading and Piezo1 Chemical Activation on 22-Months-Old Female Mouse Bone Adaptation. Aging Cell 2025:e70087. [PMID: 40410950 DOI: 10.1111/acel.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/23/2025] [Accepted: 04/11/2025] [Indexed: 05/26/2025] Open
Abstract
With age, bones mechanosensitivity is reduced, which limits their ability to adapt to loading. The exact mechanism leading to this loss of mechanosensitvity is still unclear, making developing effective treatment challenging. Current treatments mostly focus on preventing bone mass loss (such as bisphosphonates) or promoting bone formation (such as Sclerostin inhibitors) to limit the decline of bones mass. However, treatments do not target the cause of bone mass loss which may be, in part, due to the bone's inability to initiate a normal bone mechanoadaptation response. In this work, we investigated the effects of 2 weeks of tibia loading, and Piezo1 agonist injection in vivo on 22-month-old mouse bone adaptation response. We used an optimized loading profile, which induced high fluid flow velocity and low strain magnitude in adult mouse tibia. We found that tibia loading and Yoda2 injection have an additive effect on increasing cortical bone parameters in 22-month-old mice. In vivo osteocytes calcium signaling imaging suggests that Yoda2 is able to reach osteocytes and activate Piezo1. This combination of mechanical and chemical stimulation could be a promising treatment strategy to help promote bone formation in patients who have low bone mass due to aging.
Collapse
Affiliation(s)
- Quentin A Meslier
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Robert Oehrlein
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
| | - Sandra J Shefelbine
- Department of Bioengineering, Northeastern University, Boston, Massachusetts, USA
- Institute for Chemical Imaging of Living Systems, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Lai J, Wu Q, Gao B, Cai W, Wang Y. Piezo Channels in Dentistry: Decoding the Functional Effects of Forces. J Dent Res 2025:220345251329376. [PMID: 40353513 DOI: 10.1177/00220345251329376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
The oral system is a highly complex mechanosensory structure that continuously adapts to changes in mechanical stimuli, exerting mechanical forces on cells and tissues. Understanding how these forces are converted into biochemical signals and how they mediate gene expression and cellular activities has been a significant focus in dentistry. Piezo channels, including Piezo1 and Piezo2, are mechanically activated cation channels characterized by an extracellular "cap" domain and 3 peripheral mechanosensitive blades. Recent research has demonstrated that mechanical forces applied to tissues can induce deformation of cell membranes, leading to conformational changes in Piezo channels that facilitate cation influx, thereby regulating cellular activities. The influx of Ca2+, the most discussed outcome of Piezo channel activation, initiates diverse signaling pathways that regulate dentin hypersensitivity, alveolar bone remodeling, and temporomandibular joint (TMJ) osteoarthritis. The chemical inhibition of Piezo channels has been shown to alleviate dentinal hypersensitivity, reduce the rate of orthodontic tooth movement, and slow the progression of TMJ osteoarthritis in rat models. Mice deficient in piezo channels exhibit impaired reactive dentin formation, reduced alveolar bone volume, and developmental deformities of the jawbone. Considering their roles in decoding the functional effects of mechanical forces, this review summarizes the involvement of Piezo channels in dentistry, organized by anatomical sites, to provide comprehensive knowledge of Piezo channels and their mediated signal crosstalk, which offers promising therapeutic prospects for the treatment of various force-related oral diseases.
Collapse
Affiliation(s)
- J Lai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Q Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, China
| | - B Gao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, China
| | - W Cai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Y Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Wang X, Wang J, Zhang Y, He Y, Chen S. Piezo1 regulates fibrocartilage stem cell in cartilage growth and osteoarthritis. Osteoarthritis Cartilage 2025:S1063-4584(25)01003-9. [PMID: 40345612 DOI: 10.1016/j.joca.2025.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 04/05/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
OBJECTIVE The temporomandibular joint (TMJ) is essential for maxillofacial mechanics, with the condyle serving as the primary site for mandibular growth and development. Mechanical forces regulate condylar development and homeostasis, while aberrant loading contributes to temporomandibular joint osteoarthritis (TMJOA). Although mechanical stimuli are known to influence TMJ function, the role of mechanosensitive ion channels, particularly Piezo1, remains underexplored. METHODS To investigate the role of Piezo1, we performed single-cell ribonucleic acid sequencing (scRNA-seq) on human TMJ samples to assess mechanosensitive ion channel expression. Conditional knockout (CKO) mice were generated using Gli1-CreERT2; Piezo1fl/fl mice to delete Piezo1 in Gli1+ cells. Unilateral partial discectomy was performed to model TMJOA, followed by histological and immunohistochemical analyses to evaluate the effects of Piezo1 deletion on condylar development and homeostasis. RESULTS scRNA-seq revealed high expression of PIEZO1 in both cartilage and bone of the human condyle. Piezo1-tdTomato reporter mice confirmed its consistent expression across different ages. CKO of Piezo1 in Gli1+ cells impaired condylar development and homeostasis by disrupting fibrocartilage stem cell (FCSC) differentiation. In the TMJOA model, Piezo1 deletion mitigated arthritic symptoms, preserved condylar morphology, and reduced cartilage damage. CONCLUSION Piezo1 is a key mechanosensitive ion channel in the cartilage, regulating condylar development and homeostasis. Its deletion in Gli1+ cells disrupts FCSC differentiation and endochondral ossification, leading to developmental deficiencies. However, in pathological conditions, Piezo1 deletion protects against TMJOA, highlighting its potential as a therapeutic target for osteoarthritis prevention and treatment.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Jie Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Yi Zhang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Yang He
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Shuo Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| |
Collapse
|
8
|
Zhang FR, Tang J, Lai Y, Mo SQ, Lin ZM, Lei QQ, Han CC, Zhou AD, Lv XF, Wang C, Ou JS, Zhou JG, Pang RP. Smooth muscle cell Piezo1 is essential for phenotypic switch and neointimal hyperplasia. Br J Pharmacol 2025; 182:2031-2048. [PMID: 39900041 DOI: 10.1111/bph.17436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 10/17/2024] [Accepted: 11/10/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND AND PURPOSE Disturbed blood flow is a critical factor in activation of vascular smooth muscle cells (VSMCs) and initiation of neointimal hyperplasia. The Piezo1 channel is a recent yet well-characterised mechanosensor that plays a vital role in vascular development and homeostasis. However, the role of VSMC Piezo1 in neointima development remains largely unknown. The purpose of this study is to investigate the functional role of Piezo1 channel in neointimal hyperplasia. EXPERIMENTAL APPROACH We measured the expression of Piezo1 in VSMC-rich neointima from human coronary artery samples and two mouse neointimal hyperplasia models which were induced by cast implantation or guidewire injury. We utilised VSMC-specific Piezo1 knockout mice to explore its function and the underlying mechanism of neointimal hyperplasia, both in vivo and in vitro. KEY RESULTS In human and mouse neointima samples, we observed a significant up-regulation of Piezo1 expression in the VSMC-rich neointima compared to the medial layer. VSMC-specific knockout of Piezo1 significantly reduced neointimal hyperplasia in both animal models. Activation of Piezo1 facilitates, whereas Piezo1 deficiency inhibits disturbed flow-induced cell proliferation, migration and synthetic phenotype switch. Mechanistic studies suggest that Piezo1 activates YAP and TAZ through Ca2+ and its downstream effectors calmodulin kinase II and calcineurin, which in turn drive VSMC proliferation and migration, thereby facilitating neointimal hyperplasia. CONCLUSIONS AND IMPLICATIONS These findings demonstrate a critical role of mechanosensitive Piezo1 channel in neointimal hyperplasia via modulating VSMC phenotype. Piezo1 channels may represent a novel therapeutic target for maladaptive vascular remodelling and occlusive vascular diseases.
Collapse
Affiliation(s)
- Fei-Ran Zhang
- Program of Kidney and Cardiovascular Diseases, the Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jie Tang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ying Lai
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shi-Qi Mo
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Zhuo-Miao Lin
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qing-Qing Lei
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Cong-Cong Han
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - An-Dong Zhou
- Department of Clinical Medicine, the Second Clinical Medical School, Guangdong Medical University, Dongguan, China
| | - Xiao-Fei Lv
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Cheng Wang
- Program of Kidney and Cardiovascular Diseases, the Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing-Song Ou
- Division of Cardiac Surgery, Heart Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jia-Guo Zhou
- Program of Kidney and Cardiovascular Diseases, the Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Program of Vascular Diseases, the Eighth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Cardiology, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Cardiovascular diseases, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Rui-Ping Pang
- Department of Physiology, Pain Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Wang L, Ren Q, Chen S, Lou L, Hu X, Xing W, Suo J, Sun J, Greenblatt MB, Feng H, Zou W. Piezo1 balances the osteogenic-tenogenic plasticity of periosteal progenitor cells through the YAP pathway. Cell Rep 2025; 44:115630. [PMID: 40286268 DOI: 10.1016/j.celrep.2025.115630] [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: 01/22/2024] [Revised: 01/12/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
The extremity tendons and bones are derived from limb bud mesenchyme, which eventually becomes cells of different tissues. Despite their common origin, it was widely believed that tenocytes and skeletal cells are distinct lineages without interconversion. However, we found that periosteal skeletal progenitor cells could transform between osteogenic and tenogenic commitment. SCX can label the periosteal progenitor cell population. Loss of Piezo1 arrests the periosteal progenitor cells in a progenitor state. Piezo1 deficiency leads to upregulation of Scx expression due to the inhibited YAP nuclear localization. Loss of Piezo1 showed increased tenogenic marker genes and decreased osteogenic marker genes. The periosteal progenitor cells can regenerate the injured tendon more robustly when Piezo1 is absent. Taken together, our data reveal that periosteal SCX+ progenitor cells compromise the osteogenic ability and acquire the tenocyte-biased lineage commitment after loss of Piezo1, providing a strategy to modulate the periosteal progenitor cells for tissue regeneration.
Collapse
Affiliation(s)
- Lijun Wang
- Hainan Institute of Regenerative Orthopedics and Sports Medicine, Hainan Academy of Medical Sciences and School of Basic Medicine, Hainan Medical University, Hainan 570000, China; Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| | - Qian Ren
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuqin Chen
- Hainan Institute of Regenerative Orthopedics and Sports Medicine, Hainan Academy of Medical Sciences and School of Basic Medicine, Hainan Medical University, Hainan 570000, China
| | - Lixiang Lou
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xuye Hu
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenhui Xing
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Jun Sun
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Heng Feng
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Weiguo Zou
- Hainan Institute of Regenerative Orthopedics and Sports Medicine, Hainan Academy of Medical Sciences and School of Basic Medicine, Hainan Medical University, Hainan 570000, China; Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Institute of Microsurgery on Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China.
| |
Collapse
|
10
|
Liu X, Zhang Z, Cao Z, Yuan H, Xing C. Near-Infrared Light-Controlled Dynamic Hydrogel for Modulating Mechanosensitive Ion Channels in 3-Dimensional Environment. Biomater Res 2025; 29:0182. [PMID: 40207256 PMCID: PMC11979339 DOI: 10.34133/bmr.0182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/26/2025] [Accepted: 03/15/2025] [Indexed: 04/11/2025] Open
Abstract
The extracellular matrix (ECM) creates a dynamic mechanical environment for cellular functions, continuously influencing cellular activities via the mechanotransduction pathway. Mechanosensitive ion channels, recently identified as key mechanotransducers, convert mechanical stimuli into electrical or chemical signals when they detect membrane deformation. This process facilitates extracellular Ca2+ influx, cytoskeletal reorganization, and transcriptional regulation, all of which are essential for cellular physiological functions. In this study, we developed a fibrous hydrogel composite (PIC/OEG-NPs) with near-infrared (NIR) light-controlled dynamic mechanical properties to modulate mechanosensitive ion channels in cells, by using oligo-ethylene glycol (OEG)-assembled polyisocyanide (PIC) polymer and OEG-grafted conjugated polymer nanoparticles (OEG-NPs). PIC and OEG-NPs assemble into PIC/OEG-NPs composites through OEG-mediated hydrophobic interactions when heated. Under NIR stimulation, the PIC/OEG-NPs composites exhibit increased mechanical tension and form tighter fibrous networks due to their thermoresponsive behavior. These changes are reversible and allow for the dynamic regulation of mechanosensitive ion channels, including Piezo1 in transfected HEK-293T cells and the endogenous TRPV4 in human umbilical vein endothelial cells (HUVECs), by switching NIR on and off. Furthermore, this process enhances the angiogenic potential of HUVECs. In summary, we present a simple and effective platform for in situ modulation of mechanosensitive ion channels in 3 dimensions.
