1
|
Yang K, Li H, Peng R, Wu B, Shen Y, Zhao T, Li C, Wang W, Wang H. The MTMR11 variants identified in a short stature cohort compromise the dephosphorylation ability of MTM1 on SMAD5 to up-regulate BMP signaling. Genes Dis 2025; 12:101393. [PMID: 40236666 PMCID: PMC11999603 DOI: 10.1016/j.gendis.2024.101393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/09/2024] [Indexed: 04/17/2025] Open
Affiliation(s)
- Kai Yang
- Obstetrics & Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200032, China
| | - Hongdou Li
- Obstetrics & Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200032, China
| | - Rui Peng
- Obstetrics & Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200032, China
| | - Bo Wu
- Prenatal Diagnosis Center of Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, Guangdong 518028, China
| | - Yiping Shen
- Genetic and Metabolic Central Laboratory, Birth Defects Prevention and Control Institute of Guangxi Zhuang Autonomous Region, Maternal and Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi 530003, China
| | - Tongjin Zhao
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai 200438, China
| | - Chentao Li
- Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Weimin Wang
- Department of Pharmacy College of Life Sciences, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Hongyan Wang
- Obstetrics & Gynecology Hospital, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200032, China
- Prenatal Diagnosis Center of Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, Guangdong 518028, China
- Children's Hospital, Fudan University, Shanghai 201102, China
| |
Collapse
|
2
|
Kang WY, Jung S, Jeong H, Woo HM, Kang MH, Bae H, Cha JM. Effect of Mechanical Environment Alterations in 3D Stem Cell Culture on the Therapeutic Potential of Extracellular Vesicles. Biomater Res 2025; 29:0189. [PMID: 40416939 PMCID: PMC12099057 DOI: 10.34133/bmr.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/13/2025] [Accepted: 03/20/2025] [Indexed: 05/27/2025] Open
Abstract
Stem-cell-derived extracellular vesicles (EVs) have emerged as a promising therapeutic option, addressing the limitations of conventional stem cell therapies. However, the variability and poorly defined therapeutic contents of EVs produced under standard 2-dimensional culture conditions present challenges for their clinical application. In this study, we investigated how the therapeutic properties of mesenchymal stem cell (MSC)-derived EVs can be enhanced by culturing MSCs within 3-dimensional hydrogels that have tunable mechanical properties. Our results demonstrate that different mechanical cues from the culture environment can induce specific gene expression changes in MSCs without compromising their inherent characteristics. Furthermore, EVs derived from these MSCs exhibited distinct angiogenic and immunomodulatory activities, which were dependent on the mechanical properties of the hydrogels used. A comprehensive analysis of the cytokines and microRNAs present in the EVs provided additional validation of these findings. By utilizing a noninvasive culture method that eliminates the need for genetic modification or exogenous biochemical supplementation, our approach presents a novel platform for the tailored production of EVs, thereby enhancing their therapeutic potential in regenerative medicine.
Collapse
Affiliation(s)
- Wu Young Kang
- Department of Biomedical & Robotics Engineering, College of Engineering,
Incheon National University, Incheon 22012, Republic of Korea
- 3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology,
Incheon National University, Incheon 22012, Republic of Korea
| | - Sunyoung Jung
- Department of Biomedical & Robotics Engineering, College of Engineering,
Incheon National University, Incheon 22012, Republic of Korea
- Department of BioMedical Sciences,
Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Hyundoo Jeong
- Department of Biomedical & Robotics Engineering, College of Engineering,
Incheon National University, Incheon 22012, Republic of Korea
| | - Hyun-Myung Woo
- Department of Biomedical & Robotics Engineering, College of Engineering,
Incheon National University, Incheon 22012, Republic of Korea
| | - Min-Ho Kang
- Department of BioMedical-Chemical Engineering (BMCE),
The Catholic University of Korea, Bucheon 14662, Republic of Korea
- Department of Biotechnology,
The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hojae Bae
- Department of Stem Cell and Regenerative Biotechnology, KU Convergence Science and Technology Institute,
Konkuk University, Seoul 05029, Republic of Korea
- Institute of Advanced Regenerative Science,
Konkuk University, Seoul 05029, Republic of Korea
| | - Jae Min Cha
- Department of Biomedical & Robotics Engineering, College of Engineering,
Incheon National University, Incheon 22012, Republic of Korea
- 3D Stem Cell Bioengineering Laboratory, Research Institute for Engineering and Technology,
Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
3
|
Liu B, Mao X, Gao ZJY, Wang H. Natural traditional Chinese medicine products: emerging therapeutic targets for the treatment of osteoporosis. J Orthop Surg Res 2025; 20:469. [PMID: 40380244 PMCID: PMC12083174 DOI: 10.1186/s13018-025-05879-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 05/01/2025] [Indexed: 05/19/2025] Open
Abstract
Osteoporosis is a systemic metabolic degenerative bone disease characterised by decreased bone mass, impaired bone microstructure, weakened bone strength and susceptibility to fracture. In China, the prevention and treatment of osteoporosis is faced with a high disease prevalence rate but low awareness, diagnosis and treatment rates. Bone resorption inhibitors and bone formation promoters often dominate osteoporosis treatment. Although conventional drugs can alleviate symptoms and reduce fracture risk, they often come with musculoskeletal, allergic and digestive side effects. Natural traditional Chinese medicine (TCM) products, known for their multi-targeting, high safety, efficacy and low cost, have been widely used in the treatment and prevention of osteoporosis in recent years and have gradually been recognised by many experts locally and abroad. This paper summarises recent research progress on natural TCM products in preventing and treating osteoporosis and provides a theoretical and experimental basis for the development of new drugs and the improvement of osteoporosis management.
Collapse
Affiliation(s)
- Bo Liu
- Department of Orthopedics, The Fourth People's Hospital of Shenyang, 20 Huanghe Dajie, Huanggu District, Shenyang, 110031, Liaoning, China
| | - Xue Mao
- Department of Orthopedics, Fushun Hospital of Chinese Medicine, No.9, East 6 th Road, Zhanqian Street, Xinfu District, Fushun, 113000, Liaoning, China
| | - Zhe-Jian-Yi Gao
- Department of Orthopedics, Fushun Hospital of Chinese Medicine, No.9, East 6 th Road, Zhanqian Street, Xinfu District, Fushun, 113000, Liaoning, China.
| | - Huan Wang
- Department of Orthopedics, Liaoning University of Traditional Chinese Medicine, 79 Chongshan Road, Huanggu District, Shenyang, 110847, Liaoning, China.
| |
Collapse
|
4
|
Wen R, Huang R, Xu K, Yi X. Insights into the role of histone lysine demethylases in bone homeostasis and skeletal diseases: A review. Int J Biol Macromol 2025; 306:141807. [PMID: 40054804 DOI: 10.1016/j.ijbiomac.2025.141807] [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: 02/14/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 05/11/2025]
Abstract
Histone lysine demethylases (KDMs), as important epigenetic regulators, are involved in various biological processes such as energy metabolism, apoptosis, and autophagy. Recent research shows that KDMs activate or silence downstream target genes by removing lysine residues from histone tails, and participate in the regulation of bone marrow mesenchymal stem cells (BM-MSCs), osteoblasts (OB), osteoclasts (OC), chondrocytes and other skeletal cell development, differentiation and formation. Moreover, several members of the KDM family affect the occurrence and development of bone diseases such as osteoporosis (OP), osteoarthritis (OA), osteosarcoma (OS), by regulating target genes. Specific regulation mechanisms of KDMs suggest new strategies for bone disease treatment and prevention. Despite the unique function and importance of KDMs in the skeletal system, previous studies have never systematically summarized their specific role, molecular mechanism, and clinical treatment in bone physiology and pathology. Therefore, this review summarises the expression pattern, intracellular signal transduction, and mechanism of action of the KDM family in several bone physiological and pathological conditions, aiming to highlight the important role of KDMs in bone diseases and provide a reference for the future treatment of bone diseases.
Collapse
Affiliation(s)
- Ruiming Wen
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Ruiqi Huang
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China; School of Physical Education, Liaoning Normal University, Dalian, Liaoning, China
| | - Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Xuejie Yi
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China.
| |
Collapse
|
5
|
Triwardhani A, Alida A, Winoto ER, Pramusita A, Putranti NAR, Ariadi KS, Pribadi OB, Anwar AA, Purnamasari AE, Mappananrang RA, Situmorang PC, Riawan W, Noor TNEBTA, Nugraha AP, Nugraha AP. Moringa oleifera L. Nanosuspension Extract Administration Affects Heat Shock Protein-10 and -70 under Orthodontics Mechanical Force In Vivo. Eur J Dent 2025; 19:523-530. [PMID: 39788532 PMCID: PMC12020578 DOI: 10.1055/s-0044-1791937] [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: 01/12/2025] Open
Abstract
OBJECTIVE The mechanical stimulation known as orthodontic mechanical force (OMF) causes biological reactions in orthodontic tooth movement (OTM). Heat shock protein-70 (HSP-70) needs pro-inflammatory cytokines to trigger bone resorption in OTM; nevertheless, heat shock protein-10 (HSP-10), a "Alarmin" cytokine, should control these pro-inflammatory cytokines to get the best alveolar bone remodeling (ABR). Moringa oleifera L. nanosuspension extract (MONE) has anti-inflammatory, antioxidant, and ABR-stimulating properties. The aim of the study was to examine in vivo HSP-10 and HSP-70 expressions under OMF following MONE application in Wistar rats (Rattus norvegicus). MATERIAL AND METHODS A total of 36 Wistar rats (R. norvegicus) were split up into eight groups: one for treatment (OMF + MONE) and one for control (OMF + MONE administration for days 1, 7, 14, and 21). By employing nickel-titanium coil springs and using 10 g of light force per millimeter to implant the orthodontic device, the OMF was completed. According to the day of observation, all of the samples were sacrificed. To perform an immunohistochemistry investigation, the premaxilla of the sample was isolated. Tukey's Honest Significant Different (HSD) test (p < 0.05) was performed after an Analysis of Variance (ANOVA) analysis of the data. RESULTS In both the OMF and MONE groups, HSP-70 peaked on day 14 and began to fall on day 21. HSP-10 peaked on day 21, but along with MONE, it also began to progressively decline on days 14 and 21, with significant differences (p < 0.05). CONCLUSION According to immunohistochemistry evidence, postadministration of MONE markedly elevated HSP-10 but lowered HSP-70 expression in the alveolar bone of Wistar rats under OMF.
Collapse
Affiliation(s)
- Ari Triwardhani
- Department of Orthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Alida Alida
- Department of Orthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Ervina Restiwulan Winoto
- Department of Orthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Adya Pramusita
- Department of Orthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | - Kristian Satrio Ariadi
- Department of Orthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Okso Brillian Pribadi
- Department of Orthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| | | | | | | | - Putri Cahaya Situmorang
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan, Indonesia
| | - Wibi Riawan
- Department of Molecular Biochemistry, Faculty of Medicine, Universitas Brawijaya, Malang, East Java, Indonesia
| | | | | | - Alexander Patera Nugraha
- Department of Orthodontic, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia
| |
Collapse
|
6
|
Jia Y, Qin Y, Yuan FL, Shen JH. Macrophage-to-Myofibroblast Transition Contributes to Cutaneous Scarring Formation Through the TGF-β/Smad3 Signaling Pathways. Cell Biol Int 2025; 49:494-507. [PMID: 39949174 DOI: 10.1002/cbin.70002] [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: 06/07/2024] [Revised: 01/11/2025] [Accepted: 02/02/2025] [Indexed: 04/15/2025]
Abstract
Cutaneous scarring typically arises after surgery, trauma, and infection, occurring when normal skin tissue is replaced by fibrous tissue during the healing process. Myofibroblasts have been identified as a significant contributor to this scarring. While the differentiation of fibroblasts into myofibroblasts is well-recognized as essential for wound healing and tissue repair, the mechanisms underlying the macrophage-myofibroblast transition (MMT) remain largely unexplored. This study aimed to investigate the role and potential mechanisms of MMT in cutaneous scarring. In specimens of hypertrophic scars, keloid and scleroderma, we confirmed the coexistence of MMT markers CD68 and α-smooth muscle actin (α-SMA) in areas of skin fibrosis. Additionally, most MMT cells in human cutaneous scar co-expressed the M2-type macrophage marker CD206. Fate-mapping in Lyz2-Cre/Rosa26-tdTomato mice further demonstrated that the majority of myofibroblasts in cutaneous scars were derived from bone marrow macrophages. Furthermore, higher levels of TGF-β were released from scar fibroblasts, which contributed to MMT through the Smad3 pathways. In vivo studies inhibiting Smad3 reduced MMT and scarring. Macrophage depletion with clodronate liposomes also reduced cutaneous scar formation. Our findings indicate that MMT plays a pivotal role in cutaneous scarring through the TGF-β/Smad3 pathways. Consequently, inhibiting MMT may be a novel strategy for the treatment of cutaneous scarring.
Collapse
Affiliation(s)
- Yuan Jia
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
- The 82nd Hospital of the Chinese People's Liberation Army, Huaian, China
| | - Yi Qin
- Xuzhou Medical University, Xuzhou, China
| | - Feng-Lai Yuan
- Institute of Integrated Chinese and Western Medicine, Affiliated to Jiangnan University, Wuxi, China
| | - Jie-Hong Shen
- Affiliated Hospital of Jiangnan University, Wuxi, China
| |
Collapse
|
7
|
Liu H, Zhang X, Ge X, Hsu C, Wang Y, Chen S, Yan X, Xu R, Ma J, Guo S. Optineurin Cooperates With NRF2 to Regulate Tooth Root Morphogenesis by Controlling Mitochondrial Dynamics and Apoptosis. Cell Prolif 2025; 58:e13799. [PMID: 39762159 PMCID: PMC12099217 DOI: 10.1111/cpr.13799] [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: 04/23/2024] [Revised: 11/18/2024] [Accepted: 12/18/2024] [Indexed: 05/24/2025] Open
Abstract
Tooth root development is a complex process essential for tooth function, yet the role of root dentin development in tooth morphogenesis is not fully understood. Optineurin (OPTN), linked to bone disorders like Paget's disease of bone (PDB), may affect tooth root development. In this study, we used single-cell sequencing of embryonic day 16.5 (E16.5), postnatal day 1 (P1), and P7 mouse teeth, as well as embryonic and adult human teeth, to show that OPTN is vital for odontoblastic differentiation. In Optn-/- mice, we observed short root deformities and defective dentin, with impaired apical papilla differentiation and increased apoptosis. In vitro OPTN downregulation in stem cells of the apical papilla (SCAPs) exacerbated apoptosis and hindered odontoblastic differentiation. RNA-seq analysis revealed significant differences in mitochondrial dynamics between control and OPTN knockout SCAPs. We discovered that OPTN influences mitochondrial dynamics primarily by promoting fission, leading to odontoblastic differentiation and mineralisation. Mechanistically, OPTN cooperates with NRF2 to regulate mitochondrial fission via DRP1 phosphorylation and affects the transcription of BCL2. Rescue experiments using an activator of NRF2 in ex vivo organ cultures and local gingival injection experiments confirmed these findings. Therefore, we concluded that OPTN, interacting with NRF2, acts as a key regulator of SCAPs mitochondrial dynamics, mineralisation and apoptosis during tooth development. These findings provide fresh insights into the mechanisms underlying tooth root development.
