1
|
Raffaele B, Nicola M, Cinzia R, Valeria R, Paolo CF, Addolorata C. Mechanisms of ossification of the entheses in spondyloarthritis physiopathogenic aspects and possible therapeutic implication. Tissue Cell 2025; 94:102803. [PMID: 39983384 DOI: 10.1016/j.tice.2025.102803] [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: 10/24/2024] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/23/2025]
Abstract
This review examines the molecular mechanisms driving structural damage in Spondyloarthritis (SpA), a chronic inflammatory condition characterized by new bone formation that can lead to partial or complete spinal ankylosis. We explore the complex interplay between inflammation, mechanical stress, and bone metabolism in SpA, focusing on key signaling pathways and cytokines that contribute to disease progression. The review analyzes both structural and inflammatory aspects, particularly the role of enthesis biology and the impact of mechanical factors. Additionally, we assess how current therapeutic approaches, including biologic treatments targeting specific inflammatory pathways such as tumor necrosis factor inhibitors, affect disease progression. While these treatments can reduce inflammation and manage clinical symptoms, their limited ability to completely prevent new bone formation highlights the complexity of the underlying pathological processes. We also evaluate emerging therapeutic strategies targeting specific molecular pathways involved in bone formation. Understanding these intricate molecular mechanisms and their interactions is crucial for developing more effective targeted therapies that could potentially not only manage symptoms but also prevent or reverse structural damage in SpA patients.
Collapse
Affiliation(s)
- Barile Raffaele
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy.
| | - Maruotti Nicola
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy.
| | - Rotondo Cinzia
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy.
| | - Rella Valeria
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy.
| | - Cantatore Francesco Paolo
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy.
| | - Corrado Addolorata
- Rheumatology Clinic, Department of Medical and Surgical Sciences, University of Foggia, Foggia 71122, Italy.
| |
Collapse
|
2
|
Fan J, Wang Y, Yang H, Huang S, Ma Y, Guo J, Jia Y, Zhang Y, Fan Y, Xiao D, Zhang J, Li J, Dong Y, Zhao Y, Guo M, Tang Q, Li SS, Sun T, Jin X. Protosappanin B activates the Wnt pathway to protect against glucocorticoid-induced osteoblast inhibition and enhance bone formation. Chem Biol Interact 2025; 410:111436. [PMID: 39986363 DOI: 10.1016/j.cbi.2025.111436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Osteoporosis remains a major health challenge due to impaired osteoblast function and reduced bone formation, particularly in glucocorticoid-induced osteoporosis (GIOP). The Wnt/β-catenin signaling pathway plays a critical role in osteogenesis, making it a promising target for protective interventions against osteoporosis-related bone loss. In this study, virtual screening of a natural product library identified Protosappanin B (PB) as a potential Wnt pathway activator with high binding affinity for Wnt receptors. We investigated PB's protective effects on osteoblast function under glucocorticoid exposure using MC3T3-E1 cells treated with dexamethasone (DEX) and an in vivo zebrafish model of GIOP. PB significantly promoted osteoblast proliferation, facilitated cell cycle progression, and attenuated DEX-induced apoptosis in a dose-dependent manner. Additionally, PB enhanced osteoblast differentiation and mineralization, counteracting DEX's inhibitory effects on alkaline phosphatase (ALP) activity and calcium deposition. In zebrafish, PB mitigated DEX-induced skeletal defects, improving bone and craniofacial cartilage formation. Western blot analysis confirmed that PB restored β-catenin levels, activating the Wnt/β-catenin pathway. Notably, the osteogenic effects of PB were abolished by XAV939, a Wnt signaling inhibitor, further supporting its Wnt-dependent mechanism of action. These findings indicate that PB provides protective effects against glucocorticoid-induced osteoblast dysfunction and bone loss by modulating Wnt signaling. This study highlight the potential of PB as a natural agent for preventing GIOP-related bone deterioration and warrants further investigation into its clinical applicability.
Collapse
Affiliation(s)
- Jigeng Fan
- Tianjin Medical University, Tianjin, China
| | - Yahui Wang
- Tianjin Medical University, Tianjin, China
| | | | | | - Yuan Ma
- NanYang Central Hospital, NanYang, China
| | - Jie Guo
- Tianjin Medical University, Tianjin, China
| | - YuTao Jia
- Department of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Ying Zhang
- Tianjin Key Specialty of Integrated Traditional Chinese and Western Medicine, Tianjin Institute of Rehabilitation, Tianjin Union Medical Center, Tianjin, China
| | - Yonggang Fan
- School of Medicine, Nankai University, Tianjin, China
| | | | | | - Jianwei Li
- School of Medicine, Nankai University, Tianjin, China
| | - Yu Dong
- School of Medicine, Nankai University, Tianjin, China
| | - Ying Zhao
- School of Medicine, Nankai University, Tianjin, China
| | - Miao Guo
- School of Medicine, Nankai University, Tianjin, China
| | - Qiong Tang
- Department of Respiratory, Tianjin Union Medical Center, Tianjin, China
| | - Shan-Shan Li
- School of Medicine, Nankai University, Tianjin, China.
| | - Tianwei Sun
- Department of Spinal Surgery, Tianjin Union Medical Center, Tianjin, China.
| | - Xin Jin
- School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
3
|
Najary S, Nokhbatolfoghahaei H, Khojasteh A. The effect of Hypoxia-Inducible Factor-1a stabilization on bone regeneration during distraction osteogenesis: A systematic review of animal studies. Arch Oral Biol 2025; 172:106184. [PMID: 39893997 DOI: 10.1016/j.archoralbio.2025.106184] [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: 09/15/2024] [Revised: 01/07/2025] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
OBJECTIVE This systematic review described Hypoxia-Inducible Factor-1a stabilization or upregulation approaches along with underlying signaling pathways and assessed bone regeneration, angiogenesis, and consolidation time during DO in animal models. DESIGN A comprehensive and systematic search of electronic databases including PubMed, Scopus, and ScienceDirect was performed till December 26, 2023. The search was limited to English articles, and no time restrictions were applied. RESULTS A total of 14 studies met the inclusion criteria and were included for final review. Four methods have been shown to activate the HIF pathway including genetic, pharmacological, mechanical, and cell preconditioning approaches. Deferoxamine (DFO) was administered as a pharmacological hypoxia-mimicking agent in many studies reporting acceptable outcomes on bone regeneration and acceleration of bone consolation. Applying mechanical loads at the optimal rate and amplitude serves as a minimally invasive approach with acceptable results. HIF-related signaling pathways increase osteogenesis and angiogenesis during DO, potentially through VHL/HIF-1a/VEGF, Wnt/β-catenin, and Mesenchymal-Epithelial transition (MET) signaling pathways. CONCLUSION Activation of HIF-related signaling pathways enhances and accelerates bone regeneration during the consolidation phase of distraction osteogenesis. The most feasible approach with the least side effects must be selected for further clinical studies.