Collapse
Affiliation(s)
- Xiaoning Liu
- School of Materials Science and Engineering,
Hebei University of Technology, Tianjin 300401, China
| | - Zimeng Zhang
- Key Laboratory of Molecular Biophysics of Hebei Province, School of Health Sciences and Biomedical Engineering,
Hebei University of Technology, Tianjin 300401, China
| | - Zhanshuo Cao
- School of Chemical Engineering,
Hebei University of Technology, Tianjin 300401, China.
| | - Hongbo Yuan
- Key Laboratory of Molecular Biophysics of Hebei Province, School of Health Sciences and Biomedical Engineering,
Hebei University of Technology, Tianjin 300401, China
- Molecular Imaging and Photonics, Chemistry Department, KU Leuven, 3001 Heverlee, Belgium
| | - Chengfen Xing
- School of Materials Science and Engineering,
Hebei University of Technology, Tianjin 300401, China
- Key Laboratory of Molecular Biophysics of Hebei Province, School of Health Sciences and Biomedical Engineering,
Hebei University of Technology, Tianjin 300401, China
| |
Collapse
|
11
|
Wang Y, Lee B, Yang Z, Ho T, Ci H, Jackson B, Pushon T, Wang B, Levy J, Ho S. Chewing-Activated TRPV4/PIEZO1- HIF-1α-Zn Axes in a Rat Periodontal Complex. J Dent Res 2025; 104:398-407. [PMID: 39876056 PMCID: PMC11909774 DOI: 10.1177/00220345241294001] [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] [Indexed: 01/30/2025] Open
Abstract
The upstream mechanobiological pathways that regulate the downstream mineralization rates in periodontal tissues are limitedly understood. Herein, we spatially colocalized and correlated compression and tension strain profiles with the expressions of mechanosensory ion channels (MS-ion) TRPV4 and PIEZO1, biometal zinc, mitochondrial function marker (MFN2), cell senescence indicator (p16), and oxygen status marker hypoxia-inducible factor-1α (HIF-1α) in rats fed hard and soft foods. The observed zinc and related cellular homeostasis in vivo were ascertained by TRPV4 and PIEZO1 agonists and antagonists on human periodontal ligament fibroblasts ex vivo. Four-week-old male Sprague-Dawley rats were fed hard (n = 3) or soft (n = 3) foods for 4 wk (in vivo). Significant changes in alveolar socket and root shapes with decreased periodontal ligament space and increased cementum volume fraction were observed in maxillae on reduced loads (soft food). Reduced loads impaired distally localized compression-stimulated PIEZO1 and mesially localized tension-stimulated TRPV4, decreased mitochondrial function (MFN2), and increased cell senescence in mesial and distal periodontal regions. The switch in HIF-1α from hard food-distal to soft food-mesial indicated a plausible effect of shear-regulated blood and oxygen flows in the periodontal complex. Blunting or activating TRPV4 or PIEZO1 MS-ion channels by channel-specific antagonists or agonists in human periodontal ligament fibroblast cultures (in vitro) indicated a positive correlation between zinc levels and zinc transporters but not with MS-ion channel expressions. The effects of reduced chewing loads in vivo were analogous to TRPV4 and PIEZO1 antagonists in vitro. Study results collectively illustrated that tension-induced TRPV4 and compression-induced PIEZO1 activations are necessary for cell metabolism. An increased hypoxic state with reduced functional loads can be a conducive environment for cementum growth. From a practical standpoint, dose rate-controlled loads can modulate tension and compression-specific MS-ion channel activation, cellular zinc, and HIF-1α transcription. These mechanobiochemical events indicate the plausible catalytic role of biometal zinc in mineralization, periodontal maintenance, and dentoalveolar joint function.
Collapse
Affiliation(s)
- Y. Wang
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - B.H. Lee
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - Z. Yang
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - T.J. Ho
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
| | - H. Ci
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- International Research Center for Computational Mechanics, Dalian University of Technology, Dalian, China
- Ningbo Institute of Dalian University of Technology, Ningbo, China
- DUT-BSU Joint Institute, Dalian University of Technology, Dalian, China
| | - B. Jackson
- Deparment of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - T. Pushon
- Deparment of Biological Sciences, Dartmouth College, Hanover, NH, USA
| | - B. Wang
- State Key Laboratory of Structural Analysis for Industrial Equipment, Department of Engineering Mechanics, Dalian University of Technology, Dalian, China
- International Research Center for Computational Mechanics, Dalian University of Technology, Dalian, China
- Ningbo Institute of Dalian University of Technology, Ningbo, China
- DUT-BSU Joint Institute, Dalian University of Technology, Dalian, China
| | - J. Levy
- Department of Pathology and Computational Biomedicine, Cedars Sinai, Los Angeles, CA, USA
| | - S.P. Ho
- Department of Preventive and Restorative Dental Sciences, School of Dentistry, University of California, San Francisco, CA, USA
- Department of Urology, School of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
12
|
Zhou Y, Li M, Lin S, Zhu Z, Zhuang Z, Cui S, Chen L, Zhang R, Wang X, Shen B, Chen C, Yang R. Mechanical sensing protein PIEZO1 controls osteoarthritis via glycolysis mediated mesenchymal stem cells-Th17 cells crosstalk. Cell Death Dis 2025; 16:231. [PMID: 40169556 PMCID: PMC11961634 DOI: 10.1038/s41419-025-07577-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 03/04/2025] [Accepted: 03/20/2025] [Indexed: 04/03/2025]
Abstract
Aberrant mechanical stimuli can cause tissue attrition and activate mechanosensitive intracellular signaling, impacting the progression of osteoarthritis (OA). However, the precise relationship between mechanical loading and bone metabolism remains unclear. Here, we present evidence that Piezo1 senses the mechanical stimuli to coordinate the crosstalk between mesenchymal stem cells (MSCs) and T helper 17 (Th17) cells, leading to the deterioration of bone and cartilage in osteoarthritis (OA). Mechanical loading impaired the property of MSCs by inhibiting their osteo-chondrogenic differentiation and promoting inflammatory signaling to enhance Th17 cells. Mechanistically, mechanical stimuli activated Piezo1, thereby facilitating Ca2+ influx which upregulated the activity of Hexokinase 2(HK2), the rate-limiting enzyme of glycolysis. The resultant increase in glycolytic activity enhanced communication between MSCs and T cells, thus promoting Th17 cell polarization in a macrophage migration inhibitory factor (MIF) dependent manner. Functionally, Wnt1cre; Piezo1fl/fl mice reduced bone and cartilage erosion in the temporomandibular joint condyle following mechanical loading compared to control groups. Additionally, we observed activated Piezo1 and HK2-mediated glycolysis in patients with temporomandibular joint OA, thereby confirming the clinical relevance of our findings. Overall, our results provide insights into how Piezo1 in MSCs coordinates with mechano-inflammatory signaling to regulate bone metabolism. The schema shows that mechanical sensing protein PIEZO1 in MSCs controls osteoarthritis via glycolysis mediated MSCs and Th17 cells crosstalk in a MIF dependent manner.
Collapse
Affiliation(s)
- Yikun Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for 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 Beijing, Beijing, China
- Department of Orthodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Mingzhao Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for 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 Beijing, Beijing, China
| | - Shuai Lin
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for 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 Beijing, Beijing, China
| | - Zilu Zhu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for 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 Beijing, Beijing, China
| | - Zimeng Zhuang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for 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 Beijing, Beijing, China
| | - Shengjie Cui
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for 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 Beijing, Beijing, China
| | - Liujing Chen
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for 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 Beijing, Beijing, China
| | - Ran Zhang
- Department of Oral Pathology, Peking University School and Hospital of Stomatology, Beijing, China
| | - Xuedong Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for 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 Beijing, Beijing, China
| | - Bo Shen
- National Institute of Biological Sciences, Beijing, China, Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| | - Chider Chen
- Department of Oral and Maxillofacial Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Ruili Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Center for 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 Beijing, Beijing, China.
| |
Collapse
|
13
|
Wada H, Abe M, Wada N, Yoshimoto S, Fujii S, Moriyama M, Mori Y, Kido MA, Kiyoshima T. Piezo1 Is Related to the Enamel Matrix Formation in Mouse Tooth Germ Development. J Cell Physiol 2025; 240:e70036. [PMID: 40237554 DOI: 10.1002/jcp.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 03/23/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025]
Abstract
Cellular responses to mechanical stimulation are involved in tissue development and the maintenance of biological functions. Teeth function as receptors for mastication and occlusal pressure. During tooth development, the tooth germ begins with an invagination of the epithelium, and its morphology matures through dynamic interactions between epithelial cells and mesenchymal cells, suggesting that mechanosensors may play an important role in this process. We analyzed the expression and function of Piezo1, a mechanically activated ion channel, during tooth development and clarified the involvement of Piezo1 in tooth morphogenesis. The expression of Piezo1 was observed in both the enamel organ and the surrounding mesenchymal cells at the early stage and in the ameloblasts and odontoblasts during enamel and dentin matrix formation. Yoda1, a Piezo1 activator, inhibited cell proliferation in mouse dental epithelial (mDE6) cells and E15 tooth germs, and suppressed cell migration in mDE6 cells. Meanwhile, GsMTx4, a Piezo1 inactivator, showed opposite results. Furthermore, in the organ culture of E15 tooth germs, the activation and inactivation of Piezo1 were found to affect the expression of ameloblast differentiation marker genes and control the arrangement of ameloblasts. Interestingly, the expression of E-cadherin was reduced in the cell membrane of ameloblasts at the cusp in the GsMTx4-treated tooth germs of organ culture, and enamel formation was significantly decreased. Yoda1-treated mDE6 cells showed upregulated E-cadherin expression, which was downregulated by calpain inhibitor. These findings suggest that Piezo1 may be involved in tooth morphogenesis during ameloblast development by playing an essential role in cell proliferation, migration, arrangement, differentiation, and mineralization.
Collapse
Affiliation(s)
- Hiroko Wada
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, Japan
| | - Misaki Abe
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Naohisa Wada
- Department of General Dentistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Shohei Yoshimoto
- Section of Pathology, Department of Morphological Biology, Division of Biomedical Sciences, Fukuoka Dental College, Fukuoka, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Shinsuke Fujii
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Masafumi Moriyama
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Yoshihide Mori
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
- Division of Research and Treatment for Oral and Maxillofacial Congenital Anomalies, School of Dentistry, Aichi Gakuin University, Nagoya, Japan
| | - Mizuho A Kido
- Department of Anatomy and Physiology, Faculty of Medicine, Saga University, Saga, Japan
| | - Tamotsu Kiyoshima
- Laboratory of Oral Pathology, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
14
|
Lin Z, Xu G, Lu X, Wang H, Lu F, Xia X, Song J, Jiang J, Ma X, Zou F. Piezo1 exacerbates inflammation-induced cartilaginous endplate degeneration by activating mitochondrial fission via the Ca 2+/CaMKII/Drp1 axis. Aging Cell 2025; 24:e14440. [PMID: 39610146 PMCID: PMC11984661 DOI: 10.1111/acel.14440] [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: 07/18/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
Mitochondrial homeostasis plays a crucial role in degenerative joint diseases, including cartilaginous endplate (CEP) degeneration. To date, research into mitochondrial dynamics in IVDD is at an early stage. Since Piezo1 is a novel Ca2+-permeable channel, we asked whether Piezo1 could modulate mitochondrial fission through Ca2+ signalling during CEP degeneration. In vitro and in vivo models of inflammation-induced CEP degeneration were established with lipopolysaccharide (LPS). We found increased expression of Piezo1 in degenerated CEP tissues and LPS-treated CEP cells. The Piezo1 activator Yoda1 exacerbated CEP cell senescence and apoptosis by triggering Ca2+ influx. Yoda1 also induced mitochondrial fragmentation and dysfunction. In contrast, the Piezo1 inhibitor GsMTx4 exerted cytoprotective effects in LPS-treated CEP cells. Additionally, the CaMKII inhibitor KN-93 reversed Yoda1-induced mitochondrial fission and restored mitochondrial function. Mechanistically, the phosphorylation and mitochondrial translocation of Drp1 were regulated by the Ca2+/CaMKII signalling. The Drp1 inhibitor Mdivi-1 suppressed mitochondrial fission, then reduced mitochondrial dysfunction and CEP cell death. Moreover, knockdown of Piezo1 by siRNA hindered CaMKII and Drp1 activation, facilitating the redistribution of mitochondrial Drp1 to the cytosol in LPS-treated CEP cells. Piezo1 silencing improved mitochondrial morphology and function, thereby rescuing CEP cell senescence and apoptosis under inflammatory conditions. Finally, subendplate injection of GsMTx4 or AAV-shPiezo1 alleviated CEP degeneration in a rat model. Thus, Piezo1 may exacerbate inflammation-induced CEP degeneration by triggering mitochondrial fission and dysfunction via the Ca2+/CaMKII/Drp1 axis.
Collapse
Affiliation(s)
- Zhidi Lin
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Guangyu Xu
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Xiao Lu
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Hongli Wang
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Feizhou Lu
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Xinlei Xia
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Jian Song
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Jianyuan Jiang
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Xiaosheng Ma
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| | - Fei Zou
- Department of Orthopedics, Huashan HospitalFudan UniversityShanghaiChina
| |
Collapse
|
15
|
Wang D, Silvani G, Schroeter L, Brynn R, Chou J, Poole K. The mechanosensitive channel ELKIN1 regulates cellular adaptations to simulated microgravity. NPJ Microgravity 2025; 11:10. [PMID: 40090965 PMCID: PMC11911437 DOI: 10.1038/s41526-025-00466-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
In conditions of microgravity the human body undergoes extensive alterations in physiological function. However, it has proven challenging to determine how these changes are mediated at the molecular and cellular level. Here, we investigated whether ELKIN1, a mechanically activated ion channel, regulates changes in cellular and molecular structures in conditions of simulated microgravity. Deletion of ELKIN1 inhibited the simulated microgravity-induced alterations of cellular structure and attachment. In addition, cells lacking ELKIN1 did not exhibit changes in focal adhesion structures and redistribution of the YAP1 transcription factor in response to simulated microgravity, consistent with wild type cells. Finally, melanoma cell invasion of a collagen gel, from organotypic spheroids, was reduced in simulated microgravity, in an ELKIN1 dependent manner. Thus, the force sensing molecule, ELKIN1, modulates the impact of microgravity at both the molecular and cellular levels, revealing one of the molecular mechanisms that underpins cellular adaptations to conditions of microgravity.