Collapse
Affiliation(s)
- Haojie Liu
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- State Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral Diseases (Nanjing Medical University)NanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjingChina
| | - Xinyu Zhang
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- State Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral Diseases (Nanjing Medical University)NanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjingChina
| | - Xiao Ge
- State Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral Diseases (Nanjing Medical University)NanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjingChina
| | - ChingCho Hsu
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
| | - Yan Wang
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- State Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral Diseases (Nanjing Medical University)NanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjingChina
| | - Simai Chen
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- State Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral Diseases (Nanjing Medical University)NanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjingChina
| | - Xingzhi Yan
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- State Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral Diseases (Nanjing Medical University)NanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjingChina
| | - Rongyao Xu
- State Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral Diseases (Nanjing Medical University)NanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjingChina
| | - Junqing Ma
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- State Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral Diseases (Nanjing Medical University)NanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjingChina
| | - Shuyu Guo
- Department of OrthodonticsThe Affiliated Stomatological Hospital of Nanjing Medical UniversityNanjingChina
- State Key Laboratory Cultivation Base of ResearchPrevention and Treatment for Oral Diseases (Nanjing Medical University)NanjingChina
- Jiangsu Province Engineering Research Center of Stomatological Translational MedicineNanjingChina
| |
Collapse
|
8
|
Wen R, Huang R, Yang M, Yang J, Yi X. Regulation of protein arginine methyltransferase in osteoporosis: a narrative review. Front Cell Dev Biol 2025; 13:1453624. [PMID: 40342926 PMCID: PMC12058719 DOI: 10.3389/fcell.2025.1453624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 04/14/2025] [Indexed: 05/11/2025] Open
Abstract
Osteoporosis (OP), a systemic bone disease characterised by increased bone fragility and susceptibility to fracture, is mainly caused by a decline in bone mineral density (BMD) and quality caused by an imbalance between bone formation and resorption. Protein arginine methyltransferases (PRMTs) are epigenetic factors and post-translational modification (PTM) enzymes participating in various biological processes, including mRNA splicing, DNA damage repair, transcriptional regulation, and cell signalling. They act by catalysing the transfer and modification of arginine residues and, thus, have become therapeutic targets for OP. In-depth studies have found that these enzymes also play key roles in bone matrix protein metabolism, skeletal cell proliferation and differentiation, and signal pathway regulation to regulate bone formation, bone resorption balance, or both and jointly maintain bone health and stability. However, the expression changes and mechanisms of action of multiple members of the PRMT family differ in OP. Therefore, this paper discusses the biological functions, mechanisms of action, and influencing factors of PRMTs in OP, which is expected to provide a new understanding of the pathogenesis of OP. Furthermore, we present theoretical support for the development of more precise and effective treatment strategies as well as for further study of the molecular mechanisms of PRMTs.
Collapse
Affiliation(s)
| | | | | | | | - Xuejie Yi
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| |
Collapse
|
9
|
Naidu P, Das M, Hansda S, Prateeksha P, Howlader MSI, Siraj MA, Das H. Mechanisms of Ellagic Acid (EA)-Mediated Osteogenic Differentiation of Human Dental Pulp-Derived Stem Cells. ACS OMEGA 2025; 10:15229-15242. [PMID: 40290905 PMCID: PMC12019503 DOI: 10.1021/acsomega.4c10642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/30/2025]
Abstract
Ellagic acid (EA) is a potent antioxidant that reduces oxidative stress and promotes differentiation. By lowering the harmful levels of reactive oxygen species (ROS), EA fosters an environment conducive to the osteoblastic differentiation (OB) of stem cells. In addition, it promotes autophagy and mitophagy, which are vital for promoting differentiation. Effective autophagic activity recycles damaged organelles and proteins, meeting the energy required during differentiation and shielding from apoptosis. However, molecular mechanisms underlying the osteogenic differentiation of mesenchymal stem cells remain inadequately explored. Therefore, the current study aims to define the regulatory role of EA during the OB of dental pulp-derived stem cells (DPSC) and to study how autophagy and mitophagy are being modulated during this differentiation process. Herein, we showed that the expression level of osteoblast-specific markers, autophagy, and mitophagy-associated markers was significantly elevated during EA-mediated OB differentiation of DPSC. Moreover, we found that the EA induced the osteoblastic-specific markers through canonical BMP2 pathway molecules, reduced ROS in both basal and activated states, and induced autophagy and mitophagy molecules along with enhanced mitochondrial functions. Cell cycle analysis revealed that the G1 phase was arrested via phosphorylation of γ-H2AX, ATM, and CHK2 proteins. Furthermore, in silico analysis revealed that EA strongly binds with osteonectin, a crucial noncollagen protein involved in bone remodeling, and confirmed by Western blot analysis. These results support that EA could be a promising natural compound for bone repair and regeneration applications.
Collapse
Affiliation(s)
- Prathyusha Naidu
- Department
of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Manjusri Das
- Department
of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Surajit Hansda
- Department
of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Prateeksha Prateeksha
- Department
of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Md Sariful Islam Howlader
- Department
of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Md Afjalus Siraj
- Department
of Therapeutic Radiology, Yale School of Medicine, Yale University, New Haven, Connecticut 06520, United States
| | - Hiranmoy Das
- Department
of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| |
Collapse
|
10
|
Mohar NP, Langland CJ, Darr Z, Viles J, Moore SA, Darbro BW, Wallrath LL. A genetic variant in SMAD7 acts as a modifier of LMNA-associated muscular dystrophy, implicating SMAD signaling as a therapeutic target. SCIENCE ADVANCES 2025; 11:eads7903. [PMID: 40249815 PMCID: PMC12007578 DOI: 10.1126/sciadv.ads7903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 03/12/2025] [Indexed: 04/20/2025]
Abstract
Mutations in LMNA cause multiple types of muscular dystrophy (LMNA-MD). The symptoms of LMNA-MD are highly variable and sensitive to genetic background. To identify genetic contributions to this phenotypic variability, we performed whole-genome sequencing on four siblings possessing the same LMNA mutation with differing degrees of skeletal muscle disease severity. We identified a variant in SMAD7 that segregated with severe muscle disease. To functionally test the SMAD7 variant, we generated a Drosophila model possessing the LMNA mutation and the SMAD7 variant in the orthologous fly genes. The SMAD7 variant increased SMAD signaling and enhanced muscle defects caused by the mutant lamin. Conversely, overexpression of wild-type SMAD7 rescued muscle function. These findings were extended to humans by showing that SMAD signaling is increased in muscle biopsy tissue from individuals with LMNA-MD compared to age-matched controls. Collectively, our findings support SMAD7 as the first functionally tested genetic modifier for LMNA-MD and suggest components of the SMAD pathway as therapeutic targets.
Collapse
Affiliation(s)
- Nathaniel P. Mohar
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Christopher J. Langland
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Zachary Darr
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Jill Viles
- Independent researcher, Gowrie, Iowa, USA
| | - Steven A. Moore
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Benjamin W. Darbro
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Lori L. Wallrath
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Biochemistry and Molecular Biology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
11
|
Dalle Carbonare L, Cominacini M, Trabetti E, Bombieri C, Pessoa J, Romanelli MG, Valenti MT. The bone microenvironment: new insights into the role of stem cells and cell communication in bone regeneration. Stem Cell Res Ther 2025; 16:169. [PMID: 40221779 PMCID: PMC11993959 DOI: 10.1186/s13287-025-04288-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Mesenchymal stem cells (MSCs) play a crucial role in bone formation and remodeling. Intrinsic genetic factors and extrinsic environmental cues regulate their differentiation into osteoblasts. Within the bone microenvironment, a complex network of biochemical and biomechanical signals orchestrates bone homeostasis and regeneration. In addition, the crosstalk among MSCs, immune cells, and neighboring cells-mediated by extracellular vesicles and non-coding RNAs (such as circular RNAs and micro RNAs) -profoundly influences osteogenic differentiation and bone remodeling. Recent studies have explored specific signaling pathways that contribute to effective bone regeneration, highlighting the potential of manipulating the bone microenvironment to enhance MSC functionality. The integration of advanced biomaterials, gene editing techniques, and controlled delivery systems is paving the way for more targeted and efficient regenerative therapies. Furthermore, artificial intelligence could improve bone tissue engineering, optimize biomaterial design, and enable personalized treatment strategies. This review explores the latest advancements in bone regeneration, emphasizing the intricate interplay among stem cells, immune cells, and signaling molecules. By providing a comprehensive overview of these mechanisms and their clinical implications, we aim to shed light on future research directions in this rapidly evolving field.
Collapse
Affiliation(s)
- L Dalle Carbonare
- Department of Engineering for the Innovation Medicine, University of Verona, 37100, Verona, Italy
| | - M Cominacini
- Department of Engineering for the Innovation Medicine, University of Verona, 37100, Verona, Italy
| | - E Trabetti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy
| | - C Bombieri
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy
| | - J Pessoa
- Department of Medical Sciences and Institute of Biomedicine-Ibimed, University of Aveiro, 3810 - 193, Aveiro, Portugal
| | - M G Romanelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy
| | - M T Valenti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy.
| |
Collapse
|
12
|
Li S, Cai X, Guo J, Li X, Li W, Liu Y, Qi M. Cell communication and relevant signaling pathways in osteogenesis-angiogenesis coupling. Bone Res 2025; 13:45. [PMID: 40195313 PMCID: PMC11977258 DOI: 10.1038/s41413-025-00417-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 02/18/2025] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
Osteogenesis is the process of bone formation mediated by the osteoblasts, participating in various bone-related physiological processes including bone development, bone homeostasis and fracture healing. It exhibits temporal and spatial interconnectivity with angiogenesis, constructed by multiple forms of cell communication occurring between bone and vascular endothelial cells. Molecular regulation among different cell types is crucial for coordinating osteogenesis and angiogenesis to facilitate bone remodeling, fracture healing, and other bone-related processes. The transmission of signaling molecules and the activation of their corresponding signal pathways are indispensable for various forms of cell communication. This communication acts as a "bridge" in coupling osteogenesis to angiogenesis. This article reviews the modes and processes of cell communication in osteogenesis-angiogenesis coupling over the past decade, mainly focusing on interactions among bone-related cells and vascular endothelial cells to provide insights into the mechanism of cell communication of osteogenesis-angiogenesis coupling in different bone-related contexts. Moreover, clinical relevance and applications are also introduced in this review.
Collapse
Affiliation(s)
- Shuqing Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Xinjia Cai
- Central Laboratory, Peking University School and Hospital for 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 & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Jiahe Guo
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Xiaolu Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Wen Li
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China
| | - Yan Liu
- Central Laboratory, Peking University School and Hospital for 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 & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| | - Mengchun Qi
- Department of Oral & Maxillofacial Surgery, College of Stomatology, North China University of Science and Technology, Tangshan, Hebei, China.
| |
Collapse
|
13
|
Qian CZ, Wu JJ, Zhang Z. Comments on "The Efficacy and Safety of Botulinum Toxin Type A in Prevention of Hypertrophic Scars After Epicanthoplasty: A Split-Face Double-Blinded Randomized Controlled Trial". Aesthetic Plast Surg 2025:10.1007/s00266-025-04847-3. [PMID: 40183927 DOI: 10.1007/s00266-025-04847-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025]
Affiliation(s)
- Chang-Zhong Qian
- School of Clinical Medicine, Jiangsu University, Zhenjiang, 212000, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214041, Jiangsu, China
| | - Zhenyu Zhang
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214041, Jiangsu, China.
| |
Collapse
|
14
|
Wang Z, Zhu P, Li H, Ye B, Luo Q, Cheng J, Cai Y. Sodium Hyaluronate-PDGF Repairs Cartilage and Subchondral Bone Microenvironment via HIF-1α-VEGF-Notch and SDF-1-CXCR4 Inhibition in Osteoarthritis. J Cell Mol Med 2025; 29:e70515. [PMID: 40159624 PMCID: PMC11955409 DOI: 10.1111/jcmm.70515] [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: 11/05/2024] [Revised: 03/09/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
Chronic degenerative changes in cartilage and subchondral bone that lead to instability of the cartilage microenvironment are essential for the development of osteoarthritis (OA) in the old. Synchronous repair of cartilage and subchondral bone may be a key strategy for OA treatment. PDGF-BB effectively promoted chondrocyte regeneration and angiogenesis. However, the mechanisms by which PDGF-BB affects subchondral bone and the delivery of PDGF-BB to the joint cavity need to be further explored. In this study, we used sodium hyaluronate to deliver PDGF-BB (SH-PDGF) to the joint space and aimed to determine the mechanisms of SH-PDGF in repairing cartilage and subchondral bone and stabilising the cartilage microenvironment. In this research, we determined the pharmacokinetics of PDGF-BB and SH-PDGF in cartilage. Moreover, we investigated the effects of PDGF-BB and SH-PDGF on cartilage and the subchondral bone microenvironment by identifying changes in the HIF-VEGF-Notch axis and SDF-1-CXCR4 axis in an OA rat model. The results showed that PDGF-BB increased cell viability, decreased HIF-1α levels, inhibited inflammation and improved matrix metabolism in osteoarthritic chondrocytes under hyperoxic or hypoxic conditions. We also found that PDGF-BB and SH-PDGF showed similar effects on repairing cartilage and subchondral bone simultaneously. However, SH-PDGF had some advantages over PDGF-BB in prolonging the injection interval and decreasing the injection time. These protective effects were mediated by the inhibition of both the HIF-1α-VEGF-Notch axis and the SDF-1-CXCR4 axis. The underlying mechanisms include the inhibition of HIF-1α-VEGF-Notch-mediated vessel invasion and SDF-1-CXCR4 axis-mediated crosstalk between cartilage and subchondral tissue.