Collapse
Affiliation(s)
- Shaghayegh Najary
- Student Research Committee, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hanieh Nokhbatolfoghahaei
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arash Khojasteh
- Dental Research Center, Research Institute of Dental Sciences, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Cranio-Maxillofacial Surgery, University Hospital, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
4
|
Foroutani MR, Salamat MR, Bagherzadeh S, Keshtkar M, Khoshhali M, Asgari M. The role of trunk region body composition in lumbar spine bone mineral content and density. J Clin Densitom 2025; 28:101580. [PMID: 40209294 DOI: 10.1016/j.jocd.2025.101580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 02/22/2025] [Accepted: 03/07/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND The aim of this study was to investigate the differential effects of trunk region body composition-specifically fat mass (FM) and lean mass (LM)-on lumbar spine bone mineral density (BMD) and bone mineral content (BMC), with a focus on variations by gender and menopausal status. MATERIALS AND METHODS We identified 331 adult patients (69 men, 161 premenopausal women, and 101 postmenopausal women) who underwent dual-energy X-ray absorptiometry (DXA) to simultaneously measure trunk body composition and lumbar spine BMD (L2-L4). The Pearson correlation coefficient was used to assess the linear relationships between body composition components and lumbar spine BMD and BMC, stratified by gender and menopausal status. Additionally, multiple linear regression analysis with the forward stepwise elimination procedure was applied to find a reasonable subset of predictor variables. RESULT The highest correlation coefficients between body composition and BMD were seen for men and post- and premenopausal individuals in the following areas: pelvic fat mass (r = 0.325; P < 0.01), chest lean mass (r = 0.260; P < 0.01), and total lean mass (r = 0.312; P < 0.01), respectively. Additionally, total lean mass (r = 0.477; P < 0.01), chest lean mass (r = 0.360; P < 0.01), and total lean mass (r = 0.459; P < 0.01) had the strongest correlation coefficients between body composition and BMC. Forward stepwise regression identified age, chest lean mass, and midriff fat mass as predictors of BMC in postmenopausal women; BMI and total lean mass in premenopausal women; and total lean mass in men. For BMD, chest lean mass (postmenopausal), total lean mass (premenopausal), and BMI (men) were significant predictors. CONCLUSIONS Fat mass does not affect BMD or BMC, while lean mass, strongly predicts bone health. Trunk body composition showed varying relationships with BMD and BMC, making it challenging to pinpoint how lean mass distribution in the chest and midriff specifically impacts bone health.
Collapse
Affiliation(s)
- Mohammad Reza Foroutani
- Department of Nursing, School of Nursing, Larestan University of Medical sciences, Larestan, Iran
| | - Mohammad Reza Salamat
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sakineh Bagherzadeh
- Department of Medical Physics, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Keshtkar
- Department of Medical Physics and Radiology, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Mehri Khoshhali
- Limited Liability Company of Dade Azma Salamat Pajooh, Shiraz, Iran
| | - Mahdi Asgari
- Department of Medical Physics, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
5
|
Ahamad S, Saquib M, Hussain MK, Bhat SA. Targeting Wnt signaling pathway with small-molecule therapeutics for treating osteoporosis. Bioorg Chem 2025; 156:108195. [PMID: 39864370 DOI: 10.1016/j.bioorg.2025.108195] [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: 10/03/2024] [Revised: 11/29/2024] [Accepted: 01/17/2025] [Indexed: 01/28/2025]
Abstract
Small molecules are emerging as potential candidates for treating osteoporosis by activating canonical Wnt signaling. These candidates work either by inhibiting DKK-1, sclerostin, SFRP-1, NOTUM, and S1P lyase or by preventing β-catenin degradation through inhibition of GSK-3β, or by targeting Dvl-CXXC5 and axin/β-catenin interactions. While many of these anti-osteoporotic small molecules are in preclinical development, the paucity of FDA-approved small molecules, or promising candidates, that have progressed to clinical trials for treating bone disorders through this mechanism poses a challenge. Despite advancements in computer-aided drug design, it is rarely employed for designing Wnt signaling activators to treat osteoporosis, and high-throughput screen (HTS) remains the primary method for discovering initial hits. Acknowledging the promising therapeutic potential of these compounds in addressing bone diseases, this review underscores the need for further mechanistic elucidation to enhance our understanding of their applications. Additionally, caution must be exercised in the design of small molecule-based Wnt activators due to their association with oncological risks.
Collapse
Affiliation(s)
- Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University Aligarh 202002 India.
| | - Mohammad Saquib
- Department of Chemistry, University of Allahabad, Prayagraj (Allahabad) 211002, UP, India; Department of Chemistry, G. R. P. B. Degree College, P. R. S. University, Prayagraj (Allahabad) 211010, UP, India
| | | | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
6
|
Abo-Elenin MHH, Kamel R, Nofal S, Ahmed AAE. The crucial role of beta-catenin in the osteoprotective effect of semaglutide in an ovariectomized rat model of osteoporosis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2677-2693. [PMID: 39254876 PMCID: PMC11920005 DOI: 10.1007/s00210-024-03378-z] [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: 04/26/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024]
Abstract
Postmenopausal osteoporosis is a common chronic medical illness resulting from an imbalance between bone resorption and bone formation along with microarchitecture degeneration attributed to estrogen deficiency and often accompanied by other medical conditions such as weight gain, depression, and insomnia. Semaglutide (SEM) is a recently introduced GLP-1 receptor agonist (GLP-1RA) for the treatment of obesity and type 2 diabetes mellitus by mitigating insulin resistance. It has been discovered that the beneficial effects of GLP-1 are associated with alterations in lipolysis, adipogenesis, and anti-inflammatory processes. GLP-1 analogs transmit signals directly to adipose tissue. Mesenchymal stem cells (MSCs) are multidisciplinary cells that originate from bone marrow, migrate to injury sites, and promote bone regeneration. MSCs can differentiate into osteoblasts, adipose cells, and cartilage cells. Our aim is to investigate the role of semaglutide on bone formation and the Wnt signaling pathway. Osteoporosis was induced in female rats by ovariectomy, and the ovariectomized rats were treated with alendronate as standard treatment with a dose of 3 mg/kg orally and semaglutide with two doses (150 mcg/kg and 300 mcg/kg) S.C. for 10 successive weeks. Semaglutide ameliorates bone detrimental changes induced by ovariectomy. It improves bone microarchitecture and preserves bone mineral content. Semaglutide ameliorates ovariectomy-induced osteoporosis and increases the expression of β-catenin, leading to increased bone formation and halted receptor activator of nuclear factor kappa-Β ligand (RANKL's) activation. Semaglutide can be used as a potential prophylactic and therapeutic drug against osteoporosis, possibly by activating Wnt signaling and decreasing bone resorption.
Collapse
Affiliation(s)
| | - Rehab Kamel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo City, Egypt
| | - Shahira Nofal
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo City, Egypt
| | - Amany Ali Eissa Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo City, Egypt
| |
Collapse
|
7
|
Akbar A, Zaheer A, Kharal MM, Komel A, Khan MH, Ahsan A, Singh AK. Evolving strategies for osteoporosis management in postmenopausal women: From tradition to innovation. Medicine (Baltimore) 2025; 104:e41605. [PMID: 39960896 PMCID: PMC11835067 DOI: 10.1097/md.0000000000041605] [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: 06/17/2024] [Accepted: 10/23/2024] [Indexed: 02/20/2025] Open
Abstract
Osteoporosis is a chronic condition primarily affecting postmenopausal women, significantly impacting their well-being and quality of life. Traditional treatment approaches include medications, vitamins, and exercise, but there is a growing interest in alternative therapies that enhance bone health. This review was conducted by searching multiple databases, including PubMed, Medline, and Google Scholar, for studies related to osteoporosis treatment. Articles focusing on both traditional therapies such as bisphosphonates, calcium, and vitamin D supplementation, and newer advancements like vibration therapy and bone-building devices such as Osteoboost were included. Traditional treatments, such as vitamin supplementation, exercise, and bisphosphonates, remain foundational in osteoporosis management, helping to maintain bone density and reduce fracture risks. Recent developments, including vibration therapy and Osteoboost, show promising results in bone regeneration without the use of medication. While traditional therapies continue to play an essential role, advancements like vibration therapy present novel alternatives for managing osteoporosis. Further research is necessary to optimize these approaches, ensuring they maximize benefits while minimizing risks, ultimately improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Anum Akbar
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE
| | - Amna Zaheer
- Liaquat National Hospital and Medical College, Karachi, Pakistan
| | | | - Aqsa Komel
- Nishtar Medical University, Multan, Pakistan
| | | | - Areeba Ahsan
- Foundation University Medical College, Islamabad, Pakistan
| | | |
Collapse
|
8
|
Chen R, Yang C, Xiao H, Yang A, Chen C, Yang F, Peng B, Geng B, Xia Y. PRKD2 as a novel target for targeting the diabetes-osteoporosis nexus. Sci Rep 2025; 15:4703. [PMID: 39922871 PMCID: PMC11807170 DOI: 10.1038/s41598-025-89235-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/04/2025] [Indexed: 02/10/2025] Open
Abstract
Diabetes mellitus (DM) and osteoporosis (OP) co-morbidity (DMOP) pose major health challenges owing to their complex pathophysiological interactions. The aim of this study was to identify and validate key genes implicated in the pathogenesis of both conditions. By employing the Mfuzz time-series gene clustering method combined with transcriptome sequencing of patient serum, we systematically delineated gene expression patterns during the transition from a healthy state through DM to DMOP. These findings were further validated using external datasets, and a series of functional enrichment analyses, gene set enrichment analyses, and immune cell infiltration studies were conducted. Our analyses revealed a distinct progression pattern from a normal state through DM to DMOP, characterized by dynamic gene expression changes. Notably, PRKD2 emerged as a significantly downregulated gene in DMOP, highlighting its crucial role in disease pathogenesis. Further analyses revealed the involvement of PRKD2 in key signaling pathways, especially the Wnt and IL-18 pathways, which are critical for bone and glucose metabolism. Validation in cellular and animal models confirmed the role of PRKD2 in apoptosis and bone metabolism, emphasizing its therapeutic potential. In conclusion, our findings establish PRKD2 as a pivotal molecule in DMOP, offering fresh insights into its mechanisms and affirming its value as a therapeutic target.