Collapse
Affiliation(s)
- Daphne Wang
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, 2052, Sydney, NSW, Australia
- Eye Genetics Research Unit, Children's Medical Research Institute, The Children's Hospital at Westmead, Save Sight Institute, University of Sydney, Sydney, NSW, Australia
| | - Giulia Silvani
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
- Laboratory for Advanced Biomaterials & Matrix Engineering, School of Chemistry and School of Materials Science and Engineering, University of New South Wales, 2052, Sydney, NSW, Australia
| | - Lioba Schroeter
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, 2052, Sydney, NSW, Australia
| | - Remi Brynn
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, 2052, Sydney, NSW, Australia
| | - Joshua Chou
- School of Biomedical Engineering, Faculty of Engineering and Information Technology, University of Technology Sydney, Ultimo, NSW, Australia
- EXPLOR Biologics, NSW, 2000, Sydney, Australia
| | - Kate Poole
- School of Biomedical Sciences, Faculty of Medicine & Health, University of New South Wales, 2052, Sydney, NSW, Australia.
| |
Collapse
|
16
|
Yan X, Fu S, Xie Y, Zhang C, Wu X. Piezo1-driven mechanotransduction as a key regulator of cartilage degradation in early osteoarthritis. BIOMOLECULES & BIOMEDICINE 2025; 25:905-913. [PMID: 39388709 PMCID: PMC11959384 DOI: 10.17305/bb.2024.11156] [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: 08/17/2024] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024]
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease characterized by pain and cartilage damage in its later stages, while early OA is marked by the loss of cartilage's mechanical function. Recent studies suggest that Piezo1, a mechanotransducer, may contribute to cartilage degradation under abnormal physical stress. This study investigates the mechanism by which Piezo1 mediates the loss of cartilage's mechanical properties. Using rat chondrocytes cultured in a 3D in vitro model, we found that fluid flow-induced physical stress activates constitutively expressed Piezo1, leading to increased catabolic activity and apoptosis, which, in turn, disrupts the matrix structure. Ex vivo cartilage experiments further demonstrated that the mechanical stress-induced loss of cartilage's physical properties (approximately 10% reduction in relaxation modulus) is mediated by Piezo1 and depends on cell viability. Notably, Piezo1 agonists alone did not alter the mechanical behavior of cartilage tissue. In vivo, using an OA rat model induced by anterior cruciate ligament transection, we observed cartilage integrity degradation and loss of mechanical properties, which were partially mitigated by Piezo1 inhibition. RNA sequencing revealed significant modulation of the PI3K signaling and matrix regulation pathways. Collectively, this study demonstrates that Piezo1-mediated catabolic activity in chondrocytes is a key driver of the loss of cartilage's mechanical function during the relaxation phase.
Collapse
Affiliation(s)
- Xu Yan
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Su Fu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Xie
- Department of Blood Transfusion, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chunlin Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuejian Wu
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
17
|
Chen X, Ji X, Lao Z, Pan B, Qian Y, Yang W. Role of YAP/TAZ in bone diseases: A transductor from mechanics to biology. J Orthop Translat 2025; 51:13-23. [PMID: 39902099 PMCID: PMC11787699 DOI: 10.1016/j.jot.2024.12.003] [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: 08/01/2024] [Revised: 10/24/2024] [Accepted: 12/09/2024] [Indexed: 02/05/2025] Open
Abstract
Wolff's Law and the Mechanostat Theory elucidate how bone tissues detect and convert mechanical stimuli into biological signals, crucial for maintaining bone equilibrium. Abnormal mechanics can lead to diseases such as osteoporosis, osteoarthritis, and nonunion fractures. However, the detailed molecular mechanisms by which mechanical cues are transformed into biological responses in bone remain underexplored. Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), key regulators of bone homeostasis, are instrumental in this process. Emerging research highlights bone cells' ability to sense various mechanical stimuli and relay these signals intracellularly. YAP/TAZ are central in receiving these mechanical cues and converting them into signals that influence bone cell behavior. Abnormal YAP/TAZ activity is linked to several bone pathologies, positioning these proteins as promising targets for new treatments. Thus, this review aims to provide an in-depth examination of YAP/TAZ's critical role in the interpretation of mechanical stimuli to biological signals, with a special emphasis on their involvement in bone cell mechanosensing, mechanotransduction, and mechanoresponse. The translational potential of this article: Clinically, appropriate stress stimulation promotes fracture healing, while bed rest can lead to disuse osteoporosis and excessive stress can cause osteoarthritis or bone spurs. Recent advancements in the understanding of YAP/TAZ-mediated mechanobiological signal transduction in bone diseases have been significant, yet many aspects remain unknown. This systematic review summarizes current research progress, identifies unaddressed areas, and highlights potential future research directions. Advancements in this field facilitate a deeper understanding of the molecular mechanisms underlying bone mechanics regulation and underscore the potential of YAP/TAZ as therapeutic targets for bone diseases such as fractures, osteoporosis, and osteoarthritis.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Xing Ji
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhaobai Lao
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Bin Pan
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Yu Qian
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| | - Wanlei Yang
- Department of Orthopedics Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310006, China
| |
Collapse
|
18
|
Zhao Y, Duan J, Han ID, van de Leemput J, Ray PE, Han Z. Piezo, Nephrocyte Function, and Slit Diaphragm Maintenance in Drosophila. J Am Soc Nephrol 2025; 36:393-405. [PMID: 39431457 PMCID: PMC11888958 DOI: 10.1681/asn.0000000529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024] Open
Abstract
Key Points Piezo channels, known for detecting mechanical pressure, were found to be expressed at the lacuna channel membranes of nephrocytes. Piezo loss of function caused nephrocyte dysfunction, including disrupted slit diaphragm structure and altered lacuna channel morphology. Piezo deficiency led to internalized slit diaphragm proteins, reduced autophagy, increased endoplasmic reticulum stress, and impaired calcium homeostasis. Background The Piezo gene encodes a highly conserved cell membrane protein responsible for sensing pressure. The glomerular kidney and the slit diaphragm filtration structure depend on pressure for filtration. However, how Piezo is involved in kidney function and in maintaining the slit diaphragm filtration structure is not clear. Methods We used Drosophila pericardial nephrocytes, filtration kidney cells with striking structural and functional similarities to human podocytes, in a loss-of-function model (mutant and knockdown) to study the roles of Piezo in nephrocyte filtration and function. Results Piezo was highly expressed at the invaginated membranes (lacuna channels) of nephrocytes. A Piezo loss-of-function mutant showed significant nephrocyte functional decline. Nephrocyte-specific silencing of Piezo showed disruption of the slit diaphragm filtration structure and significant functional defects. Electron microscopy showed that silencing Piezo in nephrocytes led to reduced slit diaphragm density and abnormal shape of lacuna channels. Moreover, the Piezo-deficient nephrocytes showed internalized slit diaphragm component proteins, reduced autophagy, increased endoplasmic reticulum stress, and reduced calcium influx. Conclusions Together, our findings suggest that Piezo plays an important role in the calcium homeostasis of nephrocytes and is required for maintaining nephrocyte function and the slit diaphragm filtration structure.
Collapse
Affiliation(s)
- Yunpo Zhao
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jianli Duan
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Iden D. Han
- Department of Pediatrics, Child Health Research Center, School of Medicine, University of Virginia, Charlottesville, Virginia
| | - Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Patricio E. Ray
- Department of Pediatrics, Child Health Research Center, School of Medicine, University of Virginia, Charlottesville, Virginia
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
19
|
Liu T, Xiong B, Cui X, Zhang C. Numerical study of interstitial fluid flow behavior in osteons under dynamic loading. BMC Musculoskelet Disord 2025; 26:187. [PMID: 39994737 PMCID: PMC11854024 DOI: 10.1186/s12891-025-08425-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND The porous structure in bone tissue is essential for maintaining the physiological functions and overall health of intraosseous cells. The lacunar-canalicular net (LCN), a microscopic porous structure within osteons, facilitates the transport of nutrients and signaling molecules through interstitial fluid flow. However, the transient behavior of fluid flow within these micro-pores under dynamic loading conditions remains insufficiently studied. METHODS The study constructs a fluid-solid coupling model including the Haversian canal, canaliculi, lacunae, and interstitial fluid, to examine interstitial fluid flow behavior within the LCN under dynamic loading with varying frequencies and amplitudes. The relationship between changes of LCN pore volume and fluid velocity, and pressure is researched. RESULTS The results demonstrate that increasing strain amplitude leads to significant changes of LCN pore volume within osteons. In a complete loading cycle, with the increase of compressive strain, the pore volume in the osteon gradually shrinks, and the pressure gradient in the LCN increases, which promotes the increase of interstitial fluid velocity. When the compressive strain reaches the peak value, the flow velocity also reaches the maximum. In the subsequent unloading process, the pore volume began to recover, the pressure gradient gradually decreased, the flow rate decreased accordingly, and finally returned to the steady state level. At a loading amplitude of 1000 µε, the pore volume within LCN decreases by 1.1‰. At load amplitudes of 1500 µε, 2000 µε, and 2500 µε, the pore volume decreases by 1.6‰, 2.2‰ and 2.7‰ respectively, and the average flow velocity at the center of the superficial lacuna is 1.36 times, 1.77 times, and 2.14 times that at 1000 µε, respectively. Additionally, at a loading amplitude of 1000 µε under three different loading frequencies, the average flow velocities at the center of the superficial bone lacuna are 0.60 μm/s, 1.04 μm/s, and 1.54 μm/s, respectively. This indicates that high-frequency and high-amplitude dynamic loading can promote more vigorous fluid flow and pressure fluctuations with changes in LCN pore volume. CONCLUSIONS Dynamic mechanical loading can significantly enhance the interstitial fluid flow in LCN by the changes of LCN pore volume. and dynamic loading promoted fluid flow in shallow lacunae significantly higher than that in deep lacunae. The relationship between changes of LCN pore volume and interstitial fluid flow behavior has implications for drug delivery and bone tissue engineering research.
Collapse
Affiliation(s)
- Tianyu Liu
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, P.R. China
| | - Baochuan Xiong
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, P.R. China
| | - Xin Cui
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, P.R. China
| | - Chunqiu Zhang
- Tianjin Key Laboratory for Advanced Mechatronic System Design and Intelligent Control, School of Mechanical Engineering, Tianjin University of Technology, Tianjin, 300384, P.R. China.
| |
Collapse
|
20
|
Zhang S, Li C, Feng Y, Lei W, Sun X. Emerging roles of Piezo1 channels in bone: Cells and diseases. Chin Med J (Engl) 2025; 138:00029330-990000000-01419. [PMID: 39920897 PMCID: PMC11882270 DOI: 10.1097/cm9.0000000000003483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Indexed: 02/10/2025] Open
Affiliation(s)
- Siqi Zhang
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Chengfei Li
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yafei Feng
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Wei Lei
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Xiqing Sun
- Department of Aerospace Medical Training, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| |
Collapse
|
21
|
He Z, Du Y, Peng C, Xu B, Tang J, Liu R, Yang K. Piezo1 Promotes Osteogenesis Through CaMKII Signalling in a Rat Maxillary Expansion Model. Orthod Craniofac Res 2025; 28:196-206. [PMID: 39350680 DOI: 10.1111/ocr.12862] [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: 07/17/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 01/07/2025]
Abstract
OBJECTIVES Rapid maxillary expansion (RME) is a widely used technique to treat maxillary transverse deficiency. Piezo1 is a cation channel that is activated by mechanical force and regulates bone formation. This study aims to elucidate the role of Piezo1 in bone remodelling during the RME process. MATERIALS AND METHODS In this study, the periosteal-derived stem cells (PDSCs) were cultured and stretched by the Flexcell system. The effects of Piezo1 on osteogenesis were assessed via RNA sequencing, real-time quantitative PCR, and western blot analyses. Moreover, for in vivo analyses, the rat RME model was established. The function of Piezo1 in mid-palatal suture bone remodelling was evaluated using micro-CT, haematoxylin-eosin (HE) staining, and immunohistochemistry analyses. RESULTS It was revealed that under tension force, the osteogenic factors (Runt-related transcription factor 2, Osterix, and Alkaline Phosphatase) and Ca2+/calmodulin -dependent protein kinase (CaMKII) were significantly enhanced in PDSCs over time. Furthermore, these were also upregulated in the RME model with the expansion of the mid-palatal suture. However, Piezo1 inhibition by Grammostola spatulata mechanotoxin 4 downregulated the levels of these factors in the RME model. CONCLUSIONS This study indicated that Piezo1 is associated with the osteogenesis of PDSCs and bone remodelling in the RME process. CaMKII might also participate in this process.
Collapse
Affiliation(s)
- Zhengquan He
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
- Department of Orthodontics, Changsha Stomatological Hospital, Changsha, China
| | - Yugui Du
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Chuhan Peng
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Bowen Xu
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Jiayue Tang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Runxuan Liu
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| | - Kai Yang
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
22
|
Ly AT, Freites JA, Bertaccini GA, Evans EL, Dickinson GD, Tobias DJ, Pathak MM. Single-particle tracking reveals heterogeneous PIEZO1 diffusion. Biophys J 2025:S0006-3495(25)00023-2. [PMID: 39844465 DOI: 10.1016/j.bpj.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 12/21/2024] [Accepted: 01/16/2025] [Indexed: 01/24/2025] Open
Abstract
The mechanically activated ion channel PIEZO1 is critical to numerous physiological processes, and is activated by diverse mechanical cues. The channel is gated by membrane tension and has been found to be mobile in the plasma membrane. We employed single-particle tracking (SPT) of endogenous, tdTomato-tagged PIEZO1 using total internal reflection fluorescence microscopy in live cells. Application of SPT unveiled a surprising heterogeneity of diffusing PIEZO1 subpopulations, which we labeled "mobile" and "immobile." We sorted these trajectories into the two aforementioned categories using trajectory spread. To evaluate the effects of the plasma membrane composition on PIEZO1 diffusion, we manipulated membrane composition by depleting or supplementing cholesterol, or by adding margaric acid to stiffen the membrane. To examine effects of channel activation on PIEZO1 mobility, we treated cells with Yoda1, a PIEZO1 agonist, and GsMTx-4, a channel inhibitor. We collected thousands of trajectories for each condition, and found that cholesterol removal and Yoda1 incubation increased the channel's propensity for mobility. Conversely, we found that GsMTx-4 incubation and cholesterol supplementation resulted in a lower chance of mobile trajectories, whereas margaric acid incubation did not have a significant effect on PIEZO1 mobility. The mobile trajectories were analyzed further by fitting the time-averaged mean-squared displacement as a function of lag time to a power law model, revealing that mobile PIEZO1 puncta exhibit anomalous subdiffusion. These studies illuminate the fundamental properties governing PIEZO1 diffusion in the plasma membrane and set the stage to determine how cellular processes and interactions may influence channel activity and mobility.