Collapse
Affiliation(s)
- Zhengchao Wang
- Department of Sports MedicineWuhan Fourth HospitalWuhanChina
- Hubei Provincial Sports Medicine CenterWuhanChina
| | - Pengfei Zhu
- Hubei Provincial Sports Medicine CenterWuhanChina
- Department of CardiovascularWuhan Fourth HospitalWuhanChina
| | - Hongmei Li
- Zibo First Hospital, Zibo Prevention and Treatment Hospital for Occupation DiseasesZiboChina
| | - Bo Ye
- Hubei Provincial Sports Medicine CenterWuhanChina
- Department of RehabilitationWuhan Fourth HospitalWuhanChina
| | - Qiong Luo
- Hubei Provincial Sports Medicine CenterWuhanChina
- Department of RehabilitationWuhan Fourth HospitalWuhanChina
| | - Jiangxia Cheng
- Hubei Provincial Sports Medicine CenterWuhanChina
- Department of AnesthesiologyWuhan Fourth HospitalWuhanChina
| | - Yu Cai
- Hubei Provincial Sports Medicine CenterWuhanChina
- Department of RehabilitationWuhan Fourth HospitalWuhanChina
| |
Collapse
|
15
|
Zulkifli A, Kong P, Hrk S, Yasin NF, Nam HY, Kamarul T. Hypoxia-induced HIF-1α accumulation promotes superior tenogenic differentiation potential of human adipose-derived mesenchymal stromal cells. Biotech Histochem 2025; 100:100-118. [PMID: 40135543 DOI: 10.1080/10520295.2025.2482934] [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: 03/27/2025] Open
Abstract
Tendon injuries remains a challenge to treat owing to its poor intrinsic reparative ability. It is hypothesised that hypoxic conditioning of mesenchymal stem cells (MSC) through the activation of hypoxia-inducible factor-1 alpha (HIF-1α), may enhance tendon repair process by promoting cellular proliferation and tenogenic differentiation. To demonstrate this, a study using roxadustat, a specific hypoxia mimetic mediator and HIF-1α inducer was conducted on adipose-derived mesenchymal stromal cells (AD-MSCs). Cellular morphology, proliferation rates, tenogenic protein and gene expression levels in untreated AD-MSCs (Group 1), roxadustat pre-conditioned AD-MSCs (Group 2), AD-MSCs subjected to CAY10585 (Group 3), roxadustat pre-conditioned AD-MSCs with CAY10585 (Group 4) and untreated primary tenocytes (Group 5) were evaluated. MSCs pre-conditioned with 12.5µM roxadustat for 24 hours showed the highest expression of HIF-1α without affecting the proliferation rates of AD-MSCs. However, significant reduction of HIF-1α levels was observed when the cells were treated with 3.5µM CAY10585. Roxadustat significantly up-regulated collagen I and III expressions by 6.6 and 6.3-fold respectively. HIF-1α promoted Scleraxis, Tenascin-C and Collagen III expressions, resulting in an increase of 6, 7, and 3 folds respectively. Conversely, using CAY10585 reduced these expressions to 3, 2 and 1 folds respectively. These trends were observed in the gene expression levels across Groups 1 to 4. However, the expression of these genes in Group 2 was significantly lower as compared to Group 5. Conclusion: HIF-1α accumulation promotes superior cell proliferation and tenogenic differentiation of AD-MSCs, indicating that roxadustat may be a potential therapeutic mediator in tendon repair strategies.
Collapse
Affiliation(s)
- Amirah Zulkifli
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, Universiti Malaya Malaya, Kuala Lumpur, Malaysia
| | - Peggy Kong
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, Universiti Malaya Malaya, Kuala Lumpur, Malaysia
| | - Shaliny Hrk
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, Universiti Malaya Malaya, Kuala Lumpur, Malaysia
| | - Nor Faissal Yasin
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, Universiti Malaya Malaya, Kuala Lumpur, Malaysia
| | - Hui Yin Nam
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, Universiti Malaya Malaya, Kuala Lumpur, Malaysia
- Nanotechnology & Catalysis Research Centre (NANOCAT), Institute for Advanced Studies, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Tunku Kamarul
- Tissue Engineering Group, Department of Orthopaedic Surgery (NOCERAL), Faculty of Medicine, Universiti Malaya Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
16
|
Zhu L, Liu Y, Sun Y, Che Z, Li Y, Liu T, Li X, Yang C, Huang L. Sustained slow-release TGF-β3 in a three-dimensional-printed titanium microporous scaffold composite system promotes ligament-to-bone healing. Mater Today Bio 2025; 31:101549. [PMID: 40182658 PMCID: PMC11966733 DOI: 10.1016/j.mtbio.2025.101549] [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: 05/28/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 04/05/2025] Open
Abstract
The treatment of tendon/ligament-to-bone injury is a long-standing research challenge in orthopedics and bone tissue engineering. Orderly healing of the fibrocartilage layer and mineralized bone layer is crucial for treating tendon-bone interface injuries. We designed a three-dimensional printed porous titanium scaffold composite system with thermosensitive collagen hydrogel loaded with transforming growth factor β3 (TGF-β3), formulated for the sustained slow release of TGF-β3 at a constant rate. In vitro, the composite system exhibited good biocompatibility and was beneficial for the adhesion and proliferation of bone marrow mesenchymal stem cells (BMSCs), which showed high growth activity. Moreover, the composite system promoted the differentiation of BMSCs via osteogenesis and chondrogenesis. In vivo, the composite system provided active substances at the injured site, promoting the repair of the fibrocartilage layer and of the mineralized bone layer at the interface between the ligament and bone. Micro-CT results demonstrated that the complex promotes the osseointegration of titanium scaffolds in bone defects. Hard tissue sections showed that the new bone, ligament, and the titanium alloy scaffold system formed a closely integrated whole; the composite system provided suitable attachment points for ligament growth. Additionally, the biomechanical strength of the tendon interface improved to some extent. Our results indicate that the composite system has potential as a bioactive implant interface for repairing ligament and bone injuries.
Collapse
Affiliation(s)
- Liwei Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Yuzhe Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Yifu Sun
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Zhenjia Che
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Youbin Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Tengyue Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Xudong Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Chengzhe Yang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| | - Lanfeng Huang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, 130041, PR China
| |
Collapse
|
17
|
De Pace R, Iaquinta MR, Benkhalqui A, D'Agostino A, Trevisiol L, Nocini R, Mazziotta C, Rotondo JC, Bononi I, Tognon M, Martini F, Mazzoni E. Revolutionizing bone healing: the role of 3D models. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:7. [PMID: 40113735 PMCID: PMC11926310 DOI: 10.1186/s13619-025-00225-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/31/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025]
Abstract
The increasing incidence of bone diseases has driven research towards Bone Tissue Engineering (BTE), an innovative discipline that uses biomaterials to develop three-dimensional (3D) scaffolds capable of mimicking the natural environment of bone tissue. Traditional approaches relying on two-dimensional (2D) models have exhibited significant limitations in simulating cellular interactions and the complexity of the bone microenvironment. In response to these challenges, 3D models such as organoids and cellular spheroids have emerged as effective tools for studying bone regeneration. Adult mesenchymal stem cells have proven crucial in this context, as they can differentiate into osteoblasts and contribute to bone tissue repair. Furthermore, the integration of composite biomaterials has shown substantial potential in enhancing bone healing. Advanced technologies like microfluidics offer additional opportunities to create controlled environments for cell culture, facilitating more detailed studies on bone regeneration. These advancements represent a fundamental step forward in the treatment of bone pathologies and the promotion of skeletal health. In this review, we report on the evolution of in vitro culture models applied to the study of bone healing/regrowth, starting from 2 to 3D cultures and microfluids. The different methodologies of in vitro model generation, cells and biomaterials are presented and discussed.
Collapse
Affiliation(s)
- Raffaella De Pace
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, 44121, Italy
| | - Maria Rosa Iaquinta
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- University Center for Studies On Gender Medicine, University of Ferrara, Ferrara, Italy
| | - Assia Benkhalqui
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- Department of Surgery, University of Verona, Verona, Italy
| | | | - Lorenzo Trevisiol
- Centre for Medical Sciences (CISMed), University of Trento, Trento, Italy
- Unit of Maxillofacial Surgery, Santa Chiara Regional Hospital, APSS, Trento, Italy
| | | | - Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- University Center for Studies On Gender Medicine, University of Ferrara, Ferrara, Italy
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- University Center for Studies On Gender Medicine, University of Ferrara, Ferrara, Italy
| | - Ilaria Bononi
- Centralized Laboratory of Pre-Clinical Research, University of Ferrara, Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
- University Center for Studies On Gender Medicine, University of Ferrara, Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Elisa Mazzoni
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, 44121, Italy.
| |
Collapse
|
18
|
Mehreen A, Faisal M, Zulfiqar B, Hays D, Dhananjaya K, Yaseen F, Liang Y. Connecting Bone Remodeling and Regeneration: Unraveling Hormones and Signaling Pathways. BIOLOGY 2025; 14:274. [PMID: 40136530 PMCID: PMC11939909 DOI: 10.3390/biology14030274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/02/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025]
Abstract
Recent advancements in tissue engineering and stem cell science have positioned bone disease treatment as a promising frontier in regenerative medicine. This review explores the hormonal and signaling pathways critical to bone regeneration, with a focus on their clinical relevance. Key endocrine factors, including thyroid hormones (T3 and T4), insulin-like growth factor 1 (IGF-1), bone morphogenetic proteins (BMPs), parathyroid hormone (PTH), calcitonin, and fibroblast growth factor 23 (FGF23), play pivotal roles in bone remodeling by regulating osteoblast activity, bone resorption, and mineralization. These factors primarily act through the Wnt/β-catenin, BMP, and FGF signaling pathways, which govern bone repair and regeneration. While animal models, such as axolotls, zebrafish, and Xenopus laevis, provide valuable findings about these mechanisms, translating these findings into human applications presents challenges. This review underscores the therapeutic potential of modulating these hormonal networks to enhance bone regeneration while cautioning against possible adverse effects, such as uncontrolled tissue proliferation or metabolic imbalances. By integrating knowledge from regenerative models, this work provides a foundation for optimizing hormone-based therapies for clinical applications in bone repair and disease treatment.
Collapse
Affiliation(s)
- Afshan Mehreen
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (A.M.); (M.F.); (D.H.); (K.D.); (F.Y.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Muhammad Faisal
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (A.M.); (M.F.); (D.H.); (K.D.); (F.Y.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Bilal Zulfiqar
- Discovery Biology, Griffith University, Nathan, QLD 4111, Australia;
| | - Deli Hays
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (A.M.); (M.F.); (D.H.); (K.D.); (F.Y.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Kavishka Dhananjaya
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (A.M.); (M.F.); (D.H.); (K.D.); (F.Y.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Faiza Yaseen
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (A.M.); (M.F.); (D.H.); (K.D.); (F.Y.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Yujun Liang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education), Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (A.M.); (M.F.); (D.H.); (K.D.); (F.Y.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
19
|
Ward T, Morton SU, Venturini G, Tai W, Jang MY, Gorham J, Delaughter D, Wasson LK, Khazal Z, Homsy J, Gelb BD, Chung WK, Bruneau BG, Brueckner M, Tristani-Firouzi M, DePalma SR, Seidman C, Seidman JG. Modeling SMAD2 Mutations in Induced Pluripotent Stem Cells Provides Insights Into Cardiovascular Disease Pathogenesis. J Am Heart Assoc 2025; 14:e036860. [PMID: 40028843 DOI: 10.1161/jaha.124.036860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/17/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND SMAD2 is a coregulator that binds a variety of transcription factors in human development. Heterozygous SMAD2 loss-of-function and missense variants are identified in patients with congenital heart disease (CHD) or arterial aneurysms. Mechanisms that cause distinct cardiovascular phenotypes remain unknown. We aimed to define transcriptional and epigenetic effects of SMAD2 variants and their role in CHD. We also assessed the function of SMAD2 missense variants of uncertain significance. METHODS AND RESULTS Rare SMAD2 variants (minor allele frequency ≤10-5) were identified in exome sequencing of 11 336 participants with CHD. We constructed isogenic induced pluripotent stem cells with heterozygous or homozygous loss-of-function and missense SMAD2 variants identified in CHD probands. Wild-type and mutant induced pluripotent stem cells were analyzed using bulk RNA sequencing, chromatin accessibility (Assay for Transposase-Accessible Chromatin With Sequencing), and integrated with published SMAD2/3 chromatin immunoprecipitation data. Cardiomyocyte differentiation and contractility were evaluated. Thirty participants with CHD had heterozygous loss-of-function or missense SMAD2 variants. SMAD2 haploinsufficiency altered chromatin accessibility at promoters and dysregulated expression of 385 SMAD regulated genes, including 10 CHD-associated genes. Motifs enriched in differential Assay for Transposase-Accessible Chromatin peaks predicted that SMAD2 haploinsufficiency disrupts interactions with transcription factors NANOG (homeobox protein NANOG), ETS, TEAD3/4 (transcriptional enhanced associate domain 3/4), CREB1 (cAMP response element binding protein 1), and AP1 (activator protein 1). Compared with SMAD2-haploinsufficient cells, induced pluripotent stem cells with R114C or W274C variants exhibited distinct and shared chromatin accessibility and transcription factor binding changes. CONCLUSIONS SMAD2 haploinsufficiency disrupts transcription factor binding and chromatin interactions critical for cardiovascular development. Differences between the molecular consequences of loss-of-function and missense variants likely contribute to phenotypic heterogeneity. These findings indicate opportunities for molecular analyses to improve reclassification of SMAD2 variants of uncertain clinical significance.