Collapse
Affiliation(s)
- Rongjin Chen
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou, 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
- Department of Orthopedics, Tianshui Hand and Foot Surgery Hospital, Tianshui, 741000, China
| | - Chenhui Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou, 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
- Department of Orthopedics, Tianshui Hand and Foot Surgery Hospital, Tianshui, 741000, China
| | - Hefang Xiao
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou, 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Ao Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou, 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Changshun Chen
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou, 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Fei Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou, 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Bo Peng
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou, 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Bin Geng
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, 730030, China
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou, 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Yayi Xia
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou, 730030, China.
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou, 730030, China.
- The Second Clinical Medical School, Lanzhou University, Lanzhou, 730030, China.
| |
Collapse
|
9
|
Hu X, Lei X, Lin W, Li X, Zhong W, Luo B, Xie J, Liang Z, Li Y, Qiu J, Wang P, Zhu X, Zhang R, Yang L. Quercetin promotes osteogenic differentiation of bone marrow mesenchymal stem cells by modulating the miR-214-3p/Wnt3a/β-catenin signaling pathway. Exp Cell Res 2025; 444:114386. [PMID: 39694404 DOI: 10.1016/j.yexcr.2024.114386] [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: 10/14/2024] [Revised: 12/12/2024] [Accepted: 12/15/2024] [Indexed: 12/20/2024]
Abstract
Postmenopausal osteoporosis, primarily driven by estrogen deficiency, is predominantly mediated through estrogen receptors such as ERα. However, the underlying mechanisms necessitate further investigation. In this study, we established an ERα-deficient model in rBMSCs to elucidate the role of ERα in osteogenic differentiation and miRNA expression profiles. Our findings demonstrate that knockdown of ERα inhibits osteogenic differentiation in rBMSCs, resulting in upregulation of 25 miRNAs and downregulation of 184 miRNAs, including a significant increase in the expression of miR-214-3p. Validation using qPCR, Western blotting, and bioinformatics analysis revealed that miR-214-3p negatively regulates osteogenic differentiation via the Wnt/β-catenin signaling pathway. Furthermore, we explored the potential therapeutic effects of quercetin (QUE) on rBMSCs. CCK8, alkaline phosphatase activity assays, and Alizarin Red staining demonstrated that QUE dose-dependently enhances rBMSCs proliferation, alkaline phosphatase activity, and mineralization within the concentration range of 0.1-1 μM. Importantly, QUE was found to downregulate miR-214-3p expression and activate the Wnt3a/β-catenin signaling pathway. Rescue experiments confirmed that QUE could counteract the inhibitory effects of miR-214-3p on the Wnt3a/β-catenin signaling pathway. Collectively, our study provides compelling evidence that knockdown of ERα inhibits the osteogenic differentiation of rBMSCs by affecting the miRNA expression profile, while QUE can reverse the inhibitory effect exerted by miR-214-3p on the Wnt3a/β-catenin signaling pathway, thereby offering novel insights into diagnosis, prevention, and treatment strategies for postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Xueling Hu
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China
| | - Xiaotong Lei
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China
| | - Weiwen Lin
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China
| | - Xiaoyun Li
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China
| | - Wenqiang Zhong
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China
| | - Bingjie Luo
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China
| | - Ji Xie
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China
| | - Ziwen Liang
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China
| | - Yunchuan Li
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China
| | - Jingli Qiu
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China
| | - Panpan Wang
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China; College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Xiaofeng Zhu
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China; College of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Ronghua Zhang
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China.
| | - Li Yang
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Informatization, Guangzhou, 510632, China.
| |
Collapse
|
10
|
Hong S, Lee HJ, Jung DS, Erdenebileg S, Hwang H, Kwon HC, Kwon J, Yoo G. Exploring the Anti-Osteoporotic Effects of n-Hexane Fraction from Cotoneaster wilsonii Nakai: Activation of Runx2 and Osteoblast Differentiation In Vivo. Pharmaceuticals (Basel) 2025; 18:45. [PMID: 39861108 PMCID: PMC11768920 DOI: 10.3390/ph18010045] [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: 11/19/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Osteoporosis is characterized by the microstructural depletion of bone tissue and decreased bone density, leading to an increased risk of fractures. Cotoneaster wilsonii Nakai, an endemic species of the Korean Peninsula, grows wild in Ulleungdo. In this study, we aimed to investigate the effects of C. wilsonii and its components on osteoporosis. METHODS AND RESULTS The alkaline phosphatase (ALP) activity of C. wilsonii extracts and fractions was evaluated in MC3T3-E1 pre-osteoblasts, and the n-hexane fraction (CWH) showed the best properties for ALP activity. The effects of the CWH on bone formation were assessed in MC3T3-E1 cells and ovariectomized mice. Biochemical assays and histological analyses focused on the signaling activation of osteoblast differentiation and osteogenic markers, such as ALP, collagen, and osterix. The CWH significantly activated TGF-β and Wnt signaling, enhancing osteoblast differentiation and bone matrix formation. Notably, CWH treatment improved micro-CT indices, such as femoral bone density, and restored serum osteocalcin levels compared to OVX controls. CONCLUSIONS These results highlight the potential of the C. wilsonii Nakai n-hexane fraction as a promising therapeutic agent for managing osteoporosis.
Collapse
Affiliation(s)
- Soyeon Hong
- Smart Farm Research Center, Korean Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (S.H.); (D.S.J.); (S.E.)
| | - Hee Ju Lee
- Center for Natural Product Systems Biology, Korean Institute of Science and Technology, Gangneung 25451, Republic of Korea; (H.J.L.); (H.H.); (H.C.K.)
| | - Da Seul Jung
- Smart Farm Research Center, Korean Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (S.H.); (D.S.J.); (S.E.)
| | - Saruul Erdenebileg
- Smart Farm Research Center, Korean Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (S.H.); (D.S.J.); (S.E.)
| | - Hoseong Hwang
- Center for Natural Product Systems Biology, Korean Institute of Science and Technology, Gangneung 25451, Republic of Korea; (H.J.L.); (H.H.); (H.C.K.)
| | - Hak Cheol Kwon
- Center for Natural Product Systems Biology, Korean Institute of Science and Technology, Gangneung 25451, Republic of Korea; (H.J.L.); (H.H.); (H.C.K.)
| | - Jaeyoung Kwon
- Center for Natural Product Systems Biology, Korean Institute of Science and Technology, Gangneung 25451, Republic of Korea; (H.J.L.); (H.H.); (H.C.K.)
| | - Gyhye Yoo
- Smart Farm Research Center, Korean Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (S.H.); (D.S.J.); (S.E.)