Collapse
Affiliation(s)
- Alan T Ly
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | | | - Gabriella A Bertaccini
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | - Elizabeth L Evans
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | - George D Dickinson
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California
| | - Douglas J Tobias
- Department of Chemistry, UC Irvine, Irvine, California; Center for Complex Biological Systems, UC Irvine, Irvine, California.
| | - Medha M Pathak
- Department of Physiology & Biophysics, UC Irvine, Irvine, California; Department of Biomedical Engineering, UC Irvine, Irvine, California; Center for Complex Biological Systems, UC Irvine, Irvine, California; Sue and Bill Gross Stem Cell Research Center, UC Irvine, Irvine, California.
| |
Collapse
|
23
|
Yan C, Jin G, Li L. Spinal scoliosis: insights into developmental mechanisms and animal models. Spine Deform 2025; 13:7-18. [PMID: 39164474 PMCID: PMC11729078 DOI: 10.1007/s43390-024-00941-9] [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: 04/16/2024] [Accepted: 07/29/2024] [Indexed: 08/22/2024]
Abstract
Spinal scoliosis, a prevalent spinal deformity impacting both physical and mental well-being, has a significant genetic component, though the exact pathogenic mechanisms remain elusive. This review offers a comprehensive exploration of current research on embryonic spinal development, focusing on the genetic and biological intricacies governing axial elongation and straightening. Zebrafish, a vital model in developmental biology, takes a prominent role in understanding spinal scoliosis. Insights from zebrafish studies illustrate genetic and physiological aspects, including notochord development and cerebrospinal fluid dynamics, revealing the anomalies contributing to scoliosis. In this review, we acknowledge existing challenges, such as deciphering the unique dynamics of human spinal development, variations in physiological curvature, and disparities in cerebrospinal fluid circulation. Further, we emphasize the need for caution when extrapolating findings to humans and for future research to bridge current knowledge gaps. We hope that this review will be a beneficial frame of reference for the guidance of future studies on animal models and genetic research for spinal scoliosis.
Collapse
Affiliation(s)
- Chongnan Yan
- Department of Spine Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Guoxin Jin
- Department of Spine Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Lei Li
- Department of Spine Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
24
|
Randhawa A, Ganguly K, Dutta SD, Patil TV, Lim KT. Transcriptomic profiling of human mesenchymal stem cells using a pulsed electromagnetic-wave motion bioreactor system for enhanced osteogenic commitment and therapeutic potentials. Biomaterials 2025; 312:122713. [PMID: 39084096 DOI: 10.1016/j.biomaterials.2024.122713] [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: 02/13/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024]
Abstract
Traditional bioreactor systems involve the use of three-dimensional (3D) scaffolds or stem cell aggregates, limiting the accessibility to the production of cell-secreted biomolecules. Herein, we present the use a pulse electromagnetic fields (pEMFs)-assisted wave-motion bioreactor system for the dynamic and scalable culture of human bone marrow-derived mesenchymal stem cells (hBMSCs) with enhanced the secretion of various soluble factors with massive therapeutic potential. The present study investigated the influence of dynamic pEMF (D-pEMF) on the kinetic of hBMSCs. A 30-min exposure of pEMF (10V-1Hz, 5.82 G) with 35 oscillations per minute (OPM) rocking speed can induce the proliferation (1 × 105 → 4.5 × 105) of hBMSCs than static culture. Furthermore, the culture of hBMSCs in osteo-induction media revealed a greater enhancement of osteogenic transcription factors under the D-pEMF condition, suggesting that D-pEMF addition significantly boosted hBMSCs osteogenesis. Additionally, the RNA sequencing data revealed a significant shift in various osteogenic and signaling genes in the D-pEMF group, further suggesting their osteogenic capabilities. In this research, we demonstrated that the combined effect of wave and pEMF stimulation on hBMSCs allows rapid proliferation and induces osteogenic properties in the cells. Moreover, our study revealed that D-pEMF stimuli also induce ROS-scavenging properties in the cultured cells. This study also revealed a bioactive and cost-effective approach that enables the use of cells without using any expensive materials and avoids the possible risks associated with them post-implantation.
Collapse
Affiliation(s)
- Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea; Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea; Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
25
|
Ochiai N, Etani Y, Noguchi T, Miura T, Kurihara T, Fukuda Y, Hamada H, Uemura K, Takashima K, Tamaki M, Ishibashi T, Ito S, Yamakawa S, Kanamoto T, Okada S, Nakata K, Ebina K. The pivotal role of the Hes1/Piezo1 pathway in the pathophysiology of glucocorticoid-induced osteoporosis. JCI Insight 2024; 9:e179963. [PMID: 39641269 PMCID: PMC11623955 DOI: 10.1172/jci.insight.179963] [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: 02/21/2024] [Accepted: 10/22/2024] [Indexed: 12/07/2024] Open
Abstract
Glucocorticoid-induced osteoporosis (GIOP) lacks fully effective treatments. This study investigated the role of Piezo1, a mechanosensitive ion channel component 1, in GIOP. We found reduced Piezo1 expression in cortical bone osteocytes from patients with GIOP and a GIOP mouse model. Yoda1, a Piezo1 agonist, enhanced the mechanical stress response and bone mass and strength, which were diminished by dexamethasone (DEX) administration in GIOP mice. RNA-seq revealed that Yoda1 elevated Piezo1 expression by activating the key transcription factor Hes1, followed by enhanced CaM kinase II and Akt phosphorylation in osteocytes. This improved the lacuno-canalicular network and reduced sclerostin production and the receptor activator of NF-κB/osteoprotegerin ratio, which were mitigated by DEX. Comparative analysis of mouse models and human GIOP cortical bone revealed downregulation of mechanostimulated osteogenic factors, such as osteocrin, and cartilage differentiation markers in osteoprogenitor cells. In human periosteum-derived cells, DEX suppressed differentiation into osteoblasts, but Yoda1 rescued this effect. Our findings suggest that reduced Piezo1 expression and activity in osteocytes and periosteal cells contribute to GIOP, and Yoda1 may offer a novel therapeutic approach by restoring mechanosensitivity.
Collapse
Affiliation(s)
- Nagahiro Ochiai
- Department of Musculoskeletal Regenerative Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | - Yuki Etani
- Department of Musculoskeletal Regenerative Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | - Teruya Ishibashi
- Department of Orthopaedic Biomaterial Science, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | - Shohei Ito
- Taisho Pharmaceutical Co., Ltd., Saitama, Japan
| | | | - Takashi Kanamoto
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | | | - Ken Nakata
- Department of Health and Sport Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Kosuke Ebina
- Department of Musculoskeletal Regenerative Medicine, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
- Department of Orthopaedic Surgery
| |
Collapse
|
26
|
Xie D, Ma Y, Gao C, Pan S. Piezo1 activation on microglial cells exacerbates demyelination in sepsis by influencing the CCL25/GRP78 pathway. Int Immunopharmacol 2024; 142:113045. [PMID: 39236454 DOI: 10.1016/j.intimp.2024.113045] [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: 05/20/2024] [Revised: 08/27/2024] [Accepted: 08/27/2024] [Indexed: 09/07/2024]
Abstract
BACKGROUND In sepsis-associated encephalopathy (SAE), the activation of microglial cells and ensuing neuroinflammation are important in the underlying pathological mechanisms. Increasing evidence suggests that the protein Piezo1 functions as a significant regulator of neuroinflammation. However, the influence of Piezo1 on microglial cells in the context of SAE has not yet been determined. This study aims to investigate the role of Piezo1 in microglial cells in the context of SAE. METHODS By inducing cecal ligation and puncture (CLP), a mouse model of SAE was established, while the control group underwent a sham surgery in which the cecum was exposed without ligation and puncture. Piezo1 knockout mice were employed in this study. Morris water maze tests were conducted between Days 14 and 18 postop to assess both the motor activity and cognitive function. A proteomic analysis was conducted to assess the SAE-related pathways, whereas a Mendelian randomization analysis was conducted to identify the pathways associated with cognitive impairment. Dual-label immunofluorescence and flow cytometry were used to assess the secretion of inflammatory factors, microglial status, and oligodendrocyte development. Electron microscopy was used to evaluate axonal myelination. A western blot analysis was conducted to evaluate the influence of Piezo1 on oligodendrocyte ferroptosis. RESULTS The results of the bioinformatics analysis have revealed the significant involvement of CCL25 in the onset and progression of SAE-induced cognitive impairment. SAE leads to cognitive dysfunction by activating the microglial cells. The release of CCL25 by the activated microglia initiates the demyelination of oligodendrocytes in the hippocampus, resulting in ferroptosis and the disruption of hippocampal functional connectivity. Of note, the genetic knockout of the Piezo1 gene mitigates these changes. The treatment with siRNA targeting Piezo1 effectively reduces the secretion of inflammatory mediators CCL25 and IL-18 by inhibiting the p38 pathway, thus preventing the ferroptosis of oligodendrocytes through the modulation of the CCL25/GPR78 axis. CONCLUSION Piezo1 is involved in the activation of microglia and demyelinating oligodendrocytes in the animal models of SAE, resulting in cognitive impairment. Consequently, targeting Piezo1 suppression can be a promising approach for therapeutic interventions aimed at addressing cognitive dysfunction associated with SAE.
Collapse
Affiliation(s)
- Di Xie
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, 200092 Shanghai, China
| | - Yanli Ma
- Department of Pediatrics, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Hongkou District, 200434 Shanghai, China
| | - Chengjin Gao
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, 200092 Shanghai, China.
| | - Shuming Pan
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Yangpu District, 200092 Shanghai, China; Department of Emergency, Putuo District Central Hospital, Affiliated with Shanghai University of Traditional Chinese Medicine, Putuo District, 200062 Shanghai, China.
| |
Collapse
|
27
|
Jia S, Liu W, Zhang M, Wang L, Ren C, Feng C, Zhang T, Lv H, Hou Z, Zou W, Zhang Y, Tong W, Wang J, Chen W. Insufficient Mechanical Loading Downregulates Piezo1 in Chondrocytes and Impairs Fracture Healing Through ApoE-Induced Senescence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400502. [PMID: 39418070 PMCID: PMC11633519 DOI: 10.1002/advs.202400502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 08/10/2024] [Indexed: 10/19/2024]
Abstract
Insufficient mechanical loading impairs fracture healing; however, the underlying mechanisms remain unclear. Increasing evidence indicates that Piezo1 plays an important role in fracture healing, although the effect of Piezo1 on the endochondral ossification of chondrocytes has been overlooked. This study reports that mechanical unloading down-regulates the expression of Piezo1 in chondrocytes and leads to fracture nonunion. Single-cell sequencing of calluses revealed that specific deletion of Piezo1 in chondrocytes upregulated the expression of apolipoprotein E (ApoE) in hypertrophic chondrocytes, resulting in delayed cartilage-to-bone transition due to enhanced chondrocyte senescence. Based on these results, an injectable and thermosensitive hydrogel is developed, which released an ApoE antagonist in situ at the fracture site. This hydrogel effectively attenuated chondrocyte senescence and, thus, promoted cartilage-to-bone transition as well as the fracture healing process. Overall, this data provide a new perspective on the activity of chondrocytes in fracture healing and a new direction for the treatment of fracture nonunion caused by insufficient mechanical loading.