Collapse
Affiliation(s)
- Tarsha Ward
- Department of Genetics Harvard Medical School Boston MA USA
| | - Sarah U Morton
- Department of Genetics Harvard Medical School Boston MA USA
- Division of Newborn Medicine Boston Children's Hospital Boston MA USA
| | | | - Warren Tai
- Department of Genetics Harvard Medical School Boston MA USA
| | - Min Young Jang
- Department of Genetics Harvard Medical School Boston MA USA
| | - Joshua Gorham
- Department of Genetics Harvard Medical School Boston MA USA
| | - Dan Delaughter
- Department of Genetics Harvard Medical School Boston MA USA
| | | | - Zahra Khazal
- Department of Genetics Harvard Medical School Boston MA USA
| | - Jason Homsy
- Department of Genetics Harvard Medical School Boston MA USA
- Cardurion Pharmaceuticals, Inc. Burlington MA USA
| | - Bruce D Gelb
- Mindich Child Health and Development Institute and the Department of Pediatrics and Genetics and Genomic Sciences Icahn School of Medicine at Mount Sinai New York NY USA
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital Harvard Medical School Boston MA USA
| | - Benoit G Bruneau
- Gladstone Institutes San Francisco CA USA
- Roddenberry Center for Stem Cell Biology and Medicine at Gladstone San Francisco CA USA
- Department of Pediatrics, Cardiovascular Research Institute, Institute for Human Genetics, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research University of California San Francisco CA USA
| | - Martina Brueckner
- Department of Genetics and Pediatrics Yale University School of Medicine New Haven CT USA
| | - Martin Tristani-Firouzi
- Division of Pediatric Cardiology University of Utah and School of Medicine Salt Lake City UT USA
| | | | - Christine Seidman
- Department of Genetics Harvard Medical School Boston MA USA
- Department of Medicine Brigham and Women's Hospital Boston MA USA
- Howard Hughes Medical Institute Harvard Medical School Boston MA USA
| | - J G Seidman
- Department of Genetics Harvard Medical School Boston MA USA
| |
Collapse
|
20
|
Shen C, Han Y, Xiong H, Wang Y, Tan Z, Wei H, Ding Q, Ma L, Ding C, Zhao T. Multifunctional hydrogel scaffolds based on polysaccharides and polymer matrices promote bone repair: A review. Int J Biol Macromol 2025; 294:139418. [PMID: 39765302 DOI: 10.1016/j.ijbiomac.2024.139418] [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: 10/13/2024] [Revised: 12/19/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025]
Abstract
With the advancement of medical technology, the utilization of bioactive materials to promote bone repair has emerged as a significant research area. Hydrogels, as biomaterials, play a crucial role in bone tissue engineering. These hydrogels exhibit high biocompatibility, providing in vivo ecological conditions conducive to cell survival, and offer substantial advantages in facilitating bone repair. Different matrices of hydrogels serve distinct functions. In recent years, numerous researchers have developed a variety of novel hydrogel materials utilizing diverse matrices. These materials not only enhance the osteogenic induction capacity of hydrogels but also improve their efficacy as scaffolds in the treatment of complex bone defects, such as those resulting from trauma, tumor resection, or large bone defects due to infection. This article primarily analyzes the role of hydrogels that utilize polysaccharides and polymers as matrices in bone tissue repair, focusing on the creation of an optimal microenvironment to promote bone regeneration. These investigations deepen the understanding of the mechanisms underlying the action of hydrogels and establish a foundation for future advancements in the biomedical field.
Collapse
Affiliation(s)
- Chang Shen
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Yuanyuan Han
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Huan Xiong
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Yulai Wang
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Ziqi Tan
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Hewei Wei
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Qiteng Ding
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Lina Ma
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China.
| | - Chuanbo Ding
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China.
| | - Ting Zhao
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China.
| |
Collapse
|
21
|
Liu W, Xu Y, Hu W, Zhang L, Wang C, Wang F, Zai Z, Qian X, Peng X, Chen F. Succinate dehydrogenase mediated ROS production contributes to ASIC1a-induced chondrocyte pyroptosis in rheumatoid arthritis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167585. [PMID: 39586503 DOI: 10.1016/j.bbadis.2024.167585] [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: 09/04/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
Our previous study showed that acidic stimuli activate acid-sensitive ion channel 1a (ASIC1a), resulting in chondrocyte destruction associated with rheumatoid arthritis (RA). However, the exact underlying processes remain unclear. Recent evidence suggests that the production of reactive oxygen species (ROS) mediated by succinate dehydrogenase (SDH), contributes to chondrocyte damage. The objective of this study was to investigate the involvement of SDH in ASIC1a-induced chondrocyte destruction in RA and to explore the associated mechanisms both in vivo and in vitro. Our findings revealed that the cartilage of mice with collagen-induced arthritis (CIA) and acid-treated chondrocytes exhibited a substantial increase in SDH expression. Furthermore, SDH inhibition attenuates acidosis-induced pyroptosis in chondrocytes. Notably, ASIC1a activation through acid stimuli increases SDH activity and pyroptosis through the Ca2+/CaMKK2/AMPK pathway in chondrocytes. Mechanistically, SDH assembly factor 2 (SDHAF2) was identified as a key modulator of SDH activity induced by ASIC1a in acid-stressed chondrocytes. Moreover, the expression of SDH in CIA mouse chondrocytes decreased and the histological characteristics of ankle joint damage were reduced by the ASIC1a-particular blocker PcTx-1. Overall, these observations suggest that ASIC1a activation under acidic conditions increases SDH activity and modulates SDHAF2, thereby promoting chondrocyte pyroptosis through the Ca2+/CaMKK2/AMPK pathway.
Collapse
Affiliation(s)
- Wenqiang Liu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Yayun Xu
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Weirong Hu
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Longbiao Zhang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Cheng Wang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Fengshuo Wang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Zhuoyan Zai
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Xuewen Qian
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China
| | - Xiaoqing Peng
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China.
| | - Feihu Chen
- School of Pharmacy, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
22
|
Yuan B, Peng H, Wang Y, Li J, Zhang Y, Chen Z, Li K, Tu C, Zhang K, Zhu X, Shen B, Nie Y, Zhang X. Micro/Nanobiomimetic Iron-Based Scaffold Induces Vascularized Bone Regeneration To Repair Large Segmental Bone Defect in Load-Bearing Sites. ACS NANO 2025; 19:6840-6857. [PMID: 39933996 DOI: 10.1021/acsnano.4c11960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Biodegradable scaffolds, including metals, ceramics, and polymers, show great potential in bone tissue regeneration. However, current biodegradable scaffolds do not simultaneously possess suitable mechanical properties, biodegradability and osteoinductivity, which severely limits their clinical application for large segmental bone defect repair. Herein, we developed a biomimetic and hierarchically micro-nanoporous iron-based scaffold utilizing a synergistic approach combining 3-dimensional printing, surface dealloying treatment and electrochemical deposition. Compared to traditional periodic lattice structures, the biomimetic scaffold with a stochastic lattice structure promised superior stress transfer efficiency. Cell experiments revealed that the biomimetic scaffold notably enhanced osteogenesis and angiogenesis in vitro via EGFR-mediated Ras/Raf/MAPK signaling. Upon implantation in a rat femoral condyle defect model, the scaffold achieved a dynamic equilibrium between in vivo material degradation and bone formation. More importantly, the study conducted in a large animal model with an extended cycle of up to 1 year demonstrated that bionic iron-based scaffolds effectively facilitated the repair and functional reconstruction of large bone defects in load-bearing regions by inducing vascularized bone regeneration. This study not only introduces a potential solution for addressing critical-sized bone defects in load-bearing regions but also provides a viable approach for the design of other biomimetic biomaterials.
Collapse
Affiliation(s)
- Bo Yuan
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Huabei Peng
- College of Mechanical Engineering, Sichuan University, Chengdu 610065, P. R. China
| | - Yitian Wang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Jingming Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Yuqi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Zhikun Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Kang Li
- West China Biomedical Big Data Center, Sichuan University West China Hospital, Chengdu, Sichuan University, Chengdu 610041, P. R. China
| | - Chongqi Tu
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Kai Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
- Institute of Regulatory Science for Medical Device, Sichuan University, Chengdu 610064, P. R. China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| | - Bin Shen
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Yong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, Chengdu 610064, P. R. China
| |
Collapse
|
23
|
Zhan L, Zhou Y, Liu R, Sun R, Li Y, Tian Y, Fan B. Advances in growth factor-containing 3D printed scaffolds in orthopedics. Biomed Eng Online 2025; 24:14. [PMID: 39920740 PMCID: PMC11806769 DOI: 10.1186/s12938-025-01346-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 01/24/2025] [Indexed: 02/09/2025] Open
Abstract
Currently, bone tissue engineering is a research hotspot in the treatment of orthopedic diseases, and many problems in orthopedics can be solved through bone tissue engineering, which can be used to treat fractures, bone defects, arthritis, etc. More importantly, it can provide an alternative to traditional bone grafting and solve the problems of insufficient autologous bone grafting, poor histocompatibility of grafts, and insufficient induced bone regeneration. Growth factors are key factors in bone tissue engineering by promoting osteoblast proliferation and differentiation, which in turn increases the efficiency of osteogenesis and bone regeneration. 3D printing technology can provide carriers with better pore structure for growth factors to improve the stability of growth factors and precisely control their release. Studies have shown that 3D-printed scaffolds containing growth factors provide a better choice for personalized treatment, bone defect repair, and bone regeneration in orthopedics, which are important for the treatment of orthopedic diseases and have potential research value in orthopedic applications. This paper aims to summarize the research progress of 3D printed scaffolds containing growth factors in orthopedics in recent years and summarize the use of different growth factors in 3D scaffolds, including bone morphogenetic proteins, platelet-derived growth factors, transforming growth factors, vascular endothelial growth factors, etc. Optimization of material selection and the way of combining growth factors with scaffolds are also discussed.
Collapse
Affiliation(s)
- Longwen Zhan
- Orthopedic Centre-Spine Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, China
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Yigui Zhou
- Orthopedic Centre-Spine Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, China
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Ruitang Liu
- Orthopedic Centre-Spine Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, China
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Ruilong Sun
- Orthopedic Centre-Spine Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, China
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Yunfei Li
- Orthopedic Centre-Spine Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, China
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Yongzheng Tian
- Orthopedic Centre-Spine Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, China
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Bo Fan
- Orthopedic Centre-Spine Surgery, The 940 Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, 730050, China.
| |
Collapse
|
24
|
You JH, Kim NY, Choi YY, Choi HW, Chung BG. Dual-stimuli-responsive nanoparticles for the co-delivery of small molecules to promote neural differentiation of human iPSCs. NANOSCALE 2025; 17:2506-2519. [PMID: 39815767 DOI: 10.1039/d4nr04413d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
The differentiation of human induced pluripotent stem cells (hiPSCs) into neural progenitor cells (NPCs) is a promising approach for the treatment of neurodegenerative diseases and regenerative medicine. Dual-SMAD inhibition using small molecules has been identified as a key strategy for directing the differentiation of hiPSCs into NPCs by regulating specific cell signaling pathways. However, conventional culture methods are time-consuming and exhibit low differentiation efficiency in neural differentiation. Nanocarriers can address these obstacles as an efficient platform for the controlled release and accurate delivery of small molecules. In this paper, we developed calcium phosphate-coated mesoporous silica nanoparticles capable of delivering multiple small molecules, including LDN193189 as a bone morphogenetic protein (BMP) inhibitor and SB431542 as a transforming growth factor (TGF)-beta inhibitor, for direct differentiation of hiPSC-mediated NPCs. Our results demonstrated that this nanocarrier-mediated small molecule release system not only enhanced the in vitro formation of neural rosettes but also modulated the expression levels of key markers. In particular, it downregulated OCT4, a marker of pluripotency, while upregulating PAX6, a critical marker for the neuroectoderm. These findings suggest that this controlled small molecule release system holds significant potential for therapeutic applications in neural development and neurodegenerative diseases.
Collapse
Affiliation(s)
- Jeong Hyun You
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea.
| | - Na Yeon Kim
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea.
| | - Yoon Young Choi
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
| | - Hyung Woo Choi
- Department of Mechanical Engineering, Sogang University, Seoul 04107, Korea
| | - Bong Geun Chung
- Department of Biomedical Engineering, Sogang University, Seoul 04107, Korea.
- Institute of Integrated Biotechnology, Sogang University, Seoul 04107, Korea
- Department of Mechanical Engineering, Sogang University, Seoul 04107, Korea
- Institute of Smart Biosensor, Sogang University, Seoul 04107, Korea
| |
Collapse
|
25
|
Zhang Q, Yang Z, Ou X, Zhang M, Qin X, Wu G. The role of immunity in insulin resistance in patients with polycystic ovary syndrome. Front Endocrinol (Lausanne) 2025; 15:1464561. [PMID: 39911236 PMCID: PMC11797073 DOI: 10.3389/fendo.2024.1464561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 12/31/2024] [Indexed: 02/07/2025] Open
Abstract
Polycystic ovary syndrome (PCOS) is a prevalent disorder of the endocrine system with significant clinical implications, often leading to health complications related to adipose tissue accumulation, including obesity, insulin resistance (IR), metabolic syndrome, and type 2 diabetes mellitus. While the precise pathogenesis of PCOS remains unclear, it is now recognized that genetic, endocrine, and metabolic dysregulations all contribute significantly to its onset. The immunopathogenesis of PCOS has not been extensively explored, but there is growing speculation that immune system abnormalities may play a pivotal role. This chronic inflammatory state is exacerbated by factors such as obesity and hyperinsulinemia. Therefore, this review aims to elucidate the interplay between IR in PCOS patients, the controlled immune response orchestrated by immune cells and immunomodulatory molecules, and their interactions with adipocytes, hyperandrogenemia, chronic inflammation, and metabolic homeostasis.