- Department of Natural Product Applied Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
11
|
Sanjaya SS, Park J, Choi YH, Park HS, Sadanaga T, Jung MJ, Kim GY. Polyphenol extract from Tagetes erecta L. flowers stimulates osteogenesis via β-catenin activation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156313. [PMID: 39675109 DOI: 10.1016/j.phymed.2024.156313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/26/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Osteoporosis, a prevalent bone disorder, results in reduced bone mineral density and mass. With minimal side effects, medicinal plant-based natural remedies are increasingly explored for osteoporosis. However, the osteogenic potential of Tagetes erecta L. flower, traditionally used for cardiovascular and renal diseases, has not yet been studied. OBJECTIVE This study investigates the osteogenic effects of the polyphenol-enriched extract from T. erecta L. flowers (TE) and its main components on osteoblast differentiation, with an emphasis on anti-osteoporotic activity. METHODS The osteogenic activity of TE was assessed in MC3T3-E1 preosteoblast cells, analyzing osteogenic alkaline phosphatase (ALP) activity via a colorimetric assay and mineralization through Alizarin Red S staining over 14 d. Expression levels of osteogenic markers-transcription factor osterix (SP7), runt-related transcription factor 2 (RUNX2), and ALP-were quantified through quantitative reverse transcription-polymerase chain reaction and western blotting. In vivo effects were evaluated using zebrafish larvae for bone formation and anti-osteoporotic properties. Vertebral development was visualized by staining mineralized structures with calcein or Alizarin Red S. Prednisolone (PDS) was administered to zebrafish larvae to model osteoporosis. Furthermore, molecular docking simulations were conducted to assess the binding affinity of TE components to the ATP-binding pocket of glycogen synthase kinase-3β (GSK-3β), and their inhibitory potential on GSK-3β kinase activity was quantified by in vitro kinase assays. Cellular thermal shift assay (CETSA) was performed to monitor direct bindings of TE and its main components to GSK3-3β. RESULTS TE promoted vertebral and cranial bone formation in zebrafish larvae, elevating key osteogenic genes, such as sp7, runx2a, runx2b, and alpl. Among TE components, kaempferol and patuletin significantly enhanced vertebral formation, while isorhamnetin showed moderate effects. Patulitrin and quercetagetin did not increased vertebral formation. In MC3T3-E1 cells, TE increased ALP activity, mineralization, and the expression of SP7, RUNX2, and ALP. It also induced GSK-3β phosphorylation at serine 9 and promoted β-catenin nuclear translocation. Inhibition of β-catenin signaling reversed TE-induced osteogenic effects. Molecular docking suggested strong GSK-3β binding by TE components, with patuletin showing notable inhibition GSK-3β activity (half-maximal inhibitory concentration = 379.3 ng/mL) and enhancing vertebral formation. CETSA confirmed that TE and its main components, kaempferol and patuletin, degrades GSK-3β. Additionally, TE alleviated PDS-induced osteoporosis in both cellular and zebrafish models. CONCLUSION By targeting GSK-3β and activating β-catenin-mediated pathways, TE shows promise as a novel anti-osteoporotic agent. This study highlights the potential of TE for therapeutic use in bone health, warranting further clinical trials to confirm its applicability.
Collapse
Affiliation(s)
| | - Jinkuk Park
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Republic of Korea
| | - Hee Sun Park
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Takayuki Sadanaga
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Min-Jeong Jung
- GENE & BIO, 145 Sindae-ro, Jeju 63134, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
12
|
Wang J, Zhang Y, Wang S, Wang X, Jing Y, Su J. Bone aging and extracellular vesicles. Sci Bull (Beijing) 2024; 69:3978-3999. [PMID: 39455324 DOI: 10.1016/j.scib.2024.10.013] [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: 04/03/2024] [Revised: 08/01/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Bone aging, a major global health concern, is the natural decline in bone mass and strength. Concurrently, extracellular vesicles (EVs), tiny membrane-bound particles produced by cells, have gained recognition for their roles in various physiological processes and age-related diseases. The interaction between EVs and bone aging is of growing interest, particularly their effects on bone metabolism, which become increasingly critical with advancing age. In this review, we explored the biology, types, and functions of EVs and emphasized their regulatory roles in bone aging. We examined the effects of EVs on bone metabolism and highlighted their potential as biomarkers for monitoring bone aging progression. Furthermore, we discussed the therapeutic applications of EVs, including targeted drug delivery and bone regeneration, and addressed the challenges associated with EV-based therapies, including the technical complexities and regulatory issues. We summarized the current research and clinical trials investigating the role of EVs in bone aging and suggested future research directions. These include the potential for personalized medicine using EVs and the integration of EV research with advanced technologies to enhance the management of age-related bone health. This analysis emphasized the transformative potential of EVs in understanding and managing bone aging, thereby marking a significant advancement in skeletal health research.
Collapse
Affiliation(s)
- Jian Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; School of Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China
| | - Yuanwei Zhang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China
| | - Sicheng Wang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China
| | - Xinglong Wang
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, AZ 85721, USA.
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China.
| | - Jiacan Su
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Trauma Orthopedics Center, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
13
|
Sapra L, Srivastava RK. Immunotherapy in the management of inflammatory bone loss in osteoporosis. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 144:461-491. [PMID: 39978975 DOI: 10.1016/bs.apcsb.2024.10.013] [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: 02/22/2025]
Abstract
Osteoporosis, a progressive skeletal disorder characterized by decreased bone mass and increased fracture risk, has traditionally been treated with pharmacological agents targeting bone remodeling. However, emerging research highlights the critical role of immune system in regulating bone metabolism, introducing the concept of Osteoimmunology. Chronic low-grade inflammation is now recognized as a significant contributor to osteoporosis, particularly in postmenopausal women and the elderly. Immune cells, such as T cells and B cells, and their secreted cytokines directly influence bone resorption and formation, tipping the balance toward net bone loss in inflammatory environments. Immunotherapy, a treatment modality traditionally associated with cancer and autoimmune diseases, is now gaining attention in the management of osteoporosis. By targeting immune dysregulation and reducing inflammatory bone loss, immunotherapies offer a novel approach to treating osteoporosis that goes beyond merely inhibiting bone resorption or promoting bone formation. This therapeutic strategy includes monoclonal antibodies targeting inflammatory cytokines, cell-based therapies to enhance the function of regulatory T and B cells, and interventions aimed at modulating immune pathways linked to bone health. This chapter reviews the emerging role of immunotherapy in addressing inflammatory bone loss in osteoporosis. Present chapter also explores the underlying immune mechanisms contributing to bone degradation, current immunotherapeutic strategies under investigation, and the potential of these approaches to revolutionize the management of osteoporosis.
Collapse
Affiliation(s)
- Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| |
Collapse
|
14
|
Yeh TT, Chen CK, Kuthati Y, Mende LK, Wong CS, Kong ZL. Amorphous Calcium Carbonate Enhances Fracture Healing in a Rat Fracture Model. Nutrients 2024; 16:4089. [PMID: 39683484 DOI: 10.3390/nu16234089] [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: 09/11/2024] [Revised: 10/25/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Delayed and failed fracture repair and bone healing remain significant public health issues. Dietary supplements serve as a safe, inexpensive, and non-surgical means to aid in different stages of fracture repair. Studies have shown that amorphous calcium carbonate (ACC) is absorbed 2 to 4.6 times more than crystalline calcium carbonate in humans. Objectives: In the present study, we assessed the efficacy of ACC on femoral fracture healing in a male Wistar rat model. Methods: Eighty male Wistar rats were randomly divided into five groups (n = six per group): sham, fracture + water, fracture + 0.5× (206 mg/kg) ACC, fracture + 1× ACC (412 mg/kg), and fracture + 1.5× (618 mg/kg) ACC, where ACC refers to the equivalent supplemental dose of ACC for humans. A 21-gauge needle was placed in the left femoral shaft, and we then waited for three weeks. After three weeks, the sham group of rats was left without fractures, while the remaining animals had their left mid-femur fractured with an impactor, followed by treatment with different doses of oral ACC for three weeks. Weight-bearing capacity, microcomputed tomography, and serum biomarkers were evaluated weekly. After three weeks, the rats were sacrificed, and their femur bones were isolated to conduct an evaluation of biomechanical strength and histological analysis. Results: Weight-bearing tests showed that treatment with ACC at all the tested doses led to a significant increase in weight-bearing capacity compared to the controls. In addition, microcomputed tomography and histological studies revealed that ACC treatment improved callus formation dose-dependently. Moreover, biomechanical strength was improved in a dose-dependent fashion in ACC-treated rats compared to the controls. In addition, supplementation with ACC significantly lowered bone formation and resorption marker levels two-three weeks post-fracture induction, indicating accelerated fracture recovery. Conclusions: Our preliminary data demonstrate that ACC supplementation improves fracture healing, with ACC-supplemented rats healing in a shorter time than control rats.