Collapse
Affiliation(s)
- Siming Jia
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
- Hebei Medical University Clinical Medicine Postdoctoral Station (Hebei Medical University Third Hospital)ShijiazhuangHebei050051China
| | - Weijian Liu
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430022China
| | - Mo Zhang
- Hebei Medical University Clinical Medicine Postdoctoral Station (Hebei Medical University Third Hospital)ShijiazhuangHebei050051China
- School of PharmacyKey Laboratory of Innovative Drug Development and EvaluationHebei Medical UniversityShijiazhuang050017China
| | - Lijun Wang
- Hainan Institute of Regenerative Orthopedics and Sports Medicine, Hainan Academy of Medical Sciences and School of Basic MedicineHainan Medical UniversityHainan570000China
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Chuan Ren
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Chen Feng
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Tao Zhang
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Hongzhi Lv
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Zhiyong Hou
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Weiguo Zou
- Hainan Institute of Regenerative Orthopedics and Sports Medicine, Hainan Academy of Medical Sciences and School of Basic MedicineHainan Medical UniversityHainan570000China
- Key Laboratory of RNA Innovation, Science and Engineering, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, University of Chinese Academy of SciencesChinese Academy of SciencesShanghai200031China
| | - Yingze Zhang
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Wei Tong
- Department of OrthopaedicsUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubei430022China
| | - Juan Wang
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| | - Wei Chen
- Department of Orthopaedic Surgery, NHC Key Laboratory of Intelligent Orthopaedic EquipmentHebei Medical University Third HospitalShijiazhuangHebei050051China
| |
Collapse
|
28
|
Li Z, Hao L, Chen S, Fu W, Zhang H, Yin Z, Wang Y, Wang J. Forkhead box C1 promotes the pathology of osteoarthritis in subchondral bone osteoblasts via the Piezo1/YAP axis. Cell Signal 2024; 124:111463. [PMID: 39396563 DOI: 10.1016/j.cellsig.2024.111463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/15/2024]
Abstract
Subchondral bone sclerosis is a key characteristic of osteoarthritis (OA). Prior research has shown that Forkhead box C1 (FoxC1) plays a role in the synovial inflammation of OA, but its specific role in the subchondral bone of OA has not been explored. Our research revealed elevated expression levels of FoxC1 and Piezo1 in OA subchondral bone tissues. Further experiments on OA subchondral bone osteoblasts with FoxC1 or Piezo1 overexpression showed increased cell proliferation activity, expression of Yes-associated Protein 1 (YAP) and osteogenic markers, and secretion of proinflammatory factors. Mechanistically, the overexpression of FoxC1 through Piezo1 activation, in combination with downstream YAP signaling, led to increased levels of alkaline phosphatase (ALP), collagen type 1 (COL1) A1, RUNX2, Osteocalcin, matrix metalloproteinase (MMP) 3, and MMP9 expression. Notably, inhibition of Piezo1 reversed the regulatory function of FoxC1. The binding of FoxC1 to the targeted area (ATATTTATTTA, residues +612 to +622) and the activation of Piezo1 transcription were verified by the dual luciferase assays. Additionally, Reduced subchondral osteosclerosis and microangiogenesis were observed in knee joints from FoxC1-conditional knockout (CKO) and Piezo1-CKO mice, indicating reduced lesions. Collectively, our study reveals the significant involvement of FoxC1 in the pathologic process of OA subchondral bone via the Piezo1/YAP signaling pathway, potentially establishing a novel therapeutic target.
Collapse
Affiliation(s)
- Zhengyuan Li
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Anhui, China
| | - Lin Hao
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Anhui, China
| | - Shenghong Chen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Anhui, China
| | - Wenhan Fu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China; Anhui Province Key Laboratory of Zoonoses, Anhui Medical University, Anhui, China
| | - Hui Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China
| | - Zongsheng Yin
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China.
| | - Yin Wang
- Department of Wound Repair & Plastic and Aesthetic Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui, China; Anhui Public Health Clinical Center, Anhui, China.
| | - Jun Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, 218 JiXi Road, Hefei 230022, Anhui, China.
| |
Collapse
|
29
|
Schwartzman JD, McCall M, Ghattas Y, Pugazhendhi AS, Wei F, Ngo C, Ruiz J, Seal S, Coathup MJ. Multifunctional scaffolds for bone repair following age-related biological decline: Promising prospects for smart biomaterial-driven technologies. Biomaterials 2024; 311:122683. [PMID: 38954959 DOI: 10.1016/j.biomaterials.2024.122683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/09/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
The repair of large bone defects due to trauma, disease, and infection can be exceptionally challenging in the elderly. Despite best clinical practice, bone regeneration within contemporary, surgically implanted synthetic scaffolds is often problematic, inconsistent, and insufficient where additional osteobiological support is required to restore bone. Emergent smart multifunctional biomaterials may drive important and dynamic cellular crosstalk that directly targets, signals, stimulates, and promotes an innate bone repair response following age-related biological decline and when in the presence of disease or infection. However, their role remains largely undetermined. By highlighting their mechanism/s and mode/s of action, this review spotlights smart technologies that favorably align in their conceivable ability to directly target and enhance bone repair and thus are highly promising for future discovery for use in the elderly. The four degrees of interactive scaffold smartness are presented, with a focus on bioactive, bioresponsive, and the yet-to-be-developed autonomous scaffold activity. Further, cell- and biomolecular-assisted approaches were excluded, allowing for contemporary examination of the capabilities, demands, vision, and future requisites of next-generation biomaterial-induced technologies only. Data strongly supports that smart scaffolds hold significant promise in the promotion of bone repair in patients with a reduced osteobiological response. Importantly, many techniques have yet to be tested in preclinical models of aging. Thus, greater clarity on their proficiency to counteract the many unresolved challenges within the scope of aging bone is highly warranted and is arguably the next frontier in the field. This review demonstrates that the use of multifunctional smart synthetic scaffolds with an engineered strategy to circumvent the biological insufficiencies associated with aging bone is a viable route for achieving next-generation therapeutic success in the elderly population.
Collapse
Affiliation(s)
| | - Max McCall
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Yasmine Ghattas
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Abinaya Sindu Pugazhendhi
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Fei Wei
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Christopher Ngo
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA
| | - Jonathan Ruiz
- College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Sudipta Seal
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA; Advanced Materials Processing and Analysis Centre, Nanoscience Technology Center (NSTC), Materials Science and Engineering, College of Medicine, University of Central Florida, USA, Orlando, FL
| | - Melanie J Coathup
- College of Medicine, University of Central Florida, Orlando, FL, USA; Biionix Cluster, University of Central Florida, Orlando, FL, USA.
| |
Collapse
|
30
|
Tang H, Hao R, Ma D, Yao Y, Ding C, Zhang X, Zhang A. Structural Modification and Pharmacological Evaluation of (Thiadiazol-2-yl)pyrazines as Novel Piezo1 Agonists for the Intervention of Disuse Osteoporosis. J Med Chem 2024; 67:19837-19851. [PMID: 39462841 DOI: 10.1021/acs.jmedchem.4c02224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Piezo1 plays a pivotal role in regulating bone remodeling and homeostasis and has emerged as a promising target for chemical intervention in disuse osteoporosis. Nevertheless, the development of small-molecule Piezo1 agonists is still in its infancy, and highly efficacious Piezo1 agonists are urgently required. In this study, by shedding light on the structural novelty of the canonical Piezo1 agonist Yoda1, we initiated a structural optimization campaign based on the (thiadiazol-2-yl)pyrazine scaffold. A deuterated compound 12a was identified to be the most potent candidate against Piezo1 with an EC50 value of 2.21 μM, which was over 20-fold more potent than the reference Yoda1. This compound effectively activated Piezo1 and initiated Ca2+ influx in MSCs and promoted MSC osteogenesis via activating the Ca2+-related Erk signaling pathway. Furthermore, compound 12a was found to alleviate disuse osteoporosis with a desirable safety profile in a HU (hindlimb-unloading) rat model, thus warranting it as a potential probe for further investigation.
Collapse
Affiliation(s)
- Hairong Tang
- Shanghai Frontiers Science Center of Targeted Drugs, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Ruihan Hao
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200092, China
| | - Ding Ma
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200092, China
| | - Yujia Yao
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, China
| | - Chunyong Ding
- Shanghai Frontiers Science Center of Targeted Drugs, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200092, China
| | - Ao Zhang
- Shanghai Frontiers Science Center of Targeted Drugs, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200092, China
- National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
31
|
Zhang T, Bi C, Li Y, Zhao L, Cui Y, Ouyang K, Xiao B. Phosphorylation of Piezo1 at a single residue, serine-1612, regulates its mechanosensitivity and in vivo mechanotransduction function. Neuron 2024; 112:3618-3633.e6. [PMID: 39270653 DOI: 10.1016/j.neuron.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 09/15/2024]
Abstract
Piezo1 is a mechanically activated cation channel that converts mechanical force into diverse physiological processes. Owing to its large protein size of more than 2,500 amino acids and complex 38-transmembrane helix topology, how Piezo1 is post-translationally modified for regulating its in vivo mechanotransduction functions remains largely unexplored. Here, we show that PKA activation potentiates the mechanosensitivity and slows the inactivation kinetics of mouse Piezo1 and identify the major phosphorylation site, serine-1612 (S1612), that also responds to PKC activation and shear stress. Mutating S1612 abolishes PKA and PKC regulation of Piezo1 activities. Primary endothelial cells derived from the Piezo1-S1612A knockin mice lost PKA- and PKC-dependent phosphorylation and functional potentiation of Piezo1. The mutant mice show activity-dependent elevation of blood pressure and compromised exercise endurance, resembling endothelial-specific Piezo1 knockout mice. Taken together, we identify the major PKA and PKC phosphorylation site in Piezo1 and demonstrate its contribution to Piezo1-mediated physiological functions.
Collapse
Affiliation(s)
- Tingxin Zhang
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Frontier Research Center of Biological, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Cheng Bi
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Frontier Research Center of Biological, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Yiran Li
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Frontier Research Center of Biological, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Lingyun Zhao
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Yaxiong Cui
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Frontier Research Center of Biological, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
| | - Kunfu Ouyang
- Department of Cardiovascular Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China.
| | - Bailong Xiao
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Frontier Research Center of Biological, School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
32
|
Jin J, Zandieh-Doulabi B. Low, but Not High, Pulsating Fluid Shear Stress Affects Matrix Extracellular Phosphoglycoprotein Expression, Mainly via Integrin β Subunits in Pre-Osteoblasts. Curr Issues Mol Biol 2024; 46:12428-12441. [PMID: 39590332 PMCID: PMC11593251 DOI: 10.3390/cimb46110738] [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: 10/02/2024] [Revised: 10/30/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
Matrix extracellular phosphoglycoprotein (Mepe), present in bone and dentin, plays important multifunctional roles in cell signaling, bone mineralization, and phosphate homeostasis. Mepe expression in bone cells changes in response to pulsating fluid shear stress (PFSS), which is transmitted into cells through integrin-based adhesion sites, i.e., α and β subunits. Whether and to what extent PFSS influences Mepe expression through the modulation of integrin α and/or β subunit expression in pre-osteoblasts is uncertain. Therefore, we aimed to test whether low and/or high PFSS affects Mepe expression via modulation of integrin α and/or β subunit expression. MC3T3-E1 pre-osteoblasts were treated with ± 1 h PFSS (magnitude: 0.3 Pa (low PFSS) or 0.7 Pa (high PFSS); frequency: 1 Hz). Single integrin fluorescence intensity in pre-osteoblasts was increased, but single integrin area was decreased by low and high PFSS. Expression of two integrin α subunit-related genes (Itga1 and Itga5 2) was increased by low PFSS, and one (Itga5 2) by high PFSS. Expression of five integrin β subunit genes (Itgb1, Itgb3, Itgb5, Itgb5 13, and Itgb5 123) was increased by low PFSS, and three (Itgb5, Itgb5 13, and Itgb5 123) by high PFSS. Interestingly, Mepe expression in pre-osteoblasts was only modulated by low, but not high, PFSS. In conclusion, both low and high PFSS affected integrin α and β subunit expression in pre-osteoblasts, while integrin β subunit expression was more altered by low PFSS. Importantly, Mepe gene expression was only affected by low PFSS. These results might explain the different ways that Mepe-induced changes in pre-osteoblast mechanosensitivity may drive signaling pathways of bone cell function at low or high impact loading. These findings might have physiological and biomedical implications and require future research specifically addressing the precise role of integrin α or β subunits and Mepe during dynamic loading in bone health and disease.
Collapse
Affiliation(s)
- Jianfeng Jin
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, 1081 LA Amsterdam, The Netherlands;
| | | |
Collapse
|
33
|
Kong X, Qin Y, Pei W, Zhou G. Recent progresses on space life science research in China. LIFE SCIENCES IN SPACE RESEARCH 2024; 43:35-42. [PMID: 39521492 DOI: 10.1016/j.lssr.2024.10.002] [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: 09/14/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 11/16/2024]
Abstract
In the past decades, China has made significant progress on space life science research. Since completing the construction of the China Space Station (CSS) at the end of 2022, space life science research in China has entered a new era. Through carrying out numerous experiments on space life sciences, space medicine, and space agriculture conducted aboard the Shenzhou series, the CSS, and ground-based space environment simulation platforms, Chinese scientists have uncovered the effects of the space environment on the physiological and molecular mechanisms of live organisms. These findings provide essential theoretical support for long-term manned space exploration. In this article, we review the new discoveries made by Chinese researchers, focusing on the impacts of both actual and simulated space environment on cells, microorganisms, plants, animals, and human health.