Collapse
Affiliation(s)
- Qixuan Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhe Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangyang Ou
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mengying Zhang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangyu Qin
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gengxiang Wu
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
26
|
Xu H, Luo Y, An Y, Wu X. The mechanism of action of indole-3-propionic acid on bone metabolism. Food Funct 2025; 16:406-421. [PMID: 39764708 DOI: 10.1039/d4fo03783a] [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: 01/14/2025]
Abstract
Indole-3-propionic acid (IPA), a metabolite produced by gut microbiota through tryptophan metabolism, has recently been identified as playing a pivotal role in bone metabolism. IPA promotes osteoblast differentiation by upregulating mitochondrial transcription factor A (Tfam), contributing to increased bone density and supporting bone repair. Simultaneously, it inhibits the formation and activity of osteoclasts, reducing bone resorption, possibly through modulation of the nuclear factor-κB (NF-κB) pathway and downregulation of osteoclast-associated factors, thereby maintaining bone structural integrity. Additionally, IPA provides indirect protection to bone health by regulating host immune responses and inflammation via activation of receptors such as the Aryl hydrocarbon Receptor (AhR) and the Pregnane X Receptor (PXR). This review summarizes the roles and signaling pathways of IPA in bone metabolism and its impact on various bone metabolic disorders. Furthermore, we discuss the therapeutic potential and limitations of IPA in treating bone metabolic diseases, aiming to offer novel strategies for clinical management.
Collapse
Affiliation(s)
- Huimin Xu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yi An
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| | - Xi Wu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China.
| |
Collapse
|
27
|
Park HY, Kang MH, Lee G, Kim JW. Enhancement of skin regeneration through activation of TGF-β/SMAD signaling pathway by Panax ginseng meyer non-edible callus-derived extracellular vesicles. J Ginseng Res 2025; 49:34-41. [PMID: 39872281 PMCID: PMC11770231 DOI: 10.1016/j.jgr.2024.08.002] [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: 02/22/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 01/30/2025] Open
Abstract
Background This study aimed to investigate the effects of ginseng non-edible callus-derived extracellular vesicle (GNEV) on skin regeneration, particularly focusing on its impact on proliferation and migration in human dermal fibroblast (HDF). Methods GNEV was isolated from ginseng non-edible callus using sequential filtration and size exclusion chromatography (SEC). The extracellular vesicle was characterized using nanoparticle tracking analysis (NTA). HDF was treated with various concentrations of GNEV, and cell viability, proliferation, and migration were assessed using MTT and scratch wound healing assays. Gene expression related to collagen synthesis (TGF-β, SMAD-2, SMAD-3, COL1A1) was measured using RT-PCR. Results Treatment of HDF with GNEV resulted in a significant 2.5-fold increase in cell migration compared to the non-treated group. Furthermore, GNEV demonstrated the upregulation of collagen synthesis genes, specifically TGF-β, SMAD-2, SMAD-3, and COL1A1, by 41.7 %, 59.4 %, 60.2 %, and 21.8 %, respectively. These findings indicated that GNEV activates the TGF-β/SMAD signaling pathway, showcasing its potential to induce skin regeneration. Conclusions In conclusion, GNEV exhibits a notable ability to enhance skin regeneration through its stimulatory effects on cell migration and the upregulation of key collagen synthesis genes. The activation of the TGF-β/SMAD signaling pathway further suggests the potential of GNEV as a promising candidate for drug delivery systems in the fields of cosmetics and pharmaceuticals, opening avenues for further research and application in skincare and dermatology.
Collapse
Affiliation(s)
- Ha Young Park
- Department of Food Science, Sunmoon University, Chungcheongnam-do, Republic of Korea
| | - Min Ho Kang
- Department of Food Science, Sunmoon University, Chungcheongnam-do, Republic of Korea
| | - Guewha Lee
- Hu evergreen Pharm Inc., Incheon, Republic of Korea
| | - Jin Woo Kim
- Department of Food Science, Sunmoon University, Chungcheongnam-do, Republic of Korea
- Center for Next-Generation Semiconductor Technology, Sun Moon University, Chungnam, Republic of Korea
| |
Collapse
|
28
|
Zheng Y, Sun R, Yang H, Gu T, Han M, Yu C, Chen P, Zhang J, Jiang T, Ding Y, Liang L, Quan R, Yao S, Zhao X. Aucubin Promotes BMSCs Proliferation and Differentiation of Postmenopausal Osteoporosis Patients by Regulating Ferroptosis and BMP2 Signalling. J Cell Mol Med 2025; 29:e70288. [PMID: 39823248 PMCID: PMC11740986 DOI: 10.1111/jcmm.70288] [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: 12/13/2023] [Revised: 09/02/2024] [Accepted: 12/04/2024] [Indexed: 01/19/2025] Open
Abstract
Postmenopausal osteoporosis (PMOP) is a chronic systemic bone metabolism disorder. Promotion in the patterns of human bone marrow mesenchymal stem cells (hBMSCs) differentiation towards osteoblasts contributes to alleviating osteoporosis. Aucubin, a natural compound isolated from the well-known herbal medicine Eucommia, was previously shown to possess various pharmacological effects. However, its effects on hBMSCs of PMOP patients are unknown. The aim of this present research was to investigate the impact and underlying process of aucubin on cell proliferation and osteogenic differentiation in hBMSCs isolated from PMOP patients. The ability of aucubin to inhibit the ferroptosis induced by erastin in hBMSCs was detected; ROS production, ferrous ion levels, SOD, MDA, and GPX activities were tested by using commercial kits. Next, ALP staining, ARS staining, RT-qPCR, RNA-sequencing, and Western blot were applied for determining the mRNA and protein expression levels associated with the osteogenesis of hBMSCs. The study also explored the involvement of BMP2/Smads signalling in aucubin promoting the osteogenesis of hBMSCs and evaluated the effects of aucubin intervention on osteoporosis using an ovariectomised rat model. The results indicated that aucubin significantly inhibited ROS generation and oxidative stress induced by erastin and protected against ferroptosis in hBMSCs. Additionally, aucubin facilitated osteogenic differentiation of hBMSCs by activating the BMP2/SMADs pathway and attenuated the progression of osteoporosis in OVX rats, suggesting a potential therapeutic benefit for postmenopausal osteoporosis (PMOP).
Collapse
Affiliation(s)
- Yang Zheng
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Research Institute of OrthopedicsThe Affiliated Jiangnan Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
| | - Rongtai Sun
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Huan Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Liangzhu LaboratoryZhejiang University Medical CenterHangzhouChina
| | - Tianyuan Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Meichun Han
- Third Clinical Medical SchoolZhejiang Chinese Medical UniversityHangzhouChina
| | - Congcong Yu
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Pengyu Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Jianhua Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Ting Jiang
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Yangyang Ding
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Long Liang
- Department of OrthopedicsThe First Affiliated Hospital of Anhui University of Traditional Chinese MedicineHefeiChina
| | - Renfu Quan
- Research Institute of OrthopedicsThe Affiliated Jiangnan Hospital of Zhejiang Chinese Medical UniversityHangzhouChina
- Third Clinical Medical SchoolZhejiang Chinese Medical UniversityHangzhouChina
| | - Shasha Yao
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| | - Xing Zhao
- Department of Orthopaedic Surgery, Sir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
29
|
Sun G, Zhao H, Mu X, Li X, Wang J, Zhao M, Ji R, Lv H, Li Y, Chen C, Xie J, Zhang W, Duan X, Zhu S, Wang J. Whole-genome re-sequencing association study on body size traits at 10-weeks of age in Chinese indigenous geese. Front Vet Sci 2024; 11:1506471. [PMID: 39736931 PMCID: PMC11683055 DOI: 10.3389/fvets.2024.1506471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
To investigate the genetic factors underlying marketed body size traits in Chinese local geese, we conducted a comprehensive study involving nine body size traits in 251 samples at 10 weeks of age from five local breeds: Taihu goose (TH), Sichuan goose (SC), Guangfeng goose (GF), Xupu goose (XP), and Youjiang goose (YJ). Genotyping data were obtained through whole-genome re-sequencing, followed by a genome-wide association analysis utilizing the fixed and random model circulating probability unification (FarmCPU) approach. Our findings revealed 88 significant SNPs associated with body size traits, with 16 SNPs surpassing the genome-wide significance threshold (p = 3.98E-09) and 72 SNPs exceeding the suggestive significance threshold (p = 5E-07). Subsequent gene annotation identified these SNPs to be located within exonic regions of 86 candidate genes, including THADA, ATP5A1, ZNF462, PRDM8, and GH14523. Notably, functional enrichment analysis employing Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways highlighted 37 significantly enriched pathways, among which the "negative regulation of transforming growth factor beta receptor signaling pathway" (GO:0030512) emerged as relevant to goose skeletal development and the phenotypic expression of body size in geese. The identification of these novel SNPs and candidate genes associated with 10-week-old body size traits in geese presents valuable insights for future molecular breeding endeavors and the elucidation of underlying mechanisms governing body size trait formation in goose.
Collapse
Affiliation(s)
- Guobo Sun
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
- National Waterfowl of Gene Pool, Taizhou, China
| | - Hongchang Zhao
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
- National Waterfowl of Gene Pool, Taizhou, China
| | - Xiaohui Mu
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
- National Waterfowl of Gene Pool, Taizhou, China
| | - Xiaoming Li
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
| | - Jun Wang
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
- National Waterfowl of Gene Pool, Taizhou, China
| | - Mengli Zhao
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
- National Waterfowl of Gene Pool, Taizhou, China
| | - Rongchao Ji
- Taizhou Fengda Agriculture and Animal Husbandry Technology Co., Ltd., Taizhou, China
| | - Hailing Lv
- Taizhou Fengda Agriculture and Animal Husbandry Technology Co., Ltd., Taizhou, China
| | - Yang Li
- National Waterfowl of Gene Pool, Taizhou, China
| | - Chao Chen
- Taizhou Fengda Agriculture and Animal Husbandry Technology Co., Ltd., Taizhou, China
| | - Jia Xie
- Jiangsu Liangyu Agriculture and Animal Husbandry Co., Ltd., Suqian, China
| | - Wei Zhang
- Suqian March Food Co., Ltd., Suqian, China
| | - Xiujun Duan
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
| | - Shanyuan Zhu
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
| | - Jian Wang
- Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, China
- National Waterfowl of Gene Pool, Taizhou, China
- Taizhou Fengda Agriculture and Animal Husbandry Technology Co., Ltd., Taizhou, China
| |
Collapse
|
30
|
Geng Y, Bao C, Chen Y, Yan Z, Miao F, Wang T, Li Y, Li L, Sun W, Xu Y. NLRP3 deficiency improves bone healing of tooth extraction sockets through SMAD2/3-RUNX2-mediated osteoblast differentiation. Stem Cells 2024; 42:1085-1099. [PMID: 39404121 DOI: 10.1093/stmcls/sxae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 08/26/2024] [Indexed: 12/12/2024]
Abstract
Impaired bone healing following tooth extraction poses a significant challenge for implantation. As a crucial component of the natural immune system, the NLRP3 inflammasome is one of the most extensively studied pattern-recognition receptors, and is involved in multiple diseases. Yet, the role of NLRP3 in bone healing remains to be clarified. Here, to investigate the effect of NLRP3 on bone healing, we established a maxillary first molar extraction model in wild-type and NLRP3KO mice using minimally invasive techniques. We observed that NLRP3 was activated during the bone repair phase, and its depletion enhanced socket bone formation and osteoblast differentiation. Moreover, NLRP3 inflammasome activation was found to inhibit osteogenic differentiation in alveolar bone-derived mesenchymal stem cells (aBMSCs), an effect mitigated by NLRP3 deficiency. Mechanistically, we established that the SMAD2/3-RUNX2 signaling pathway is a downstream target of NLRP3 inflammasome activation, and SMAD2/3 knockdown partially reversed the significant decrease in expression of RUNX2, OSX, and ALP induced by NLRP3. Thus, our findings demonstrate that NLRP3 negatively modulates alveolar socket bone healing and contributes to the understanding of the NLRP3-induced signaling pathways involved in osteogenesis regulation.
Collapse
Affiliation(s)
- Ying Geng
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 212000, People's Republic of China
| | - Chen Bao
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
| | - Yue Chen
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
| | - Ziwei Yan
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
| | - Fen Miao
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
- Department of Stomatology, Children's Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
| | - Ting Wang
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
| | - Yingyi Li
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
| | - Lu Li
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 212000, People's Republic of China
| | - Wen Sun
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 212000, People's Republic of China
| | - Yan Xu
- Department of Periodontology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Nanjing 212000, People's Republic of China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases (Nanjing Medical University), Nanjing 212000, People's Republic of China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing 212000, People's Republic of China
| |
Collapse
|
31
|
Plichta J, Majos A, Kuna P, Panek M. Nasal allergen and methacholine provocation tests influence co‑expression patterns of TGF‑β/SMAD and MAPK signaling pathway genes in patients with asthma. Exp Ther Med 2024; 28:445. [PMID: 39386939 PMCID: PMC11462400 DOI: 10.3892/etm.2024.12735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 07/16/2024] [Indexed: 10/12/2024] Open
Abstract
Asthma is characterized by chronic bronchial inflammation and is a highly heterogeneous disease strongly influenced by both specific and non-specific exogenous factors. The present study was performed to assess the effect of nasal allergen provocation tests and methacholine provocation tests on the mRNA co-expression patterns of genes (SMAD1/3/6/7, MPK1/3 and TGFB1/3) involved in SMAD and non-SMAD TGF-β signaling pathways in patients with asthma. Reverse transcription-quantitative PCR was performed on blood samples taken pre-provocation and 1 h post-provocation to assess gene expression changes. Of the 59 patients studied, allergen provocations were administered to 27 patients and methacholine provocations to 32 patients. Correlations between expression levels of studied genes were found to be influenced markedly by the challenge administered, challenge test result and time elapsed since challenge. Importantly, increases in expression levels for four gene pairs (MAPK1-SMAD3, MAPK3-SMAD3, SMAD1-SMAD3 and SMAD3-TGFB1) were found to correlate significantly with asthma occurrence in the allergen provocation cohort, but not in the methacholine provocation cohort. The present study allows us to draw the conclusion that both intranasal allergen and bronchial methacholine challenges influence mRNA co-expression patterns of the SMAD1/3/6/7, MPK1/3 and TGFB1/3 genes.