Collapse
Affiliation(s)
- Tsu-Te Yeh
- Department of Orthopedic Surgery, Tri-Service General Hospital and National Defense Medical Center, 325 Cheng-Kung Road, Section 2, Taipei 114, Taiwan
| | - Chun-Kai Chen
- Department of Food Science, National Taiwan Ocean University, Keelung 202301, Taiwan
| | - Yaswanth Kuthati
- Department of Anesthesiology, Cathay General Hospital, Taipei 106, Taiwan
| | - Lokesh Kumar Mende
- Department of Anesthesiology, Cathay General Hospital, Taipei 106, Taiwan
| | - Chih-Shung Wong
- Department of Anesthesiology, Cathay General Hospital, Taipei 106, Taiwan
- National Defense Medical Center, Institute of Medical Sciences, Taipei 114, Taiwan
| | - Zwe-Ling Kong
- Department of Food Science, National Taiwan Ocean University, Keelung 202301, Taiwan
| |
Collapse
|
15
|
Spigel DR, Wang JS, Pronk L, Muskens B, Teufel M, Bashir B, Burris H. A phase I dose escalation study of the LRP5 antagonist BI 905681 in patients with advanced and metastatic solid tumors. ESMO Open 2024; 9:103730. [PMID: 39617535 DOI: 10.1016/j.esmoop.2024.103730] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND The Wnt pathway is involved in proliferation and tissue homeostasis. Aberrant activation promotes cancer cell proliferation and survival. Inhibition of the low-density lipoprotein receptor-related protein 5/6 (LRP5/6) coreceptors that regulate Wnt signaling could prevent cancer cell proliferation. BI 905681 is a novel LRP5 antagonist that has demonstrated potent in vivo antitumor activity. PATIENTS AND METHODS This was a phase I, dose escalation study (NCT04147247) evaluating BI 905681 in patients with advanced solid tumors over two dosing schedules (schedule A: every 3 weeks, 3-week cycles and schedule B: every 2 weeks, 4-week cycles). The primary endpoint was the maximum tolerated dose (MTD) of BI 905681 and the number of patients experiencing adverse events (AEs). Other endpoints were pharmacokinetics, pharmacodynamics, and efficacy. RESULTS As a result of difficulties enrolling patients, the trial was terminated early and the MTD for schedule A could not be determined. Twenty-one patients received BI 905681 over five dose cohorts (schedule A: 1.0, 2.5, 5.0, 7.0, and 8.5 mg/kg). No patients received schedule B. No dose-limiting toxicities (DLTs) were reported during the MTD evaluation period. However, during the entire treatment period, two patients (9.5%) experienced a DLT of grade 1 C-telopeptide increase in the 5.0 and 8.5 mg/kg dose cohorts. The most frequent treatment-related AEs were diarrhea (23.8%), vomiting (23.8%), nausea (19.0%), and infusion-related reactions (IRRs; 14.3%). Despite premedication to mitigate IRRs, one patient experienced a grade 2 IRR. The pharmacokinetic profiles of BI 905681 were biphasic, with a rapid distribution phase in the beginning followed by a slower elimination phase. The objective response rate was 0%; 5 (23.8%) and 14 patients (66.7%) had a best overall response of stable disease and progressive disease, respectively. CONCLUSION BI 905681 has minimal efficacy in an unselected patient population and was generally well tolerated.
Collapse
Affiliation(s)
- D R Spigel
- Sarah Cannon Research Institute, Nashville
| | - J S Wang
- Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, USA
| | - L Pronk
- Boehringer Ingelheim España S.A., Madrid, Spain
| | - B Muskens
- Venn Life Sciences ED, Breda, the Netherlands
| | - M Teufel
- Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield
| | - B Bashir
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, USA.
| | - H Burris
- Sarah Cannon Research Institute, Nashville
| |
Collapse
|
16
|
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
|
17
|
Wei S, Wu Q, Cao C, Yang Z, Shi J, Huang J, He H, Lai Y, Li J. A mechanism of action-reflective, dual cell-based bioassay for determining the bioactivity of sclerostin-neutralizing antibodies. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100187. [PMID: 39389544 DOI: 10.1016/j.slasd.2024.100187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/12/2024]
Abstract
Osteoporosis is a major threat to the elderly worldwide. The Wnt signaling pathway plays a critical role in bone development and homeostasis. Sclerostin, a Wnt ligand inhibitor, competes with Wnt ligands for low-density lipoprotein receptor-related protein 5 or 6 (LRP5/6) on osteoblasts, thereby suppressing bone formation. Sclerostin-neutralizing monoclonal antibodies (mAbs) have emerged as a potential bone-forming therapy for osteoporosis. A cell-based bioassay which determines the relative activity of a product, related to its mechanism of action, is of great importance from drug discovery to quality control and batch release. Currently used cell-based bioassays for sclerostin-neutralizing mAbs usually use Wnt1 or Wnt3a to stimulate the Wnt pathway; sclerostin is a direct inhibitor of Wnt1 but not Wnt3a. Wnt1 is a highly hydrophobic protein that binds to the producing cell membrane and acts in a juxtacrine manner to stimulate the Wnt pathway in neighboring cells. Bioassays for drugs that induce Wnt1 signaling should be performed in a juxtacrine manner. Here, we present a mechanism of action-reflective, dual cell-based reporter gene assay. In this assay, Wnt1 producer cells are co-cultured with cells containing the Wnt reporter genes, Wnt1 on the producer cells activates the Wnt signaling pathway in the reporter cells that are in direct cell-to-cell contact, and sclerostin-neutralizing mAbs specifically and effectively antagonize the sclerostin-mediated Wnt reporter gene suppression. This bioassay demonstrates good specificity, accuracy, linearity, and precision and is suitable for quality control, stability testing, batch release, and biosimilarity assessment of sclerostin-neutralizing mAbs.
Collapse
Affiliation(s)
- Suzhen Wei
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Qiang Wu
- Zhuhai United Laboratories Co., Ltd, 2428 Anji Road, Zhuhai, Guangdong, China
| | - Chunlai Cao
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Zhuoni Yang
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Jianrui Shi
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Jingqun Huang
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Hua He
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China
| | - Yongjie Lai
- Department of Microbiology and Immunology, Zunyi Medical University (Zhuhai Campus), 368 Golden Coast Avenue, Zhuhai, Guangdong, China.
| | - Jing Li
- Zhuhai United Biopharma Co., Ltd, 399 Airport West Road, Zhuhai, Guangdong, China; Zhuhai United Laboratories Co., Ltd, 2428 Anji Road, Zhuhai, Guangdong, China.