Collapse
Affiliation(s)
- Xiangyu Kong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yuhao Qin
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Weiwei Pei
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| | - Guangming Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| |
Collapse
|
34
|
Wasi M, Chu T, Guerra RM, Kooker R, Maldonado K, Li X, Lin CY, Song X, Xiong J, You L, Wang L. Mitigating aging and doxorubicin induced bone loss in mature mice via mechanobiology based treatments. Bone 2024; 188:117235. [PMID: 39147353 PMCID: PMC11475016 DOI: 10.1016/j.bone.2024.117235] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Aging leads to a reduced anabolic response to mechanical stimuli and a loss of bone mass and structural integrity. Chemotherapy agents such as doxorubicin exacerbate the degeneration of aging skeleton and further subject older cancer patients to a higher fracture risk. To alleviate this clinical problem, we proposed and tested a novel mechanobiology-based therapy. Building upon prior findings that i) Yoda1, the Piezo1 agonist, promoted bone growth in young adult mice and suppressed bone resorption markers in aged mice, and ii) moderate tibial loading protected bone from breast cancer-induced osteolysis, we hypothesized that combined Yoda1 and moderate loading would improve the structural integrity of adult and aged skeletons in vivo and protect bones from deterioration after chemotherapy. We first examined the effects of 4-week Yoda1 (dose 5 mg/kg, 5 times/week) and moderate tibial loading (4.5 N peak load, 4 Hz, 300 cycles for 5 days/week), individually and combined, on mature mice (∼50 weeks of age). Combined Yoda1 and loading was found to mitigate age-associated cortical and trabecular bone loss better than individual interventions. As expected, the non-treated controls experienced an average drop of cortical polar moment of inertia (Ct.pMOI) by -4.3 % over four weeks and the bone deterioration occurred in the majority (64 %) of the samples. Relative to no treatment, loading alone, Yoda1 alone, and combined Yoda1 and loading increased Ct.pMOI by +7.3 %, +9.5 %, +12.0 % and increased the % of samples with positive Ct.pMOI changes by +32 %, +26 %, and +43 %, respectively, suggesting an additive protection of aging-related bone loss for the combined therapy. We further tested if the treatment efficacy was preserved in mature mice following two weeks (six injections) of doxorubicin at the dose of 2.5 or 5 mg/kg. As expected, doxorubicin increased osteocyte apoptosis, altered bone remodeling, and impaired bone structure. However, the effects induced by DOX were too severe to be rescued by Yoda1 and loading, alone or combined, although loading and Yoda1 individually, or combined, increased the number of mice showing positive responsiveness by 0 %, +15 %, and +29 % relative to no intervention after doxorubicin exposure. Overall, this study supported the potentials and challenges of the Yoda1-based strategy in mitigating the detrimental skeletal effects caused by aging and doxorubicin.
Collapse
Affiliation(s)
- Murtaza Wasi
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Tiankuo Chu
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Rosa M Guerra
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA
| | - Rory Kooker
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - Kenneth Maldonado
- Department of Biomedical Engineering, Kansas State University, Manhattan, KS, USA
| | - Xuehua Li
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Chun-Yu Lin
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Xin Song
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Lidan You
- Department of Mechanical and Industrial Engineering, Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| | - Liyun Wang
- Department of Mechanical Engineering, University of Delaware, Newark, DE, USA; Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
35
|
Li Q, Chen X, Li X, Jiang X, Li X, Men X, Li Y, Chen S. Plexin-B2 Mediates Orthodontic Tension-Induced Osteogenesis via the RhoA/F-Actin/YAP Pathway. J Periodontal Res 2024. [PMID: 39485327 DOI: 10.1111/jre.13358] [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: 08/11/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024]
Abstract
AIMS This study aims to investigate the role of Plexin-B2 in tension-induced osteogenesis of periodontal ligament stem cells (PDLSCs) and its biomechanical mechanism. METHODS In vitro, cyclic tension simulated orthodontic forces to assess Plexin-B2 expression in PDLSCs. We then knocked out Plexin-B2 using lentivirus to explore its role in tension-induced osteogenesis. In vivo, we used nickel-titanium springs to establish orthodontic tooth movement (OTM) models in mice. Local periodontal Plexin-B2 expression was knocked down using adeno-associated viruses (AAVs) to study its influence on new bone formation under mechanical tension in OTM models. Molecular mechanisms were elucidated by manipulating Plexin-B2 and RhoA expression, assessing related proteins, and observing F-actin and Yes-associated protein (YAP) through immunofluorescence. RESULTS Plexin-B2 expression in PDLSCs increased under cyclic tension. Decrease of Plexin-B2 reduced the expression of osteogenic protein in PDLSCs and negatively affected new bone formation during OTM. RhoA expression and phosphorylation of ROCK2/LIMK2/Cofilin decreased in Plexin-B2 knockout PDLSCs but were reversed by RhoA overexpression. The level of F-actin decreased in Plexin-B2 knockout PDLSCs but increased after RhoA rescue. Nuclear YAP was reduced in Plexin-B2 knockout PDLSCs but increased after RhoA overexpression. CONCLUSIONS Plexin-B2 is involved in tension-induced osteogenesis. Mechanistically, the RhoA signaling pathway, the F-actin arrangement, and the nuclear translocation of YAP are involved in the mechanotransduction of Plexin-B2.
Collapse
Affiliation(s)
- Qiming Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyi Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoge Jiang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingjian Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinrui Men
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan Li
- Department of Stomatology, Inner Mongolia Hulunbuir Yakeshi Municipal People's Hospital, Hulunbuir, China
| | - Song Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Xie B, He X, Guo Y, Shen J, Yang B, Cai R, Chen J, He Y. Cyclic tensile stress promotes osteogenic differentiation via upregulation of Piezo1 in human dental follicle stem cells. Hum Cell 2024; 37:1649-1662. [PMID: 39190266 DOI: 10.1007/s13577-024-01123-5] [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: 03/11/2024] [Accepted: 08/17/2024] [Indexed: 08/28/2024]
Abstract
As periodontal progenitor cells, human dental follicle stem cells (hDFCs) play an important role in regenerative medicine research. Mechanical stimuli exert different regulatory effects on various functions of stem cells. Mechanosensitive ion channels can perceive and transmit mechanical signals. Piezo1 is a novel mechanosensitive cation channel dominated by Ca2+ permeation. The yes-associated protein 1 (YAP1) and mitogen-activated protein kinase (MAPK) pathways can respond to mechanical stimuli and play important roles in cell growth, differentiation, apoptosis, and cell cycle regulation. In this study, we demonstrated that Piezo1 was able to transduce cyclic tension stress (CTS) and promote the osteogenic differentiation of hDFCs by applying CTS of 2000 μstrain to hDFCs. Further investigation of this mechanism revealed that CTS activated Piezo1 in hDFCs and resulted in increased levels of intracellular Ca2+, YAP1 nuclear translocation, and phosphorylated protein expression levels of extracellular signalling-associated kinase 1/2 (ERK 1/2) and Jun amino-terminal kinase 1/2/3 (JNK 1/3) of the MAPK pathway family. However, when Piezo1 was knocked down in the hDFCs, all these increases disappeared. We conclude that CTS activates Piezo1 expression and promotes its osteogenesis via Ca2+/YAP1/MAPK in hDFCs. Appropriate mechanical stimulation promotes the osteogenic differentiation of hDFCs via Piezo1. Targeting Piezo1 may be an effective strategy to regulate the osteogenic differentiation of hDFCs, contributing to MSC-based therapies in the field of bone tissue engineering.
Collapse
Affiliation(s)
- Binqing Xie
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Xianyi He
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Ye Guo
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Jie Shen
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Binbin Yang
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Rui Cai
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China
| | - Junliang Chen
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China.
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China.
| | - Yun He
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Yunfenglu 10, Luzhou, 646000, China.
- Luzhou Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Southwest Medical University, Xianglinlu 1, Luzhou, 646000, China.
| |
Collapse
|
37
|
Xiao B. Mechanisms of mechanotransduction and physiological roles of PIEZO channels. Nat Rev Mol Cell Biol 2024; 25:886-903. [PMID: 39251883 DOI: 10.1038/s41580-024-00773-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/11/2024]
Abstract
Mechanical force is an essential physical element that contributes to the formation and function of life. The discovery of the evolutionarily conserved PIEZO family, including PIEZO1 and PIEZO2 in mammals, as bona fide mechanically activated cation channels has transformed our understanding of how mechanical forces are sensed and transduced into biological activities. In this Review, I discuss recent structure-function studies that have illustrated how PIEZO1 and PIEZO2 adopt their unique structural design and curvature-based gating dynamics, enabling their function as dedicated mechanotransduction channels with high mechanosensitivity and selective cation conductivity. I also discuss our current understanding of the physiological and pathophysiological roles mediated by PIEZO channels, including PIEZO1-dependent regulation of development and functional homeostasis and PIEZO2-dominated mechanosensation of touch, tactile pain, proprioception and interoception of mechanical states of internal organs. Despite the remarkable progress in PIEZO research, this Review also highlights outstanding questions in the field.
Collapse
Affiliation(s)
- Bailong Xiao
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
- Beijing Frontier Research Center of Biological Structure, Tsinghua University, Beijing, China.
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- State Key Laboratory of Membrane Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
38
|
Valentino A, Yazdanpanah S, Conte R, Calarco A, Peluso G. Smart Nanocomposite Hydrogels as Next-Generation Therapeutic and Diagnostic Solutions. Gels 2024; 10:689. [PMID: 39590045 PMCID: PMC11594247 DOI: 10.3390/gels10110689] [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: 10/01/2024] [Revised: 10/22/2024] [Accepted: 10/23/2024] [Indexed: 11/28/2024] Open
Abstract
Stimuli-responsive nanocomposite gels combine the unique properties of hydrogels with those of nanoparticles, thus avoiding the suboptimal results of single components and creating versatile, multi-functional platforms for therapeutic and diagnostic applications. These hybrid materials are engineered to respond to various internal and external stimuli, such as temperature, pH, light, magnetic fields, and enzymatic activity, allowing precise control over drug release, tissue regeneration, and biosensing. Their responsiveness to environmental cues permits personalized medicine approaches, providing dynamic control over therapeutic interventions and real-time diagnostic capabilities. This review explores recent advances in stimuli-responsive hybrid gels' synthesis and application, including drug delivery, tissue engineering, and diagnostics. Overall, these platforms have significant clinical potential, and future research is expected to lead to unique solutions to address unmet medical needs.
Collapse
Affiliation(s)
- Anna Valentino
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; (A.V.); (S.Y.); (G.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Sorur Yazdanpanah
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; (A.V.); (S.Y.); (G.P.)
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, Via Santa Maria di Costantinopoli 16, 80138 Naples, Italy
| | - Raffaele Conte
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; (A.V.); (S.Y.); (G.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Anna Calarco
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; (A.V.); (S.Y.); (G.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
| | - Gianfranco Peluso
- Research Institute on Terrestrial Ecosystems (IRET)-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; (A.V.); (S.Y.); (G.P.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Faculty of Medicine and Surgery, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
| |
Collapse
|
39
|
Zhan H, Xie D, Yan Z, Yi Z, Xiang D, Niu Y, Liang X, Geng B, Wu M, Xia Y, Jiang J. Fluid shear stress-mediated Piezo1 alleviates osteocyte apoptosis by activating the PI3K/Akt pathway. Biochem Biophys Res Commun 2024; 730:150391. [PMID: 39002199 DOI: 10.1016/j.bbrc.2024.150391] [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: 05/15/2024] [Revised: 07/01/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
Glucocorticoid-induced osteoporosis serves as a primary cause for secondary osteoporosis and fragility fractures, representing the most prevalent adverse reaction associated with prolonged glucocorticoid use. In this study, to elucidate the impact and underlying mechanisms of fluid shear stress (FSS)-mediated Piezo1 on dexamethasone (Dex)-induced apoptosis, we respectively applied Dex treatment for 6 h, FSS at 9 dyne/cm2 for 30 min, Yoda1 treatment for 2 h, and Piezo1 siRNA transfection to intervene in MLO-Y4 osteocytes. Western blot analysis was used to assess the expression of Cleaved Caspase-3, Bax, Bcl-2, and proteins associated with the PI3K/Akt pathway. Additionally, qRT-PCR was utilized to quantify the mRNA expression levels of these molecules. Hoechst 33258 staining and flow cytometry were utilized to evaluate the apoptosis levels. The results indicate that FSS at 9 dyne/cm2 for 30 min significantly upregulates Piezo1 in osteocytes. Following Dex-induced apoptosis, the phosphorylation levels of PI3K and Akt are markedly suppressed. FSS-mediated Piezo1 exerts a protective effect against Dex-induced apoptosis by activating the PI3K/Akt pathway. Additionally, downregulating the expression of Piezo1 in osteocytes using siRNA exacerbates Dex-induced apoptosis. To further demonstrate the role of the PI3K/Akt signaling pathway, after intervention with the PI3K pathway inhibitor, the activation of the PI3K/Akt pathway by FSS-mediated Piezo1 in osteocytes was significantly inhibited, reversing the anti-apoptotic effect. This study indicates that under FSS, Piezo1 in MLO-Y4 osteocytes is significantly upregulated, providing protection against Dex-induced apoptosis through the activation of the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Hongwei Zhan
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China; Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Daijun Xie
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
| | - Zhenxing Yan
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
| | - Zhi Yi
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
| | - Dejian Xiang
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
| | - Yongkang Niu
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
| | - Xiaoyuan Liang
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
| | - Bin Geng
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
| | - Meng Wu
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
| | - Yayi Xia
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
| | - Jin Jiang
- The Second Hospital of Lanzhou University, Orthopaedic Clinical Research Center of Gansu Province, Intelligent Orthopaedic Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
| |
Collapse
|
40
|
Li X, Zhang C, Vail CE, Sherrill JT, Xiong J. Piezo1 expression in mature osteocytes is dispensable for the skeletal response to mechanical loading. Bone 2024; 190:117276. [PMID: 39389439 DOI: 10.1016/j.bone.2024.117276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/04/2024] [Accepted: 10/06/2024] [Indexed: 10/12/2024]
Abstract
Mechanical loading is essential for bone growth and homeostasis, with osteocytes considered the primary mechanosensors. Deleting the mechanosensitive ion channel Piezo1 from Dmp1-Cre-targeted cells, which include both osteoblasts and osteocytes, resulted in reduced bone mass and impaired skeletal responses to mechanical stimuli. To specifically isolate Piezo1's role in osteocytes, we used Sost-Cre mice to generate mice with Piezo1 deletion exclusively in mature osteocytes. These mice exhibited lower bone mineral density, decreased cancellous bone mass, and reduced cortical thickness with decrease periosteal expansion. However, unlike mice lacking Piezo1 in both osteoblasts and osteocytes, those with Piezo1 deletion in mature osteocytes responded normally to mechanical loading. These findings suggest that Piezo1 expression in mature osteocytes contributes to bone maintenance under normal physiological condition, but is dispensable for the skeletal response to mechanical loading.