Collapse
Affiliation(s)
- Jacek Plichta
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, 90-153 Lodz, Poland
| | - Alicja Majos
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, 90-153 Lodz, Poland
- Department of General and Transplant Surgery, Asthma and Allergy, Medical University of Lodz, 90-153 Lodz, Poland
| | - Piotr Kuna
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, 90-153 Lodz, Poland
| | - Michał Panek
- Department of Internal Medicine, Asthma and Allergy, Medical University of Lodz, 90-153 Lodz, Poland
| |
Collapse
|
32
|
Oryan A, Afzali SA, Maffulli N. Manipulation of signaling pathways in bone tissue engineering and regenerative medicine: Current knowledge, novel strategies, and future directions. Injury 2024; 55:111976. [PMID: 39454294 DOI: 10.1016/j.injury.2024.111976] [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: 04/06/2024] [Revised: 09/21/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
During osteogenesis, a large number of bioactive molecules, macromolecules, cells, and cellular signals are activated to induce bone growth and development. The activation of molecular pathways leads to the occurrence of cellular events, ultimately resulting in observable changes. Therefore, in the studies of bone tissue engineering and regenerative medicine, it is essential to target fundamental events to exploit the mechanisms involved in osteogenesis. In this context, signaling pathways are activated during osteogenesis and trigger the activation of numerous other processes involved in osteogenesis. Direct influence of signaling pathways should allow to manipulate the signaling pathways themselves and impact osteogenesis. A combination of sequential cascades takes place to drive the progression of osteogenesis. Also, the occurrence of these processes and, more generally, cellular and molecular processes related to osteogenesis necessitate the presence of transcription factors and their activity. The present review focuses on outlining several signaling pathways and transcription factors influencing the development of osteogenesis, and describes various methods of their manipulation to induce and enhance bone formation.
Collapse
Affiliation(s)
- Ahmad Oryan
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Seyed Ali Afzali
- Department of Pathology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Nicola Maffulli
- Department of Orthopaedic and Trauma Surgery, Faculty of Medicine and Psychology, Sant'Andrea Hospital Sapienza University of Rome, Rome, Italy; Centre for Sport and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK; Faculty of Medicine, School of Pharmacy and Bioengineering, Keele University, Stoke on Trent ST47QB, UK
| |
Collapse
|
33
|
Song J, Zhang Y, Jin X, Zhu Y, Li Y, Hu M. Eucommia ulmoides Oliver polysaccharide alleviates glucocorticoid-induced osteoporosis by stimulating bone formation via ERK/BMP-2/SMAD signaling. Sci Rep 2024; 14:29647. [PMID: 39609585 PMCID: PMC11604974 DOI: 10.1038/s41598-024-80859-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024] Open
Abstract
Osteoporosis (OP) is a metabolic disease characterized by low bone mineral mass owing to osteoblast dysfunction. Eucommia ulmoides Oliver (EuO) is a Chinese herbal medicine traditionally used to treat OP. Here, a polysaccharide purified from the EuO cortex (EuOCP3) was administered to OP mice constructed with dexamethasone (Dex) to investigate its anti-OP activity. EuOCP3 significantly improved Dex-induced bone microarchitecture destruction, increased osteoblast numbers and surface, and stimulated an increase in the expression of osteoblast differentiation markers in the femurs of OP mice. Furthermore, EuOCP3 was applied to MC3T3-E1 cells to further explore its effects on osteoblast differentiation. EuOCP3 significantly promoted osteoblast differentiation and increased the level of phosphorylated extracellular signal-regulated kinase1/2 (ERK1/2) and SMAD1/5/8. The EuOCP3-mediated enhancement of osteoblast differentiation-related proteins and phosphorylated SMAD1/5/8 expression levels was strongly suppressed by an ERK inhibitor (PD98059), which confirmed the critical role of ERK signaling in EuOCP3-induced osteoblast differentiation. In summary, EuOCP3 can stimulate bone formation by improving osteoblast differentiation via ERK/BMP-2/SMAD signaling, indicating the potential use of EuOCP3 as a functional ingredient in food products for anti-OP treatment.
Collapse
Affiliation(s)
- Jiyu Song
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China
| | - Yongfeng Zhang
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Xinghui Jin
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yanfeng Zhu
- School of Life Sciences, Jilin University, Changchun, 130012, China
| | - Yutong Li
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, 130021, China.
| |
Collapse
|
34
|
Li X, Zhang KW, Zhang ZY, Wu JJ, Yuan ZD, Yuan FL, Chen J. Inhibiting dipeptidyl peptidase 4 positive fibroblasts using zinc sulfide cellulose nanofiber scaffolds to achieve scarless healing. Int J Biol Macromol 2024; 282:137525. [PMID: 39537047 DOI: 10.1016/j.ijbiomac.2024.137525] [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: 08/14/2024] [Revised: 11/05/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Wound regeneration with integral function and cutaneous appendages remains challenging in wound dressing applications. Cellulose nanofibers (CNF) exhibit remarkable characteristics in wound dressing applications; however, their utility in the wound healing process is limited by insufficient scar inhibition and regenerative healing. Herein, inspired by fibroblast heterogeneity mediating wound healing and skin regeneration, we developed a CNF scaffold designed to block Dipeptidyl Peptidase 4 positive (DPP4+) fibroblasts for regenerative healing. CNF encapsulated sitagliptin (SITA) and zinc sulfide nanoparticles (NZnS), namely CNF@SITA@NZnS, to fabricate a novel biomaterial for scar reduction and regenerative healing. The scaffold promoted scarless healing and hair follicle regeneration in rats. In vivo experiments, the wounds in the scaffold showed less skin fibrosis, a better collagen ratio and more new hair follicles. In vitro experiments showed that the scaffold material promoted scarless healing, possibly by inhibiting the secretion of extracellular matrix and fibroblast-to-myofibroblast conversion. The promotion of hair follicle regeneration by the scaffold material may be due to promotion of the migration and proliferation of human hair follicle papilla cells. RNA sequencing is performed to explore the underlying mechanisms, which can activate ECM-receptor interaction pathway in favor of the wound healing process. The inhibiting effect of CNF@SITA@NZnS scaffold on DPP4+ fibroblasts can be a potential target to reduce scarring and promote skin regeneration.
Collapse
Affiliation(s)
- Xia Li
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Kai-Wen Zhang
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Zheng-Yu Zhang
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Zheng-Dong Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Feng-Lai Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China.
| | - Jinghua Chen
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China; Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
35
|
Liu Y, Zhou Q, Zou G, Zhang W. Inhibin subunit beta B (INHBB): an emerging role in tumor progression. J Physiol Biochem 2024; 80:775-793. [PMID: 39183219 DOI: 10.1007/s13105-024-01041-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 07/30/2024] [Indexed: 08/27/2024]
Abstract
The gene inhibin subunit beta B (INHBB) encodes the inhibin βB subunit, which is involved in forming protein members of the transforming growth factor-β (TGF-β) superfamily. The TGF-β superfamily is extensively involved in cell proliferation, differentiation, adhesion, movement, metabolism, communication, and death. Activins and inhibins, which belong to the TGF-β superfamily, were first discovered in ovarian follicular fluid. They were initially described as regulators of pituitary follicle-stimulating hormone (FSH) secretion both in vivo and in vitro. Later studies found that INHBB is expressed not only in reproductive organs such as the ovary, uterus, and testis but also in numerous other organs, including the brain, spinal cord, liver, kidneys, and adrenal glands. This wide distribution implies its involvement in the normal physiological functions of various organs; however, the mechanisms underlying these functions have not yet been fully elucidated. Recent studies suggest that INHBB plays a significant, yet complex role in tumorigenesis. It appears to have dual effects, promoting tumor progression in some contexts while inhibiting it in others, although these roles are not yet fully understood. In this paper, we review the different expression patterns, functions, and mechanisms of INHBB in normal and tumor tissues to illustrate the research prospects of INHBB in tumor progression.
Collapse
Affiliation(s)
- Ying Liu
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Tongzipo Road 172, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
- Department of Clinical Laboratory, Zhengzhou Orthopedic Hospital, Zhengzhou, Henan, People's Republic of China
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China
| | - Qing Zhou
- Department of Clinical Laboratory, First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, People's Republic of China
| | - Guoying Zou
- Department of Clinical Laboratory, Brain Hospital of Hunan Province, Changsha, Hunan, People's Republic of China
| | - Wenling Zhang
- Department of Medical Laboratory Science, The Third Xiangya Hospital, Central South University, Tongzipo Road 172, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China.
- Department of Medical Laboratory Science, Xiangya School of Medicine, Central South University, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
36
|
Li Q, Wang J, Zhao C. From Genomics to Metabolomics: Molecular Insights into Osteoporosis for Enhanced Diagnostic and Therapeutic Approaches. Biomedicines 2024; 12:2389. [PMID: 39457701 PMCID: PMC11505085 DOI: 10.3390/biomedicines12102389] [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: 09/16/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Osteoporosis (OP) is a prevalent skeletal disorder characterized by decreased bone mineral density (BMD) and increased fracture risk. The advancements in omics technologies-genomics, transcriptomics, proteomics, and metabolomics-have provided significant insights into the molecular mechanisms driving OP. These technologies offer critical perspectives on genetic predispositions, gene expression regulation, protein signatures, and metabolic alterations, enabling the identification of novel biomarkers for diagnosis and therapeutic targets. This review underscores the potential of these multi-omics approaches to bridge the gap between basic research and clinical applications, paving the way for precision medicine in OP management. By integrating these technologies, researchers can contribute to improved diagnostics, preventative strategies, and treatments for patients suffering from OP and related conditions.
Collapse
Affiliation(s)
- Qingmei Li
- Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Jihan Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
| | - Congzhe Zhao
- Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| |
Collapse
|
37
|
Gambari L, Pagnotta E, Ugolini L, Righetti L, Amore E, Grigolo B, Filardo G, Grassi F. Insights into Osteogenesis Induced by Crude Brassicaceae Seeds Extracts: A Role for Glucosinolates. Nutrients 2024; 16:3457. [PMID: 39458452 PMCID: PMC11510261 DOI: 10.3390/nu16203457] [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: 09/25/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Crude extracts from the Brassica genus have recently emerged as promising phytochemicals for preventing bone loss. While the most documented evidence suggests that their general biological activity is due to glucosinolates' (GLSs') hydrolysis products, the direct activity of GLSs is beginning to be uncovered. However, the contribution of GLSs to the bone-sparing activity of crude Brassicaceae extracts has seldom been addressed. Here, we aimed to gain insights into this gap by studying in the same in vitro model of human osteogenesis the effect of two Brassica seed extracts (Eruca sativa and Lepidium sativum) obtained from defatted seed meals, comparing them to the isolated GLSs most represented in their composition, glucoerucin (GER) and glucotropaeolin (GTL), for Eruca sativa and Lepidium sativum, respectively. Methods: Osteogenic differentiation of human mesenchymal stromal cells (hMSCs) was assessed by alizarin red staining assay and real-time PCR, respectively, evaluating mineral apposition and mRNA expression of specific osteogenic genes. Results: Both Brassica extracts and GLSs increased the osteogenic differentiation, indicating that the stimulating effect of Brassica extracts can be at least partially attributed to GLSs. Moreover, these data extend previous evidence of the effect of unhydrolyzed glucoraphanin (GRA) on osteogenesis to other types of GLSs: GER and GTL. Notably, E. sativa extract and GTL induced higher osteogenic stimulation than Lepidium sativum extract and GER, respectively. Conclusions: Overall, this study expands the knowledge on the possible application of Brassica-derived bioactive molecules as natural alternatives for the prevention and treatment of bone-loss pathologies.
Collapse
Affiliation(s)
- Laura Gambari
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (L.G.); (E.A.); (B.G.)
| | - Eleonora Pagnotta
- Research Centre for Cereal and Industrial Crops (CREA-CI), CREA Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Via di Corticella 133, 40128 Bologna, Italy; (E.P.); (L.U.); (L.R.)
| | - Luisa Ugolini
- Research Centre for Cereal and Industrial Crops (CREA-CI), CREA Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Via di Corticella 133, 40128 Bologna, Italy; (E.P.); (L.U.); (L.R.)
| | - Laura Righetti
- Research Centre for Cereal and Industrial Crops (CREA-CI), CREA Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria, Via di Corticella 133, 40128 Bologna, Italy; (E.P.); (L.U.); (L.R.)
| | - Emanuela Amore
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (L.G.); (E.A.); (B.G.)
| | - Brunella Grigolo
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (L.G.); (E.A.); (B.G.)
| | - Giuseppe Filardo
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Viale Pietro Capelli 1, 6962 Lugano, Switzerland;
| | - Francesco Grassi
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; (L.G.); (E.A.); (B.G.)
| |
Collapse
|
38
|
Mills EG, Abbara A, Dhillo WS, Comninos AN. Interactions between kisspeptin and bone: Cellular mechanisms, clinical evidence, and future potential. Ann N Y Acad Sci 2024; 1540:47-60. [PMID: 39269749 DOI: 10.1111/nyas.15213] [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: 09/15/2024]
Abstract
The neuropeptide kisspeptin and its cognate receptor have been extensively studied in reproductive physiology, with diverse and well-established functions, including as an upstream regulator of pubertal onset, reproductive hormone secretion, and sexual behavior. Besides classical reproduction, both kisspeptin and its receptor are extensively expressed in bone-resorbing osteoclasts and bone-forming osteoblasts, which putatively permits direct bone effects. Accordingly, this sets the scene for recent compelling findings derived from in vitro experiments through to in vivo and clinical studies revealing prominent regulatory interactions for kisspeptin signaling in bone metabolism, as well as certain oncological aspects of bone metabolism. Herein, we comprehensively examine the experimental evidence obtained to date supporting the interaction between kisspeptin and bone. A comprehensive understanding of this emerging facet of kisspeptin biology is fundamental to exploiting the future therapeutic potential of kisspeptin-based medicines as a novel strategy for treating bone-related disorders.