| |
Collapse
|
18
|
Chen R, Jin Y, Lian R, Yang J, Liao Z, Jin Y, Deng Z, Feng S, Feng Z, Wei Y, Zhang Z, Zhao L. CRIP1 regulates osteogenic differentiation of bone marrow stromal cells and pre-osteoblasts via the Wnt signaling pathway. Biochem Biophys Res Commun 2024; 727:150277. [PMID: 38936225 DOI: 10.1016/j.bbrc.2024.150277] [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: 04/12/2024] [Revised: 06/02/2024] [Accepted: 06/17/2024] [Indexed: 06/29/2024]
Abstract
With the aging of the global demographic, the prevention and treatment of osteoporosis are becoming crucial issues. The gradual loss of self-renewal and osteogenic differentiation capabilities in bone marrow stromal cells (BMSCs) is one of the key factors contributing to osteoporosis. To explore the regulatory mechanisms of BMSCs differentiation, we collected bone marrow cells of femoral heads from patients undergoing total hip arthroplasty for single-cell RNA sequencing analysis. Single-cell RNA sequencing revealed significantly reduced CRIP1 (Cysteine-Rich Intestinal Protein 1) expression and osteogenic capacity in the BMSCs of osteoporosis patients compared to non-osteoporosis group. CRIP1 is a gene that encodes a member of the LIM/double zinc finger protein family, which is involved in the regulation of various cellular processes including cell growth, development, and differentiation. CRIP1 knockdown resulted in decreased alkaline phosphatase activity, mineralization and expression of osteogenic markers, indicating impaired osteogenic differentiation. Conversely, CRIP1 overexpression, both in vitro and in vivo, enhanced osteogenic differentiation and rescued bone mass reduction in ovariectomy-induced osteoporosis mice model. The study further established CRIP1's modulation of osteogenesis through the Wnt signaling pathway, suggesting that targeting CRIP1 could offer a novel approach for osteoporosis treatment by promoting bone formation and preventing bone loss.
Collapse
Affiliation(s)
- Ruge Chen
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yangchen Jin
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Ru Lian
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jie Yang
- Department of Chinese Medicine, Chinese People's Liberation Army Air Force Special Medical Center, Beijing, 100142, China
| | - Zheting Liao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yu Jin
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhonghao Deng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Shuhao Feng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zihang Feng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yiran Wei
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Liang Zhao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
19
|
Glorieux FH, Langdahl B, Chapurlat R, De Beur SJ, Sutton VR, Poole KES, Dahir KM, Orwoll ES, Willie BM, Mikolajewicz N, Zimmermann E, Hosseinitabatabaei S, Ominsky MS, Saville C, Clancy J, MacKinnon A, Mistry A, Javaid MK. Setrusumab for the treatment of osteogenesis imperfecta: 12-month results from the phase 2b asteroid study. J Bone Miner Res 2024; 39:1215-1228. [PMID: 39012717 PMCID: PMC11371902 DOI: 10.1093/jbmr/zjae112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 07/18/2024]
Abstract
Osteogenesis imperfecta (OI) is a rare genetic disorder commonly caused by variants of the type I collagen genes COL1A1 and COL1A2. OI is associated with increased bone fragility, bone deformities, bone pain, and reduced growth. Setrusumab, a neutralizing antibody to sclerostin, increased areal bone mineral density (aBMD) in a 21-week phase 2a dose escalation study. The phase 2b Asteroid (NCT03118570) study evaluated the efficacy and safety of setrusumab in adults. Adults with a clinical diagnosis of OI type I, III, or IV, a pathogenic variant in COL1A1/A2, and a recent fragility fracture were randomized 1:1:1:1 to receive 2, 8, or 20 mg/kg setrusumab doses or placebo by monthly intravenous infusion during a 12-mo treatment period. Participants initially randomized to the placebo group were subsequently reassigned to receive setrusumab 20 mg/kg open label. Therefore, only results from the 2, 8, and 20 mg/kg double-blind groups are presented herein. The primary endpoint of Asteroid was change in distal radial trabecular volumetric bone mineral density (vBMD) from baseline at month 12, supported by changes in high-resolution peripheral quantitative computed tomography micro-finite element (microFE)-derived bone strength. A total of 110 adults were enrolled with similar baseline characteristics across treatment groups. At 12 mo, there was a significant increase in mean (SE) failure load in the 20 mg/kg group (3.17% [1.26%]) and stiffness in the 8 (3.06% [1.70%]) and 20 mg/kg (3.19% [1.29%]) groups from baseline. There were no changes in radial trabecula vBMD (p>05). Gains in failure load and stiffness were similar across OI types. There were no significant differences in annualized fracture rates between doses. Two adults in the 20 mg/kg group experienced related serious adverse reactions. Asteroid demonstrated a beneficial effect of setrusumab on estimates of bone strength across the different types of OI and provides the basis for additional phase 3 evaluation.
Collapse
Affiliation(s)
- Francis H Glorieux
- Departments of Surgery, Pediatrics and Human Genetics, Shriners Hospitals for Children, McGill University, Montreal, Quebec H4A 0A9, Canada
| | - Bente Langdahl
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Middle Jutland 8200, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Middle Jutland 8200, Denmark
| | - Roland Chapurlat
- Inserm UMR 1033, Edouard Herriot Hospital, 69372 Lyon cedex 08, France
| | - Suzanne Jan De Beur
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Vernon Reid Sutton
- Department of Molecular & Human Genetics, Baylor College of Medicine & Texas Children’s Hospital, Houston, TX 77030, United States
| | - Kenneth E S Poole
- Department of Medicine & Cambridge NIHR Biomedical Research Centre, University of Cambridge, Cambridge CB3 0FA, United Kingdom
| | - Kathryn M Dahir
- Division of Endocrinology, Vanderbilt University Medical Center, Nashville, TN 37232, United States
| | - Eric S Orwoll
- Division of Endocrinology, Diabetes and Clinical Nutrition, School of Medicine, Oregon Health & Sciences University, Portland, OR 97239, United States
| | - Bettina M Willie
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal H3A 2T5, Canada
- Shriners Hospitals for Children, Montreal, Quebec H4A 0A9, Canada
| | - Nicholas Mikolajewicz
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal H3A 2T5, Canada
- Shriners Hospitals for Children, Montreal, Quebec H4A 0A9, Canada
| | - Elizabeth Zimmermann
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal H3A 2T5, Canada
- Shriners Hospitals for Children, Montreal, Quebec H4A 0A9, Canada
| | - Seyedmahdi Hosseinitabatabaei
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal H3A 2T5, Canada
- Shriners Hospitals for Children, Montreal, Quebec H4A 0A9, Canada
| | | | | | | | | | - Arun Mistry
- Mereo BioPharma, London W16 0QF, United Kingdom
| | - Muhammad K Javaid
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Wellington Square, Oxford OX1 2JD, United Kingdom
| |
Collapse
|
20
|
Naqvi SM, O’Sullivan LM, Allison H, Casey VJ, Schiavi-Tritz J, McNamara LM. Altered extracellular matrix and mechanotransduction gene expression in rat bone tissue following long-term estrogen deficiency. JBMR Plus 2024; 8:ziae098. [PMID: 39193115 PMCID: PMC11347883 DOI: 10.1093/jbmrpl/ziae098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 06/28/2024] [Accepted: 07/11/2024] [Indexed: 08/29/2024] Open
Abstract
Osteoporosis is primarily associated with bone loss, but changes in bone tissue matrix composition and osteocyte mechanotransduction have also been identified. However, the molecular mechanisms underlying these changes and their relation to bone loss are not fully understood. The objectives of this study were to (1) conduct comprehensive temporal gene expression analyses on cortical bone tissue from ovariectomized rats, with a specific focus on genes known to govern matrix degradation, matrix production, and mechanotransduction, and (2) correlate these findings with bone mass, trabecular and cortical microarchitecture, and mineral and matrix composition. Microarray data revealed 35 differentially expressed genes in the cortical bone tissue of the ovariectomized cohort. We report that catabolic gene expression abates after the initial accelerated bone loss period, which occurs within the first 4 wk of estrogen deficiency. However, in long-term estrogen deficiency, we report increased expression of genes associated with extracellular matrix deposition (Spp1, COL1A1, COL1A2, OCN) and mechanotransduction (Cx43) compared with age-matched controls and short-term estrogen deficiency. These changes coincided with increased heterogeneity of mineral-to-matrix ratio and collagen maturity, to which extracellular matrix markers COL1A1 and COL1A2 were positively correlated. Interestingly, mineral heterogeneity and collagen maturity, exhibited a negative correlation with PHEX and IFT88, associated with mechanosensory cilia formation and Hedgehog (Hh) signaling. This study provides the first insight into the underlying mechanisms governing secondary mineralization and heterogeneity of matrix composition of bone tissue in long-term estrogen deficiency. We propose that altered mechanobiological responses in long-term estrogen deficiency may play a role in these changes.