Collapse
Affiliation(s)
- Xuehua Li
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Connie Zhang
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Cameron E Vail
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - John T Sherrill
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jinhu Xiong
- Department of Orthopaedic Surgery, University of Arkansas for Medical Sciences, Little Rock, AR, USA; Center for Musculoskeletal Disease Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
41
|
Wang B, Shao W, Zhao Y, Li Z, Wang P, Lv X, Chen Y, Chen X, Zhu Y, Ma Y, Han L, Wu W, Feng Y. Radial extracorporeal shockwave promotes osteogenesis-angiogenesis coupling of bone marrow stromal cells from senile osteoporosis via activating the Piezo1/CaMKII/CREB axis. Bone 2024; 187:117196. [PMID: 39004161 DOI: 10.1016/j.bone.2024.117196] [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: 09/11/2023] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 07/16/2024]
Abstract
Radial extracorporeal shockwave (r-ESW) and bone marrow stromal cells (BMSCs) have been reported to alleviate senile osteoporosis (SOP), but its regulatory mechanism remains unclear. In this study, we firstly isolated human BMSCs from bone marrow samples and treated with varying r-ESW doses. And we found that r-ESW could enhance the proliferation of SOP-BMSCs in a dose-dependent manner by EdU assay. Subsequently, the impact of r-ESW on the proliferation, apoptosis and multipotency of BMSCs was assessed. And the outcomes of flow cytometry, Alizarin red S (ARS), and tube formation test demonstrated that the optimal shockwave obviously boosted SOP-BMSCs osteogenesis and angiogenesis but exhibited no significant impact on cell apoptosis. Additionally, the signaling of Piezo1 and CaMKII/CREB was examined by Western blotting, qPCR and immunofluorescence. And the results showed that r-ESW promoted the expression of Piezo1, increased intracellular Ca2+ and activated the CaMKII/CREB signaling pathway. Then, the application of Piezo1 siRNA hindered the r-ESW-induced enhancement ability of osteogenesis coupling with angiogenesis of SOP-BMSCs. The use of the CaMKII/CREB signaling pathway inhibitor KN93 suppressed the Piezo1-induced increase in osteogenesis and angiogenesis in SOP-BMSCs. Finally, we also found that r-ESW might alleviate SOP in the senescence-accelerated mouse prone 6 (SAMP6) model by activating Piezo1. In conclusion, our research offers experimental evidence and an elucidated underlying molecular mechanism to support the use of r-ESW as a credible rehabilitative treatment for senile osteoporosis.
Collapse
Affiliation(s)
- Bo Wang
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wenkai Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yubai Zhao
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Zilin Li
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Ping Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiao Lv
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yongjin Chen
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaodong Chen
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Anhui Key Laboratory of Tissue Transformation, Bengbu Medical University, Bengbu 233000, Anhui Province, PR China
| | - Yuanxiao Zhu
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yan Ma
- Department of Rehabilitation, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lizhi Han
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, Anhui Key Laboratory of Tissue Transformation, Bengbu Medical University, Bengbu 233000, Anhui Province, PR China.
| | - Wen Wu
- Department of Rehabilitation, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| | - Yong Feng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
42
|
Yuze Ma, Liu N, Shao X, Shi T, Lin J, Liu B, Shen T, Guo B, Jiang Q. Mechanical loading on osteocytes regulates thermogenesis homeostasis of brown adipose tissue by influencing osteocyte-derived exosomes. J Orthop Translat 2024; 48:39-52. [PMID: 39087139 PMCID: PMC11287067 DOI: 10.1016/j.jot.2024.06.012] [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: 01/16/2024] [Revised: 05/25/2024] [Accepted: 06/19/2024] [Indexed: 08/02/2024] Open
Abstract
Background Osteocytes are the main stress-sensing cells in bone. The substances secreted by osteocytes under mechanical loading play a crucial role in maintaining body homeostasis. Osteocytes have recently been found to release exosomes into the circulation, but whether they are affected by mechanical loading or participate in the regulation of systemic homeostasis remains unclear. Methods We used a tail-suspension model to achieve mechanical unloading on osteocytes. Osteocyte-specific CD63 reporter mice were used for osteocyte exosome tracing. Exosome detection and inhibitor treatment were performed to confirm the effect of mechanical loading on exosome secretion by osteocytes. Co-culture, GW4869 and exosome treatment were used to investigate the biological functions of osteocyte-derived exosomes on brown adipose tissue (BAT) and primary brown adipocytes. Osteocyte-specific Dicer KO mice were used to screen for loading-sensitive miRNAs. Dual luciferase assay was performed to validate the selected target gene. Results Firstly, we found the thermogenic activity was increased in BAT of mice subjected to tail suspension, which is due to the effect of unloaded bone on circulating exosomes. Further, we showed that the secretion of exosomes from osteocytes is regulated by mechanical loading, and osteocyte-derived exosomes can reach BAT and affect thermogenic activity. More importantly, we confirmed the effect of osteocyte exosomes on BAT both in vivo and in vitro. Finally, we discovered that let-7e-5p contained in exosomes is under regulation of mechanical loading and regulates thermogenic activity of BAT by targeting Ppargc1a. Conclusion Exosomes derived from osteocytes are loading-sensitive, and play a vital role in regulation on BAT, suggesting that regulation of exosomes secretion can restore homeostasis. The translational potential of this article This study provides a biological rationale for using osteocyte exosomes as potential agents to modulate BAT and even whole-body homeostasis. It also provides a new pathological basis and a new treatment approach for mechanical unloading conditions such as spaceflight.
Collapse
Affiliation(s)
- Yuze Ma
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Na Liu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Xiaoyan Shao
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Tianshu Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jiaquan Lin
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Bin Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tao Shen
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Baosheng Guo
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
43
|
Doshi A, Patel J, Gaur V, Fernandes G, Awadalkreem F. Assessment of the Peri-implant Bone Density Following Bicortical Anchored Corticobasal Implants Placement in the Maxillary Arch: A Cross-sectional Prospective Study. J Contemp Dent Pract 2024; 25:820-829. [PMID: 39791408 DOI: 10.5005/jp-journals-10024-3755] [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] [Indexed: 01/12/2025]
Abstract
AIM The aim of this cross-sectional prospective study was to evaluate the bone density changes around the bicortical corticobasal implant placed in the maxilla over 18 months of follow-up using cone-beam computed tomography (CBCT), focusing on the comparison between the anterior and posterior teeth and regions. MATERIALS AND METHODS Thirty-five subjects (20, 53.26%, were males, and 15, 46.73%, were females) received 380 implants (Basal Cortical Screwable implant, BCS®) at Narsinhbhai Patel Dental College and Hospital, India. Implant survival and success were assessed using Albrektsson criteria for implant success. The peri-implant bone density values were measured using CBCT (Vatech PaX-i 3d Smart) and InVivo software (Anatomage, San Jose, California, USA) at the baseline (immediate postoperative) and at the 18-month follow-up visit. For standardization purposes, the bone density values for only the maxillary implants were measured at the level of the second implant thread in four sites: buccal, mesial, distal, and palatal, respectively. The recorded data were tabulated and grouped according to the tooth's region (anterior/posterior) and sites (mesial, distal, buccal, and palatal). RESULTS The implant's survival rate was 100%. After 18 months, the bone density increased significantly (p < 0.05) in all the sites in both anterior and posterior regions. The study's findings revealed a higher bone density increase in the posterior region compared to the anterior region after 18 months of follow-up, except for the palatal site. CONCLUSION Within the limitations of this study, an increase in the peri-implant bone density has been associated with the use of corticobasal implants over time, with reported anterior/posterior regional variations. CLINICAL SIGNIFICANCE This study provides valuable insights into the bone density changes associated with bicortical corticobasal implants and emphasizes the importance of CBCT in evaluating bone density, as well as the significance of regional considerations in implant dentistry. By integrating these findings into clinical practice, clinicians can improve treatment outcomes and ensure long-term implant survival. How to cite this article: Doshi A, Patel J, Gaur V, et al. Assessment of the Peri-implant Bone Density Following Bicortical Anchored Corticobasal Implants Placement in the Maxillary Arch: A Cross-sectional Prospective Study. J Contemp Dent Pract 2024;25(9):820-829.
Collapse
Affiliation(s)
- Anita Doshi
- Department of Prosthodontics, Sankalchand Patel University, Visnagar, Gujarat, India, Phone: +91 9320030735, e-mail:
| | - Jayantilal Patel
- Department of Health Sciences, Sankalchand Patel University, Visnagar, Gujarat, India
| | - Vivek Gaur
- Department of Prosthodontics, Sankalchand Patel University, Visnagar, Gujarat, India
| | - Gabriela Fernandes
- Department of Periodontics and Endodontics, SUNY Buffalo, New York, United States of America
| | - Fadia Awadalkreem
- RAK College of Dental Sciences, RAK Medical and Health Sciences University, Ras Al Khaimah, United Arab Emirates
| |
Collapse
|
44
|
Zhou H, Liu H, Lin M, Wang H, Zhou J, Li M, Yang X, Fu G, Liu C. Hyperbaric oxygen promotes bone regeneration by activating the mechanosensitive Piezo1 pathway in osteogenic progenitors. J Orthop Translat 2024; 48:11-24. [PMID: 39170748 PMCID: PMC11338066 DOI: 10.1016/j.jot.2024.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/17/2024] [Accepted: 07/03/2024] [Indexed: 08/23/2024] Open
Abstract
Background Hyperbaric oxygen (HBO) therapy is widely used to treat bone defects, but the correlation of high oxygen concentration and pressure to osteogenesis is unclear. Methods Bilateral monocortical tibial defect surgeries were performed on 12-week-old Prrx1-Cre; Rosa26-tdTomato and Prrx1-Cre; Piezo1fl/+ mice. Daily HBO treatment was applied on post-surgery day (PSD) 1-9; and daily mechanical loading on tibia was from PSD 5 to 8. The mice were euthanized on PSD 10, and bone defect repair in their tibias was evaluated using μCT, biomechanical testing, and immunofluorescence deep-tissue imaging. The degree of angiogenesis-osteogenesis coupling was determined through spatial correlation analysis. Bone marrow stromal cells from knockout mice were cultured in vitro, and their osteogenic capacities of the cells were assessed. The activation of genes in the Piezo1-YAP pathway was evaluated using RNA sequencing and quantitative real-time polymerase chain reaction. Results Lineage tracing showed HBO therapy considerably altered the number of Prrx1+ cells and their progeny in a healing bone defect. Using conditional knockdown mice, we found that HBO stimulation activates the Piezo1-YAP axis in Prrx1+ cells and promotes osteogenesis-angiogenesis coupling during bone repair. The beneficial effect of HBO was similar to that of anabolic mechanical stimulation, which also acts through the Piezo1-YAP axis. Subsequent transcriptome sequencing results revealed that similar mechanosensitive pathways are activated by HBO therapy in a bone defect. Conclusion HBO therapy promotes bone tissue regeneration through the mechanosensitive Piezo1-YAP pathway in a population of Prrx1+ osteogenic progenitors. Our results contribute to the understanding of the mechanism by which HBO therapy treats bone defects. The Translational Potential of this Article Hyperbaric oxygen therapy is widely used in clinical settings. Our results show that osteogenesis was induced by the activation of the Piezo1-YAP pathway in osteoprogenitors after HBO stimulation, and the underlying mechanism was elucidated. These results may help improve current HBO methods and lead to the formulation of alternative treatments that achieve the same functional outcomes.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| | - Hongzhi Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| | - Minmin Lin
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| | - Hantang Wang
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| | - Jingjing Zhou
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| | - Ming Li
- Department of Rehabilitation Medicine, Shenzhen Children's Hospital, No. 7019 Yitian Road, Futian District, Shenzhen, Guangdong, China
| | - Xue Yang
- Department of Rehabilitation Medicine, Shenzhen Children's Hospital, No. 7019 Yitian Road, Futian District, Shenzhen, Guangdong, China
| | - Guibing Fu
- Department of Pediatric Orthopedics, Shenzhen Children's Hospital, No. 7019 Yitian Road, Futian District, Shenzhen, Guangdong, China
| | - Chao Liu
- Department of Biomedical Engineering, College of Engineering, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Engineering Building south 622, 1088 Xueyuan Avenue, Shenzhen, Guangdong, China
| |
Collapse
|
45
|
Hao R, Tang H, Ding C, Rajbanshi B, Liu Y, Ma D, Duan Z, Qi Y, Dai L, Zhang B, Zhang A, Zhang X. A Novel Piezo1 Agonist Promoting Mesenchymal Stem Cell Proliferation and Osteogenesis to Attenuate Disuse Osteoporosis. SMALL SCIENCE 2024; 4:2400061. [PMID: 40212079 PMCID: PMC11935128 DOI: 10.1002/smsc.202400061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/12/2024] [Indexed: 04/13/2025] Open
Abstract
Disuse osteoporosis (OP) is a state of bone loss due to lack of mechanical stimuli, probably induced by prolonged bed rest, neurological diseases, as well as microgravity. Currently the precise treatment strategies of disuse OP remain largely unexplored. Piezo1, a mechanosensitive calcium (Ca2+) ion channel, is a key force sensor mediating mechanotransduction and it is demonstrated to regulate bone homeostasis and osteogenesis in response to mechanical forces. Using structure-based drug design, a novel small-molecule Piezo1 agonist, MCB-22-174, which can effectively activate Piezo1 and initiate Ca2+ influx, is developed and is more potent than the canonical Piezo1 agonist, Yoda1. Moreover, MCB-22-174 is found as a safe Piezo1 agonist without any signs of serious toxicity. Mechanistically, Piezo1 activation promotes the proliferation of bone marrow mesenchymal stem cells by activating the Ca2+-related extracellular signal-related kinases and calcium-calmodulin (CaM)-dependent protein kinase II (CaMKII) pathway. Importantly, MCB-22-174 could effectively promote osteogenesis and attenuate disuse OP in vivo. Overall, the findings provide a promising therapeutic strategy for disuse OP by chemical activation of Piezo1.