Collapse
Affiliation(s)
- Edouard G Mills
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Ali Abbara
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Waljit S Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Alexander N Comninos
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
- Endocrine Bone Unit, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
39
|
Zhu Z, Huang F, Gao M, Liu M, Zhang Y, Tang L, Wu J, Yu H, He C, Chen J, Yang Z, Chen Z, Li Y, Chen H, Lei T, Zeng F, Cui Y. Osteogenic-Like Microenvironment of Renal Interstitium Induced by Osteomodulin Contributes to Randall's Plaque Formation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405875. [PMID: 39225583 PMCID: PMC11516157 DOI: 10.1002/advs.202405875] [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: 05/29/2024] [Revised: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Calcium oxalate (CaOx) kidney stones are common and recurrent, lacking pharmacological prevention. Randall's plaques (RPs), calcium deposits in renal papillae, serve as niduses for some CaOx stones. This study explores the role of osteogenic-like cells in RP formation resembling ossification. CaP crystals deposit around renal tubules, interstitium, and blood vessels in RP tissues. Human renal interstitial fibroblasts (hRIFs) exhibit the highest osteogenic-like differentiation potential compared to chloride voltage-gated channel Ka positive tubular epithelial cells, aquaporin 2 positive collecting duct cells, and vascular endothelial cells, echoing the upregulated osteogenic markers primarily in hRIFs within RP tissues. Utilizing RNA-seq, osteomodulin (OMD) is found to be upregulated in hRIFs within RP tissues and hRIFs following osteogenic induction. Furthermore, OMD colocalizes with CaP crystals and calcium vesicles within RP tissues. OMD can enhance osteogenic-like differentiation of hRIFs in vitro and in vivo. Additionally, crystal deposits are attenuated in mice with Omd deletion in renal interstitial fibroblasts following CaOx nephrocalcinosis induction. Mechanically, a positive feedback loop of OMD/BMP2/BMPR1A/RUNX2/OMD drives hRIFs to adopt osteogenic-like fates, by which OMD induces osteogenic-like microenvironment of renal interstitium to participate in RP formation. We identify OMD upregulation as a pathological feature of RP, paving the way for preventing CaOx stones.
Collapse
Affiliation(s)
- Zewu Zhu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of Internal MedicineSection EndocrinologyYale University School of MedicineNew HavenCT06519USA
| | - Fang Huang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Meng Gao
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Minghui Liu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Youjie Zhang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Liang Tang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Jian Wu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Hao Yu
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Cheng He
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Jinbo Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Zhongqing Yang
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Zhiyong Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yang Li
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Hequn Chen
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Ting Lei
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of OrthopedicsXiangya HospitalCentral South UniversityChangshaHunan410008China
- Department of Orthopaedic SurgeryThe First Affiliated HospitalSchool of MedicineZhejiang UniversityHangzhouZhejiang310006China
| | - Feng Zeng
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| | - Yu Cui
- Department of UrologyXiangya HospitalCentral South UniversityChangshaHunan410008China
| |
Collapse
|
40
|
Chatterjee P, Stevens HY, Kippner LE, Bowles-Welch AC, Drissi H, Mautner K, Yeago C, Gibson G, Roy K. Single-cell transcriptome and crosstalk analysis reveals immune alterations and key pathways in the bone marrow of knee OA patients. iScience 2024; 27:110827. [PMID: 39310769 PMCID: PMC11416684 DOI: 10.1016/j.isci.2024.110827] [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: 11/14/2023] [Revised: 06/10/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
Knee osteoarthritis (OA) is a significant medical and economic burden. To understand systemic immune effects, we performed deep exploration of bone marrow aspirate concentrates (BMACs) from knee-OA patients via single-cell RNA sequencing and proteomic analyses from a randomized clinical trial (MILES: NCT03818737). We found significant cellular and immune alterations in the bone marrow, specifically in MSCs, T cells and NK cells, along with changes in intra-tissue cellular crosstalk during OA progression. Unlike previous studies focusing on injury sites or peripheral blood, our probe into the bone marrow-an inflammation and immune regulation hub-highlights remote organ impact of OA, identifying cell types and pathways for potential therapeutic targeting. Our findings highlight increased cellular senescence and inflammatory pathways, revealing key upstream genes, transcription factors, and ligands. Additionally, we identified significant enrichment in key biological pathways like PI3-AKT-mTOR signaling and IFN responses, showing their potentially crucial role in OA onset and progression.
Collapse
Affiliation(s)
- Paramita Chatterjee
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Hazel Y. Stevens
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Linda E. Kippner
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Annie C. Bowles-Welch
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kenneth Mautner
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Carolyn Yeago
- The Parker H. Petit Institute for Bioengineering and Biosciences Georgia Institute of Technology, Atlanta, GA, USA
| | - Greg Gibson
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Krishnendu Roy
- Marcus Center for Therapeutic Cell Characterization and Manufacturing, The Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, USA
- Department of Biomedical Engineering, School of Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, School of Medicine, Vanderbilt University, Nashville, TN, USA
- Department of Chemical and Biomolecular Engineering, School of Engineering, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
41
|
Li Y, Jiang Y, Yan H, Qin Z, Peng Y, Lv D, Zhang H. Global isonicotinylome analysis identified SMAD3 isonicotinylation promotes liver cancer cell epithelial-mesenchymal transition and invasion. iScience 2024; 27:110775. [PMID: 39286495 PMCID: PMC11403401 DOI: 10.1016/j.isci.2024.110775] [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: 04/11/2024] [Revised: 07/02/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Histone lysine isonicotinylation (Kinic) induced by isoniazid (INH) was recently identified as a post-translational modification in cells. However, global cellular non-histone proteins Kinic remains unclear. Using proteomic technology, we identified 11,442 Kinic sites across 2,792 proteins and demonstrated that Kinic of non-histone proteins is involved in multiple function pathways. Non-histone proteins Kinic can be regulated by isonicotinyl-transferases, including CBP and Tip60, and deisonicotinylases, including HDAC8 and HDAC6. In particular, the Kinic of poly (ADP-ribose) (PAR) polymerase 1 (PARP1) can be catalyzed by CBP and deisonicotinylation can be catalyzed by HDAC8. Tip60 and HDAC6 are isonicotinyl-transferase and the deisonicotinylase of SMAD3, respectively. Importantly, we found the K378inic of SMAD3 increases its phosphorylation, activates TGFβ pathway, and promotes liver cancer cells migration and invasion. In conclusion, our study demonstrated non-histone proteins Kinic occur extensively in cells and plays an important role in regulation of various cellular functions, including cancer progression.
Collapse
Affiliation(s)
- Yixiao Li
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yuhan Jiang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Haoyi Yan
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Ziheng Qin
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yidi Peng
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Danyu Lv
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Hongquan Zhang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
42
|
Čandrlić M, Jerbić Radetić AT, Omrčen H, Franović B, Batičić L, Gulić T, Čaljkušić-Mance T, Zoričić Cvek S, Malešić L, Perić Kačarević Ž, Cvijanović Peloza O. Regeneration of Critical Calvarial Bone Defects Using Bovine Xenograft, Magnesium-Enriched Bovine Xenograft and Autologous Dentin in Rats: Micro-CT, Gene Expression and Immunohistochemical Analysis. J Funct Biomater 2024; 15:270. [PMID: 39330245 PMCID: PMC11433293 DOI: 10.3390/jfb15090270] [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: 07/29/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024] Open
Abstract
The aim of this study was to evaluate the efficacy of autologous dentin (AD), bovine xenograft (BX) and magnesium-enriched bovine xenograft (BX + Mg) in the healing of critical cranial bone defects (CCBDs) in rats. Eighty male Wistar rats were divided into four groups: BX, BX + Mg, AD and the control group (no intervention). Eight mm CCBDs were created and treated with the respective biomaterials. Healing was assessed 7, 15, 21 and 30 days after surgery by micro-computed tomography (micro-CT), real-time polymerase chain reaction (RT-PCR) and immunohistochemical analysis. Micro-CT analysis showed that AD had the highest bone volume and the least amount of residual biomaterial at day 30, indicating robust bone formation and efficient resorption. BX + Mg showed significant bone volume but had more residual biomaterial compared to AD. RT-PCR showed that the expression of osteocalcin (OC), the receptor activator of nuclear factor κB (RANK) and sclerostin (SOST), was highest in the AD group at day 21 and vascular endothelial growth factor (VEGF) at day 15, indicating increased osteogenesis and angiogenesis in the AD group. Immunohistochemical staining confirmed intense BMP-2/4 and SMAD-1/5/8 expression in the AD group, indicating osteoinductive properties. The favorable gene expression profile and biocompatibility of AD and BX + Mg make them promising candidates for clinical applications in bone tissue engineering. Further research is required to fully exploit their potential in regenerative surgery.
Collapse
Affiliation(s)
- Marija Čandrlić
- Department of Integrative Dental Medicine, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Ana Terezija Jerbić Radetić
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
| | - Hrvoje Omrčen
- Department of Clinical Microbiology, Teaching Institute of Public Health of Primorsko-Goranska County, Krešimirova 52a, 51000 Rijeka, Croatia
| | - Barbara Franović
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
- Doctoral School of Biomedicine and Health, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
| | - Lara Batičić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
| | - Tamara Gulić
- Department of Physiology, Immunology and Pathophysiology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
| | - Tea Čaljkušić-Mance
- Department of Ophthalmology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
- Clinic for Ophthalmology, Clinical Hospital Center Rijeka, Krešimirova 42, 51000 Rijeka, Croatia
| | - Sanja Zoričić Cvek
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
| | - Lucija Malešić
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
- Dental Clinic Rident, Franje Čandeka 39, 51000 Rijeka, Croatia
| | - Željka Perić Kačarević
- Department of Anatomy, Histology, Embriology, Pathology Anatomy and Pathology Histology, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, 31000 Osijek, Croatia
- Botiss Biomaterials GmbH, 15806 Zossen, Germany
| | - Olga Cvijanović Peloza
- Department of Anatomy, Faculty of Medicine, University of Rijeka, Braće Branchetta 20/1, 51000 Rijeka, Croatia
| |
Collapse
|
43
|
Kolipaka R, Magesh I, Bharathy MA, Karthik S, Saranya I, Selvamurugan N. A potential function for MicroRNA-124 in normal and pathological bone conditions. Noncoding RNA Res 2024; 9:687-694. [PMID: 38577015 PMCID: PMC10990750 DOI: 10.1016/j.ncrna.2024.02.018] [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: 01/07/2024] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 04/06/2024] Open
Abstract
Cells produce short single-stranded non-coding RNAs (ncRNAs) called microRNAs (miRNAs), which actively regulate gene expression at the posttranscriptional level. Several miRNAs have been observed to exert significant impacts on bone health and bone-related disorders. One of these, miR-124, is observed in bone microenvironments and is conserved across species. It affects bone cell growth and differentiation by activating different transcription factors and signaling pathways. In-depth functional analyses of miR-124 have revealed several physiological and pathological roles exerted through interactions with other ncRNAs. Deciphering these RNA-mediated signaling networks and pathways is essential for understanding the potential impacts of dysregulated miRNA functions on bone biology. In this review, we aim to provide a comprehensive analysis of miR-124's involvement in bone physiology and pathology. We highlight the importance of miR-124 in controlling transcription factors and signaling pathways that promote bone growth. This review reveals therapeutic implications for the treatment of bone-related diseases.
Collapse
Affiliation(s)
- Rushil Kolipaka
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Induja Magesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - M.R. Ashok Bharathy
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - S. Karthik
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - I. Saranya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - N. Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| |
Collapse
|
44
|
Ji PC, Xie YS, Guo WK, Fu B, Chen XM. p38 Signaling Mediates Naringin-Induced Osteogenic Differentiation of Porcine Metanephric Mesenchymal Cells. Chin J Integr Med 2024; 30:818-825. [PMID: 38850479 DOI: 10.1007/s11655-024-3761-1] [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] [Accepted: 01/23/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE To explore the potential of metanephric mesenchymal cells (MMCs) for osteogenesis and naringin's ability to enhance this process and its molecular mechanism. METHODS Porcine MMCs at 70 days of gestation were used as tool cells, cultured in osteogenic induction medium, identified by immunocytochemistry staining. Osteogenic potential of porcine MMCs and naringin's ability to enhance this process was tested by detecting changes in cell viability, alkaline phosphatase (ALP) activity, the expression of runt-related transcription factor 2 (Runx2), osteopontin (OPN) and osteocalcin (OCN), and the formation of mineralized nodules, and the application of the p38 signaling pathway inhibitor SB203580 vitiated the osteogenesis-promoting effect of naringin. RESULTS Immunocytochemical staining showed that the cells were Vimentin and Six2(+), E-cadherin and CK-18(-). Naringin can activate the p38 signaling pathway to enhance the osteogenesis of porcine MMCs by increasing cell viability, ALP activity, the expressions of Runx2, OPN and OCN, and the formation of mineralized nodules (P<0.05). The application of p38 signaling pathway inhibitor SB203580 vitiated the osteogenesis-promoting effect of naringin, manifested by decreased ALP activity, the expressions of Runx2, OPN and OCN, and the formation of mineralized nodules (P<0.05). CONCLUSION Naringin, the active ingredient of Chinese herbal medicine Rhizoma Drynariae for nourishing Shen (Kidney) and strengthening bone, enhances the osteogenic differentiation of renal MMCs through the p38 signaling pathway.
Collapse
Affiliation(s)
- Peng-Cheng Ji
- Chinese PLA Medical School, Beijing, 100853, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
| | - Yuan-Sheng Xie
- Chinese PLA Medical School, Beijing, 100853, China.
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China.