Collapse
Affiliation(s)
- Syeda Masooma Naqvi
- Mechanobiology and Medical Devices Research Group (MMDRG), Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, H91 HX31, Ireland
| | - Laura M O’Sullivan
- Mechanobiology and Medical Devices Research Group (MMDRG), Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, H91 HX31, Ireland
| | - Hollie Allison
- Mechanobiology and Medical Devices Research Group (MMDRG), Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, H91 HX31, Ireland
| | - Vincent J Casey
- Mechanobiology and Medical Devices Research Group (MMDRG), Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, H91 HX31, Ireland
| | - Jessica Schiavi-Tritz
- Mechanobiology and Medical Devices Research Group (MMDRG), Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, H91 HX31, Ireland
- University of Lorraine, CNRS, LRGP, F-54000 Nancy, France
| | - Laoise M McNamara
- Mechanobiology and Medical Devices Research Group (MMDRG), Biomedical Engineering, School of Engineering, College of Science and Engineering, University of Galway, Galway, H91 HX31, Ireland
| |
Collapse
|
21
|
Forte YS, Nascimento-Silva V, Andrade-Santos C, Ramos-Andrade I, Atella GC, Kraemer-Aguiar LG, Leal PRF, Renovato-Martins M, Barja-Fidalgo C. Unlocking the Secrets of Adipose Tissue: How an Obesity-Associated Secretome Promotes Osteoblast Dedifferentiation via TGF-β1 Signaling, Paving the Path to an Adipogenic Phenotype. Cells 2024; 13:1418. [PMID: 39272990 PMCID: PMC11394205 DOI: 10.3390/cells13171418] [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: 07/23/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Obesity poses a significant global health challenge, given its association with the excessive accumulation of adipose tissue (AT) and various systemic disruptions. Within the adipose microenvironment, expansion and enrichment with immune cells trigger the release of inflammatory mediators and growth factors, which can disrupt tissues, including bones. While obesity's contribution to bone loss is well established, the direct impact of obese AT on osteoblast maturation remains uncertain. This study aimed to explore the influence of the secretomes from obese and lean AT on osteoblast differentiation and activity. METHODS SAOS-2 cells were exposed to the secretomes obtained by culturing human subcutaneous AT from individuals with obesity (OATS) or lean patients, and their effects on osteoblasts were evaluated. RESULTS In the presence of the OATS, mature osteoblasts underwent dedifferentiation, showing an increased proliferation accompanied by a morphological shift towards a mesenchymal phenotype, with detrimental effects on osteogenic markers and the calcification capacity. Concurrently, the OATS promoted the expression of mesenchymal and adipogenic markers, inducing the formation of cytoplasmic lipid droplets in SAOS-2 cells exposed to an adipogenic differentiation medium. Additionally, TGF-β1 emerged as a key mediator of these effects, as the OATS was enriched with this growth factor. CONCLUSIONS Our findings demonstrate that obese subcutaneous AT promotes the dedifferentiation of osteoblasts and increases the adipogenic profile in these cells.
Collapse
Affiliation(s)
- Yasmin Silva Forte
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (Y.S.F.); (V.N.-S.); (C.A.-S.); (I.R.-A.)
| | - Vany Nascimento-Silva
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (Y.S.F.); (V.N.-S.); (C.A.-S.); (I.R.-A.)
| | - Caio Andrade-Santos
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (Y.S.F.); (V.N.-S.); (C.A.-S.); (I.R.-A.)
| | - Isadora Ramos-Andrade
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (Y.S.F.); (V.N.-S.); (C.A.-S.); (I.R.-A.)
| | - Georgia Correa Atella
- Institute of Medical Biochemistry, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;
| | - Luiz Guilherme Kraemer-Aguiar
- Obesity Unit, Multiuser Clinical Research Center (CePEM), Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (L.G.K.-A.); (P.R.F.L.)
| | - Paulo Roberto Falcão Leal
- Obesity Unit, Multiuser Clinical Research Center (CePEM), Hospital Universitário Pedro Ernesto, Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (L.G.K.-A.); (P.R.F.L.)
| | - Mariana Renovato-Martins
- Department of Molecular & Cellular Biology, Universidade Federal Fluminense, Rio de Janeiro 24020-141, Brazil;
| | - Christina Barja-Fidalgo
- Laboratory of Cellular & Molecular Pharmacology, Department of Cell Biology, Instituto de Biologia Roberto Alcantara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro, Rio de Janeiro 20551-030, Brazil; (Y.S.F.); (V.N.-S.); (C.A.-S.); (I.R.-A.)
| |
Collapse
|
22
|
Lai Z, Shu Q, Song Y, Tang A, Tian J. Effect of DNA methylation on the osteogenic differentiation of mesenchymal stem cells: concise review. Front Genet 2024; 15:1429844. [PMID: 39015772 PMCID: PMC11250479 DOI: 10.3389/fgene.2024.1429844] [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: 05/08/2024] [Accepted: 06/10/2024] [Indexed: 07/18/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have promising potential for bone tissue engineering in bone healing and regeneration. They are regarded as such due to their capacity for self-renewal, multiple differentiation, and their ability to modulate the immune response. However, changes in the molecular pathways and transcription factors of MSCs in osteogenesis can lead to bone defects and metabolic bone diseases. DNA methylation is an epigenetic process that plays an important role in the osteogenic differentiation of MSCs by regulating gene expression. An increasing number of studies have demonstrated the significance of DNA methyltransferases (DNMTs), Ten-eleven translocation family proteins (TETs), and MSCs signaling pathways about osteogenic differentiation in MSCs. This review focuses on the progress of research in these areas.
Collapse
Affiliation(s)
- Zhihao Lai
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qing Shu
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yue Song
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Ao Tang
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- College of Sports Medicine, Wuhan Sports University, Wuhan, China
| | - Jun Tian
- Department of Rehabilitation Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Cui W, Yang X, Dou Y, Du Y, Ma X, Hu L, Lin Y. Effects of tetrahedral DNA nanostructures on the treatment of osteoporosis. Cell Prolif 2024; 57:e13625. [PMID: 38414318 PMCID: PMC11216938 DOI: 10.1111/cpr.13625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/27/2024] [Accepted: 02/16/2024] [Indexed: 02/29/2024] Open
Abstract
Osteoporosis (OP) is a common disease characterized by bone loss and bone tissue microstructure degradation. Drug treatment is a common clinical treatment that aims to increase bone mass and bone density. Tetrahedral DNA nanostructures (TDNs) are three-dimensional tetrahedral frames formed by folding four single-stranded DNA molecules, which have good biological safety and can promote bone regeneration. In this study, a mouse model of OP was established by ovariectomy (OVX) and TDN was injected into the tail vein for 8 weeks. We found that ovariectomized mice could simulate some physiological changes in OP. After treatment with TDNs, some of this destruction in mice was significantly improved, including an increase in the bone volume fraction (BV/TV) and bone trabecular number (Tb. N), decrease in bone separation (Tb. SP), reduction in the damage to the mouse cartilage layer, reduction in osteoclast lacunae in bone trabecula, and reduction in the damage to the bone dense part. We also found that the expression of ALP, β-Catenin, Runx2, Osterix, and bone morphogenetic protein (BMP)2 significantly decreased in OVX mice but increased after TDN treatment. Therefore, this study suggests that TDNs may regulate the Wnt/β-Catenin and BMP signalling pathways to improve the levels of some specific markers of osteogenic differentiation, such as Runx2, ALP, and Osterix, to promote osteogenesis, thus showing a therapeutic effect on OP mice.