Collapse
Affiliation(s)
- Ruihan Hao
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Hairong Tang
- Shanghai Frontiers Science Center of Targeted DrugsSchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Chunyong Ding
- Shanghai Frontiers Science Center of Targeted DrugsSchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Bhavana Rajbanshi
- Department of Dermatology and VenereologyTongji University School of MedicineShanghai200092China
| | - Yuhang Liu
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Ding Ma
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Zhouyi Duan
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
| | - Yuxin Qi
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and MinistryGuangxi Medical UniversityNanningGuangxi530021China
| | - Liming Dai
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
- National Facility for Translational Medicine (Shanghai)Shanghai200240China
| | - Bingjun Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
- National Facility for Translational Medicine (Shanghai)Shanghai200240China
| | - Ao Zhang
- Shanghai Frontiers Science Center of Targeted DrugsSchool of Pharmaceutical SciencesShanghai Jiao Tong UniversityShanghai200240China
| | - Xiaoling Zhang
- Department of Orthopedic SurgeryXin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine (SJTUSM)Shanghai200092China
- National Facility for Translational Medicine (Shanghai)Shanghai200240China
| |
Collapse
|
46
|
Zeng Y, Shen J, Zhou X, Ouyang Z, Zhong J, Qin Y, Jin L, He X, Li L, Xie J, Liu X. Osteogenic differentiation of bone mesenchymal stem cells on linearly aligned triangular micropatterns. J Mater Chem B 2024; 12:8420-8430. [PMID: 39093007 DOI: 10.1039/d4tb01218f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Mesenchymal stem cells (MSCs) hold promise for regenerative medicine, particularly for bone tissue engineering. However, directing MSC differentiation towards specific lineages, such as osteogenic, while minimizing undesired phenotypes remains a challenge. Here, we investigate the influence of micropatterns on the behavior and lineage commitment of rat bone marrow-derived MSCs (rBMSCs), focusing on osteogenic differentiation. Linearly aligned triangular micropatterns (TPs) and circular micropatterns (CPs) coated with fibronectin were fabricated to study their effects on rBMSC morphology and differentiation and the underlying mechanobiological mechanisms. TPs, especially TP15 (15 μm), induced the cell elongation and thinning, while CPs also promoted the cell stretching, as evidenced by the decreased circularity and increased aspect ratio. TP15 significantly promoted osteogenic differentiation, with increased expression of osteogenic genes (Runx2, Spp1, Alpl, Bglap, Col1a1) and decreased expression of adipogenic genes (Pparg, Cebpa, Fabp4). Conversely, CPs inhibited both osteogenic and adipogenic differentiation. Mechanistically, TP15 increased Piezo1 activity, cytoskeletal remodeling including the aggregates of F-actin and myosin filaments at the cell periphery, YAP1 nuclear translocation, and integrin upregulation. Piezo1 inhibition suppressed the osteogenic genes expression, myosin remodeling, and YAP1 nuclear translocation, indicating Piezo1-mediated the mechanotransduction in rBMSCs on TPs. TP15 also induced osteogenic differentiation of BMSCs from aging rats, with upregulated Piezo1 and nuclear translocation of YAP1. Therefore, triangular micropatterns, particularly TP15, promote osteogenesis and inhibit adipogenesis of rBMSCs through Piezo1-mediated myosin and YAP1 pathways. Our study provides novel insights into the mechanobiological mechanisms governing MSC behaviors on micropatterns, offering new strategies for tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Junyi Shen
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Xintong Zhou
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Zhi Ouyang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Jian Zhong
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Yixue Qin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Linlu Jin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Xueling He
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Liang Li
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, P. R. China.
| |
Collapse
|
47
|
Aragona M, Mhalhel K, Pansera L, Montalbano G, Guerrera MC, Levanti M, Laurà R, Abbate F, Vega JA, Germanà A. Localization of Piezo 1 and Piezo 2 in Lateral Line System and Inner Ear of Zebrafish ( Danio rerio). Int J Mol Sci 2024; 25:9204. [PMID: 39273152 PMCID: PMC11395407 DOI: 10.3390/ijms25179204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Piezo proteins have been identified as mechanosensitive ion channels involved in mechanotransduction. Several ion channel dysfunctions may be associated with diseases (including deafness and pain); thus, studying them is critical to understand their role in mechanosensitive disorders and to establish new therapeutic strategies. The current study investigated for the first time the expression patterns of Piezo proteins in zebrafish octavolateralis mechanosensory organs. Piezo 1 and 2 were immunoreactive in the sensory epithelia of the lateral line system and the inner ear. Piezo 1 (28.7 ± 1.55 cells) and Piezo 2 (28.8 ± 3.31 cells) immunopositive neuromast cells were identified based on their ultrastructural features, and their overlapping immunoreactivity to the s100p specific marker (28.6 ± 1.62 cells), as sensory cells. These findings are in favor of Piezo proteins' potential role in sensory cell activation, while their expression on mantle cells reflects their implication in the maintenance and regeneration of the neuromast during cell turnover. In the inner ear, Piezo proteins' colocalization with BDNF introduces their potential implication in neuronal plasticity and regenerative events, typical of zebrafish mechanosensory epithelia. Assessing these proteins in zebrafish could open up new scenarios for the roles of these important ionic membrane channels, for example in treating impairments of sensory systems.
Collapse
Affiliation(s)
- Marialuisa Aragona
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Kamel Mhalhel
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Lidia Pansera
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Giuseppe Montalbano
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Maria Cristina Guerrera
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Maria Levanti
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Rosaria Laurà
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - Francesco Abbate
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| | - José A Vega
- Departamento de Morfología y Biología Celular, Grupo SINPOS, Universidad de Oviedo, 33006 Oviedo, Spain
- Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 7500912, Chile
| | - Antonino Germanà
- Zebrafish Neuromorphology Lab, Department of Veterinary Sciences, University of Messina, 98168 Messina, Italy
| |
Collapse
|
48
|
Nakamichi R, Asahara H. The role of mechanotransduction in tendon. J Bone Miner Res 2024; 39:814-820. [PMID: 38795012 PMCID: PMC11301520 DOI: 10.1093/jbmr/zjae074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/04/2024] [Accepted: 05/24/2024] [Indexed: 05/27/2024]
Abstract
Tendons play an important role in the maintenance of motor function by connecting muscles and bones and transmitting forces. Particularly, the role of mechanical stress has primarily focused on the key mechanism of tendon homeostasis, with much research on this topic. With the recent development of molecular biological techniques, the mechanisms of mechanical stress sensing and signal transduction have been gradually elucidated with the identification of mechanosensor in tendon cells and the master regulator in tendon development. This review provides a comprehensive overview of the structure and function of tendon tissue, including the role for physical performance and the detailed mechanism of mechanotransduction in its regulation. An important lesson is that the role of mechanotransduction in tendon tissue is only partially clarified, indicating the complexity of the mechanisms of motor function and fueling increasing interest in uncovering these mechanisms.
Collapse
Affiliation(s)
- Ryo Nakamichi
- Department of Molecular and Cellular Biology, Scripps Research, 10550 North Torrey Pines Road, MBB-102, La Jolla, CA 92037, United States
- Department of Systems Biomedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
- Department of Orthopaedic Surgery, Okayama University hospital, 2-5-1 Shikata-Cho, Kita-Ku, Okayama 700-8558, Japan
| | - Hiroshi Asahara
- Department of Molecular and Cellular Biology, Scripps Research, 10550 North Torrey Pines Road, MBB-102, La Jolla, CA 92037, United States
- Department of Systems Biomedicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-Ku, Tokyo 113-8510, Japan
| |
Collapse
|
49
|
Uchinuma M, Taketani Y, Kanaya R, Yamane Y, Shiota K, Suzuki R, Ishii M, Inomata M, Hayashi J, Shin K. Role of Piezo1 in modulating the RANKL/OPG ratio in mouse osteoblast cells exposed to Porphyromonas gingivalis lipopolysaccharide and mechanical stress. J Periodontal Res 2024; 59:749-757. [PMID: 38623787 DOI: 10.1111/jre.13265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 03/19/2024] [Accepted: 03/24/2024] [Indexed: 04/17/2024]
Abstract
AIMS Excessive occlusal force with periodontitis leads to rapid alveolar bone resorption. However, the molecular mechanism by which inflammation and mechanical stress cause bone resorption remains unclear. We examined the role of Piezo1, a mechanosensitive ion channel expressed on osteoblasts, in the changes in the receptor activator of nuclear factor-kappa B ligand (RANKL)/osteoprotegerin (OPG) ratio in mouse MC3T3-E1 osteoblast-like cells under Porphyromonas gingivalis lipopolysaccharide (P.g.-LPS) and mechanical stress. METHODS To investigate the effect of P.g.-LPS and mechanical stress on the RANKL/OPG ratio and Piezo1 expression, we stimulated MC3T3-E1 cells with P.g.-LPS. After 3 days in culture, shear stress, a form of mechanical stress, was applied to the cells using an orbital shaker. Subsequently, to investigate the role of Piezo1 in the change of RANKL/OPG ratio, we inhibited Piezo1 function by knockdown via Piezo1 siRNA transfection or by adding GsMTx4, a Piezo1 antagonist. RESULTS The RANKL/OPG ratio significantly increased in MC3T3-E1 cells cultured in a medium containing P.g.-LPS and undergoing mechanical stress compared to cells treated with P.g.-LPS or mechanical stress alone. However, the expression of Piezo1 was not increased by P.g.-LPS and mechanical stress. In addition, phosphorylation of MEK/ERK was induced in the cells under P.g.-LPS and mechanical stress. MC3T3-E1 cells treated with P.g.-LPS and mechanical stress when cocultured with RAW264.7 cells induced their differentiation into osteoclast-like cells. The increased RANKL/OPG ratio was suppressed by either Piezo1 knockdown or the addition of GsMTx4. Furthermore, GsMTx4 inhibited the phosphorylation of MEK/ERK. CONCLUSION These findings suggest that P.g.-LPS and Piezo1-mediated mechanical stress induce MEK/ERK phosphorylation and increase RANKL expression in osteoblasts. Consequently, this leads to the differentiation of osteoclast precursor cells into osteoclasts.
Collapse
Affiliation(s)
- Mabuki Uchinuma
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Yoshimasa Taketani
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Risako Kanaya
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Yusuke Yamane
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Koichiro Shiota
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Reiji Suzuki
- Division of Oral Rehabilitation, Department of Restorative & Biomaterials Sciences, Meikai University School of Dentistry, Sakado, Japan
| | - Makiko Ishii
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Megumi Inomata
- Division of Microbiology and Immunology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Joichiro Hayashi
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| | - Kitetsu Shin
- Division of Periodontology, Department of Oral Biology and Tissue Engineering, Meikai University School of Dentistry, Sakado, Japan
| |
Collapse
|
50
|
He X, Liu Y, Dai Z, Chen Y, Liu W, Dai H, Hu Y. Yoda1 pretreated BMSC derived exosomes accelerate osteogenesis by activating phospho-ErK signaling via Yoda1-mediated signal transmission. J Nanobiotechnology 2024; 22:407. [PMID: 38987801 PMCID: PMC11234696 DOI: 10.1186/s12951-024-02669-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
Segmental bone defects, arising from factors such as trauma, tumor resection, and congenital malformations, present significant clinical challenges that often necessitate complex reconstruction strategies. Hydrogels loaded with multiple osteogenesis-promoting components have emerged as promising tools for bone defect repair. While the osteogenic potential of the Piezo1 agonist Yoda1 has been demonstrated previously, its hydrophobic nature poses challenges for effective loading onto hydrogel matrices.In this study, we address this challenge by employing Yoda1-pretreated bone marrow-derived mesenchymal stem cell (BMSCs) exosomes (Exo-Yoda1) alongside exosomes derived from BMSCs (Exo-MSC). Comparatively, Exo-Yoda1-treated BMSCs exhibited enhanced osteogenic capabilities compared to both control groups and Exo-MSC-treated counterparts. Notably, Exo-Yoda1-treated cells demonstrated similar functionality to Yoda1 itself. Transcriptome analysis revealed activation of osteogenesis-associated signaling pathways, indicating the potential transduction of Yoda1-mediated signals such as ErK, a finding validated in this study. Furthermore, we successfully integrated Exo-Yoda1 into gelatin methacryloyl (GelMA)/methacrylated sodium alginate (SAMA)/β-tricalcium phosphate (β-TCP) hydrogels. These Exo-Yoda1-loaded hydrogels demonstrated augmented osteogenesis in subcutaneous ectopic osteogenesis nude mice models and in rat skull bone defect model. In conclusion, our study introduces Exo-Yoda1-loaded GELMA/SAMA/β-TCP hydrogels as a promising approach to promoting osteogenesis. This innovative strategy holds significant promise for future widespread clinical applications in the realm of bone defect reconstruction.
Collapse
Affiliation(s)
- Xi He
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, 310002, China
| | - Yanling Liu
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zhongyu Dai
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Yu Chen
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Wenbin Liu
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, 410078, China.
| | - Honglian Dai
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China.
| | - Yihe Hu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of medicine, Hangzhou, 310002, China.
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, China.
| |
Collapse
|