- School of Medicine, Nankai University, Tianjin, 300071, China.
| | - Wen-Kai Guo
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Bo Fu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
| | - Xiang-Mei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
| |
Collapse
|
45
|
Stepkowski S, Bekbolsynov D, Oenick J, Brar S, Mierzejewska B, Rees MA, Ekwenna O. The Major Role of T Regulatory Cells in the Efficiency of Vaccination in General and Immunocompromised Populations: A Review. Vaccines (Basel) 2024; 12:992. [PMID: 39340024 PMCID: PMC11436018 DOI: 10.3390/vaccines12090992] [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: 07/17/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Since their conception with the smallpox vaccine, vaccines used worldwide have mitigated multiple pandemics, including the recent COVID-19 outbreak. Insightful studies have uncovered the complexities of different functional networks of CD4 T cells (T helper 1 (Th1); Th2, Th17) and CD8 T cells (T cytotoxic; Tc), as well as B cell (BIgM, BIgG, BIgA and BIgE) subsets, during the response to vaccination. Both T and B cell subsets form central, peripheral, and tissue-resident subsets during vaccination. It has also become apparent that each vaccination forms a network of T regulatory subsets, namely CD4+ CD25+ Foxp3+ T regulatory (Treg) cells and interleukin-10 (IL-10)-producing CD4+ Foxp3- T regulatory 1 (Tr1), as well as many others, which shape the quality/quantity of vaccine-specific IgM, IgG, and IgA antibody production. These components are especially critical for immunocompromised patients, such as older individuals and allograft recipients, as their vaccination may be ineffective or less effective. This review focuses on considering how the pre- and post-vaccination Treg/Tr1 levels influence the vaccination efficacy. Experimental and clinical work has revealed that Treg/Tr1 involvement evokes different immune mechanisms in diminishing vaccine-induced cellular/humoral responses. Alternative steps may be considered to improve the vaccination response, such as increasing the dose, changing the delivery route, and/or repeated booster doses of vaccines. Vaccination may be combined with anti-CD25 (IL-2Rα chain) or anti-programmed cell death protein 1 (PD-1) monoclonal antibodies (mAb) to decrease the Tregs and boost the T/B cell immune response. All of these data and strategies for immunizations are presented and discussed, aiming to improve the efficacy of vaccination in humans and especially in immunocompromised and older individuals, as well as organ transplant patients.
Collapse
Affiliation(s)
- Stanislaw Stepkowski
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH 43614, USA; (D.B.); (B.M.)
| | - Dulat Bekbolsynov
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH 43614, USA; (D.B.); (B.M.)
| | - Jared Oenick
- Neurological Surgery, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA;
| | - Surina Brar
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH 43614, USA; (D.B.); (B.M.)
| | - Beata Mierzejewska
- Department of Medical Microbiology and Immunology, University of Toledo, Toledo, OH 43614, USA; (D.B.); (B.M.)
| | - Michael A. Rees
- Department of Urology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (M.A.R.); (O.E.)
| | - Obi Ekwenna
- Department of Urology, The University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (M.A.R.); (O.E.)
| |
Collapse
|
46
|
Chen C, Li Y, Feng T, Chen X, Li C, Li L, Zhu M, Chang Y, Wang S. LMK-235 suppresses osteoclastogenesis and promotes osteoblastogenesis by inhibiting HDAC4. Sci Rep 2024; 14:19973. [PMID: 39198677 PMCID: PMC11358535 DOI: 10.1038/s41598-024-70814-8] [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: 06/21/2024] [Accepted: 08/21/2024] [Indexed: 09/01/2024] Open
Abstract
Osteoblasts and osteoclasts play an important role in maintaining the structural integrity of bone tissue, in which osteoclasts degrade bone structure and osteoblasts restore bone tissue. The imbalance of osteoblast and osteoclast function can lead to many bone-related diseases, such as osteoporosis and inflammatory osteolysis. The drug that can both promote bone formation and inhibit bone loss will be able to treat those diseases. In this study, it was found that LMK-235, an selective HDAC4/5 inhibitor, inhibited the differentiation and maturation of osteoclasts by regulating NF-κB and p-Smad2/3 signaling pathways via inhibition of HDAC4. At the same time, we found that LMK-235 promoted osteoblast mineralization by upregulating Runx2 expression via inhibition of HDAC4. In vivo, LMK-235 was able to alleviate lipopolysaccharide (LPS)-induced calvarial osteolysis and promote the repair of bone defects. Taken together, LMK-235 suppresses osteoclast differentiation and promotes osteoblast formation by inhibiting HDAC4. This may provide a valuable treatment for bone diseases caused by abnormal osteoclast bone resorption and osteoblast bone regeneration.
Collapse
Affiliation(s)
- Chongwei Chen
- Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Yue Li
- Department of Biochemistry, Basic Medical College of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Teng Feng
- Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Xinping Chen
- Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Chengwei Li
- Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Lu Li
- Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China
| | - Mengbo Zhu
- Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Yaqiong Chang
- Department of Nursing, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
| | - Shaowei Wang
- Shanxi Key Lab of Bone and Soft Tissue Injury Repair, Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, People's Republic of China.
| |
Collapse
|
47
|
López-Cerdá S, Molinaro G, Tello RP, Correia A, Waris E, Hirvonen J, Barreto G, Santos HA. Antifibrotic and Pro-regenerative Effects of SMAD3 siRNA and Collagen I mRNA-Loaded Lipid Nanoparticles in Human Tenocytes. ACS APPLIED NANO MATERIALS 2024; 7:17736-17747. [PMID: 39144399 PMCID: PMC11320386 DOI: 10.1021/acsanm.4c02996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/02/2024] [Accepted: 07/07/2024] [Indexed: 08/16/2024]
Abstract
Tendinopathy involves the inflammation and degeneration of the tendon due to repetitive strain injury. Current treatments primarily target inflammation resolution, yet they do not aim at tissue regeneration. In this study, a microfluidics approach is harnessed to develop a platform of lipid nanoparticles (LNPs) loaded simultaneously with SMAD3 siRNA and collagen I mRNA, aiming to explore its potential dual antifibrotic and regenerative effects in human tenocytes. The developed LNPs displayed size homogeneity and colloidal stability and exhibited high cytocompatibility in human tenocytes. Moreover, LNPs allowed for efficient uptake and transfection efficiency of the RNAs. In the in vitro efficacy studies, the gene expression and production of SMAD3 and collagen I were tested by real-time quantitative chain polymerase reaction and immuno- and intracellular staining, revealing collagen I production enhancement, SMAD3 inhibition, and modulation of other tendon repair factors by the LNPs. Overall, the potential of this platform of RNA-loaded LNPs to be used as a dual therapeutic approach to prevent fibrosis and promote tissue remodeling in late stages of tendon diseases was confirmed.
Collapse
Affiliation(s)
- Sandra López-Cerdá
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Giuseppina Molinaro
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Rubén Pareja Tello
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Alexandra Correia
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Eero Waris
- Department
of Hand Surgery, University of Helsinki
and Helsinki University Hospital, 00029 Helsinki, Finland
| | - Jouni Hirvonen
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Goncalo Barreto
- Translational
Immunology Research Program, Faculty of Medicine, University of Helsinki, PL 4 (Yliopistonkatu 3), 00014 Helsinki, Finland
- Medical
Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical
Engineering, Aalto University, 02150 Espoo, Finland
- Orton
Orthopedic Hospital, Tenholantie 10, 00280 Helsinki, Finland
| | - Hélder A. Santos
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
- Department
of Biomedical Engineering, The Personalized Medicine Research Institute
(PRECISION), University Medical Center Groningen
(UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| |
Collapse
|
48
|
Zou Y, Mei X, Wang X, Zhang X, Wang X, Xiang W, Lu N. Fibrin-konjac glucomannan-black phosphorus hydrogel scaffolds loaded with nasal ectodermal mesenchymal stem cells accelerated alveolar bone regeneration. BMC Oral Health 2024; 24:878. [PMID: 39095803 PMCID: PMC11297757 DOI: 10.1186/s12903-024-04649-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Effective treatments for the alveolar bone defect remain a major concern in dental therapy. The objectives of this study were to develop a fibrin and konjac glucomannan (KGM) composite hydrogel as scaffolds for the osteogenesis of nasal mucosa-derived ectodermal mesenchymal stem cells (EMSCs) for the regeneration of alveolar bone defect, and to investigate the osteogenesis-accelerating effects of black phosphorus nanoparticles (BPNs) embedded in the hydrogels. METHODS Primary EMSCs were isolated from rat nasal mucosa and used for the alveolar bone recovery. Fibrin and KGM were prepared in different ratios for osteomimetic hydrogel scaffolds, and the optimal ratio was determined by mechanical properties and biocompatibility analysis. Then, the optimal hydrogels were integrated with BPNs to obtain BPNs/fibrin-KGM hydrogels, and the effects on osteogenic EMSCs in vitro were evaluated. To explore the osteogenesis-enhancing effects of hydrogels in vivo, the BPNs/fibrin-KGM scaffolds combined with EMSCs were implanted to a rat model of alveolar bone defect. Micro-computed tomography (CT), histological examination, real-time quantitative polymerase chain reaction (RT-qPCR) and western blot were conducted to evaluate the bone morphology and expression of osteogenesis-related genes of the bone regeneration. RESULTS The addition of KGM improved the mechanical properties and biodegradation characteristics of the fibrin hydrogels. In vitro, the BPNs-containing compound hydrogel was proved to be biocompatible and capable of enhancing the osteogenesis of EMSCs by upregulating the mineralization and the activity of alkaline phosphatase. In vivo, the micro-CT analysis and histological evaluation demonstrated that rats implanted EMSCs-BPNs/fibrin-KGM hydrogels exhibited the best bone reconstruction. And compared to the model group, the expression of osteogenesis genes including osteopontin (Opn, p < 0.0001), osteocalcin (Ocn, p < 0.0001), type collagen (Col , p < 0.0001), bone morphogenetic protein-2 (Bmp2, p < 0.0001), Smad1 (p = 0.0006), and runt-related transcription factor 2 (Runx2, p < 0.0001) were all significantly upregulated. CONCLUSIONS EMSCs/BPNs-containing fibrin-KGM hydrogels accelerated the recovery of the alveolar bone defect in rats by effectively up-regulating the expression of osteogenesis-related genes, promoting the formation and mineralisation of bone matrix.
Collapse
Affiliation(s)
- Yin Zou
- Department of Stomatology, Affiliated Children's Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China
| | - Xue Mei
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China
| | - Xinhe Wang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China
| | - Xuan Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China
| | - Xun Wang
- Jiangnan University Medical Center, Wuxi, Jiangsu Province, People's Republic of China
| | - Wen Xiang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China
| | - Naiyan Lu
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu Province, People's Republic of China.
| |
Collapse
|
49
|
Amroodi MN, Maghsoudloo M, Amiri S, Mokhtari K, Mohseni P, Pourmarjani A, Jamali B, Khosroshahi EM, Asadi S, Tabrizian P, Entezari M, Hashemi M, Wan R. Unraveling the molecular and immunological landscape: Exploring signaling pathways in osteoporosis. Biomed Pharmacother 2024; 177:116954. [PMID: 38906027 DOI: 10.1016/j.biopha.2024.116954] [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: 04/19/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 06/23/2024] Open
Abstract
Osteoporosis, characterized by compromised bone density and microarchitecture, represents a significant global health challenge, particularly in aging populations. This comprehensive review delves into the intricate signaling pathways implicated in the pathogenesis of osteoporosis, providing valuable insights into the pivotal role of signal transduction in maintaining bone homeostasis. The exploration encompasses cellular signaling pathways such as Wnt, Notch, JAK/STAT, NF-κB, and TGF-β, all of which play crucial roles in bone remodeling. The dysregulation of these pathways is a contributing factor to osteoporosis, necessitating a profound understanding of their complexities to unveil the molecular mechanisms underlying bone loss. The review highlights the pathological significance of disrupted signaling in osteoporosis, emphasizing how these deviations impact the functionality of osteoblasts and osteoclasts, ultimately resulting in heightened bone resorption and compromised bone formation. A nuanced analysis of the intricate crosstalk between these pathways is provided to underscore their relevance in the pathophysiology of osteoporosis. Furthermore, the study addresses some of the most crucial long non-coding RNAs (lncRNAs) associated with osteoporosis, adding an additional layer of academic depth to the exploration of immune system involvement in various types of osteoporosis. Finally, we propose that SKP1 can serve as a potential biomarker in osteoporosis.
Collapse
Affiliation(s)
- Morteza Nakhaei Amroodi
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, department of orthopedic, school of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Shayan Amiri
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, department of orthopedic, school of medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Khatere Mokhtari
- Department of Cellular and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Parnaz Mohseni
- Department of Pediatrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Pourmarjani
- Department of Pediatrics, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Behdokht Jamali
- Department of microbiology and genetics, kherad Institute of higher education, Busheher, lran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Pouria Tabrizian
- Bone and Joint Reconstruction Research Center, Shafa Orthopedic Hospital, department of orthopedic, school of medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Runlan Wan
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou 646000, China; Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
50
|
Wu Z, Li W, Jiang K, Lin Z, Qian C, Wu M, Xia Y, Li N, Zhang H, Xiao H, Bai J, Geng D. Regulation of bone homeostasis: signaling pathways and therapeutic targets. MedComm (Beijing) 2024; 5:e657. [PMID: 39049966 PMCID: PMC11266958 DOI: 10.1002/mco2.657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
As a highly dynamic tissue, bone is continuously rebuilt throughout life. Both bone formation by osteoblasts and bone resorption by osteoclasts constitute bone reconstruction homeostasis. The equilibrium of bone homeostasis is governed by many complicated signaling pathways that weave together to form an intricate network. These pathways coordinate the meticulous processes of bone formation and resorption, ensuring the structural integrity and dynamic vitality of the skeletal system. Dysregulation of the bone homeostatic regulatory signaling network contributes to the development and progression of many skeletal diseases. Significantly, imbalanced bone homeostasis further disrupts the signaling network and triggers a cascade reaction that exacerbates disease progression and engenders a deleterious cycle. Here, we summarize the influence of signaling pathways on bone homeostasis, elucidating the interplay and crosstalk among them. Additionally, we review the mechanisms underpinning bone homeostatic imbalances across diverse disease landscapes, highlighting current and prospective therapeutic targets and clinical drugs. We hope that this review will contribute to a holistic understanding of the signaling pathways and molecular mechanisms sustaining bone homeostasis, which are promising to contribute to further research on bone homeostasis and shed light on the development of targeted drugs.
Collapse
Affiliation(s)
- Zebin Wu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Wenming Li
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Kunlong Jiang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Zhixiang Lin
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Chen Qian
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Mingzhou Wu
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yu Xia
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Ning Li
- Department of OrthopedicsCentre for Leading Medicine and Advanced Technologies of IHMDivision of Life Sciences and MedicineThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Hongtao Zhang
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Haixiang Xiao
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Department of OrthopedicsJingjiang People's HospitalSeventh Clinical Medical School of Yangzhou UniversityJingjiangJiangsu ProvinceChina
| | - Jiaxiang Bai
- Department of OrthopedicsCentre for Leading Medicine and Advanced Technologies of IHMDivision of Life Sciences and MedicineThe First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Dechun Geng
- Department of OrthopedicsThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|