Collapse
Affiliation(s)
- Weitong Cui
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
| | - Xiao Yang
- Psychiatric Laboratory and Mental Health Center, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Yikai Dou
- Psychiatric Laboratory and Mental Health Center, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Yue Du
- Psychiatric Laboratory and Mental Health Center, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Xiaohong Ma
- Psychiatric Laboratory and Mental Health Center, the State Key Laboratory of BiotherapyWest China Hospital of Sichuan UniversityChengduChina
| | - Lei Hu
- Department of OrthopedicsSichuan Langsheng Brain Hospital & Shanghai Langsheng Brain Hospital Investment Co., Ltd.ChengduChina
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of StomatologySichuan UniversityChengduChina
- Sichuan Provincial Engineering Research Center of Oral BiomaterialsChengduChina
- National Center for Translational MedicineShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
24
|
Kulthanaamondhita P, Kornsuthisopon C, Chansaenroj A, Trachoo V, Manokawinchoke J, Samaranayake L, Srithanyarat SS, Osathanon T. MicroRNA expression in JAG1/Notch-activated periodontal ligament stem cells. BDJ Open 2024; 10:45. [PMID: 38839751 PMCID: PMC11153650 DOI: 10.1038/s41405-024-00232-5] [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: 03/12/2024] [Revised: 04/22/2024] [Accepted: 05/06/2024] [Indexed: 06/07/2024] Open
Abstract
OBJECTIVES The study explored the expression profile of miRNAs in Notch-activated periodontal ligament stem cells (PDLSCs) and examined their potential cellular targets. METHODS PDLSCs were cultured and treated with indirect immobilized Jagged1. The miRNA expression profile was examined using NanoString analysis. Bioinformatic analysis was performed together with enrichment, and miRNA expression was evaluated and validated using a quantitative polymerase chain reaction (qPCR). RESULTS A total of 26 miRNAs were differentially expressed in Jagged1 treated PDLSCs compared with the controls. Pathway analysis revealed that altered miRNAs were significantly associated with the transforming growth factor β (TGF-β) signaling pathway. Target prediction analysis demonstrated that 11,170 genes as predictable targets of these altered miRNAs. Enrichment of predicted target genes revealed that they were related to ErbB, Ras and MAPK signaling pathways and small GTPase transduction. CONCLUSIONS The research concludes that several miRNAs are differentially expressed in jagged-1 treated PDLSCs. In translational terms the differential functionality of these miRNAs offer promise for the development of targeted regenerative materials that are necessary for managing lost tissue replacement in periodontal diseases.
Collapse
Affiliation(s)
- Promphakkon Kulthanaamondhita
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chatvadee Kornsuthisopon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Ajjima Chansaenroj
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Vorapat Trachoo
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Jeeranan Manokawinchoke
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Lakshman Samaranayake
- Faculty of Dentistry, University of Hong Kong, Pok Fu Lam, 34, Hospital Road, Hong Kong
- Dean Office and Office of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Supreda Suphanantachat Srithanyarat
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
- Department of Periodontology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
- Center of Excellence for Periodontology and Dental Implants, Department of Periodontology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| | - Thanaphum Osathanon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
25
|
Sinder SB, Sharma SV, Shirvaikar IS, Pradhyumnan H, Patel SH, Cabeda Diaz I, Perez GG, Bramlett HM, Raval AP. Impact of menopause-associated frailty on traumatic brain injury. Neurochem Int 2024; 176:105741. [PMID: 38621511 DOI: 10.1016/j.neuint.2024.105741] [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: 02/03/2024] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024]
Abstract
Navigating menopause involves traversing a complex terrain of hormonal changes that extend far beyond reproductive consequences. Menopausal transition is characterized by a decrease in estradiol-17β (E2), and the impact of menopause resonates not only in the reproductive system but also through the central nervous system, musculoskeletal, and gastrointestinal domains. As women undergo menopausal transition, they become more susceptible to frailty, amplifying the risk and severity of injuries, including traumatic brain injury (TBI). Menopause triggers a cascade of changes leading to a decline in muscle mass, accompanied by diminished tone and excitability, thereby restricting the availability of irisin, a crucial hormone derived from muscles. Concurrently, bone mass undergoes reduction, culminating in the onset of osteoporosis and altering the dynamics of osteocalcin, a hormone originating from bones. The diminishing levels of E2 during menopause extend their influence on the gut microbiota, resulting in a reduction in the availability of tyrosine, tryptophan, and serotonin metabolites, affecting neurotransmitter synthesis and function. Understanding the interplay between menopause, frailty, E2 decline, and the intricate metabolisms of bone, gut, and muscle is imperative when unraveling the nuances of TBI after menopause. The current review underscores the significance of accounting for menopause-associated frailty in the incidence and consequences of TBI. The review also explores potential mechanisms to enhance gut, bone, and muscle health in menopausal women, aiming to mitigate frailty and improve TBI outcomes.
Collapse
Affiliation(s)
- Sophie B Sinder
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Sabrina V Sharma
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Isha S Shirvaikar
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Hari Pradhyumnan
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Shahil H Patel
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Indy Cabeda Diaz
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Gina G Perez
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Helen M Bramlett
- Department of Neurological Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA; The Miami Project to Cure Paralysis, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, USA
| | - Ami P Raval
- Peritz Scheinberg Cerebral Vascular Disease Research Laboratory (CVDRL), Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, USA; Bruce W. Carter Department of Veterans Affairs Medical Center, Miami, FL, USA
| |
Collapse
|
26
|
Fernández-Villabrille S, Martín-Vírgala J, Martín-Carro B, Baena-Huerta F, González-García N, Gil-Peña H, Rodríguez-García M, Fernández-Gómez JM, Fernández-Martín JL, Alonso-Montes C, Naves-Díaz M, Carrillo-López N, Panizo S. RANKL, but Not R-Spondins, Is Involved in Vascular Smooth Muscle Cell Calcification through LGR4 Interaction. Int J Mol Sci 2024; 25:5735. [PMID: 38891922 PMCID: PMC11172097 DOI: 10.3390/ijms25115735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Vascular calcification has a global health impact that is closely linked to bone loss. The Receptor Activator of Nuclear Factor Kappa B (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system, fundamental for bone metabolism, also plays an important role in vascular calcification. The Leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4), a novel receptor for RANKL, regulates bone remodeling, and it appears to be involved in vascular calcification. Besides RANKL, LGR4 interacts with R-spondins (RSPOs), which are known for their roles in bone but are less understood in vascular calcification. Studies were conducted in rats with chronic renal failure fed normal or high phosphorus diets for 18 weeks, with and without control of circulating parathormone (PTH) levels, resulting in different degrees of aortic calcification. Additionally, vascular smooth muscle cells (VSMCs) were cultured under non-calcifying (1 mM phosphate) and calcifying (3 mM phosphate) media with different concentrations of PTH. To explore the role of RANKL in VSMC calcification, increasing concentrations of soluble RANKL were added to non-calcifying and calcifying media. The effects mediated by RANKL binding to its receptor LGR4 were investigated by silencing the LGR4 receptor in VSMCs. Furthermore, the gene expression of the RANK/RANKL/OPG system and the ligands of LGR4 was assessed in human epigastric arteries obtained from kidney transplant recipients with calcification scores (Kauppila Index). Increased aortic calcium in rats coincided with elevated systolic blood pressure, upregulated Lgr4 and Rankl gene expression, downregulated Opg gene expression, and higher serum RANKL/OPG ratio without changes in Rspos gene expression. Elevated phosphate in vitro increased calcium content and expression of Rankl and Lgr4 while reducing Opg. Elevated PTH in the presence of high phosphate exacerbated the increase in calcium content. No changes in Rspos were observed under the conditions employed. The addition of soluble RANKL to VSMCs induced genotypic differentiation and calcification, partly prevented by LGR4 silencing. In the epigastric arteries of individuals presenting vascular calcification, the gene expression of RANKL was higher. While RSPOs show minimal impact on VSMC calcification, RANKL, interacting with LGR4, drives osteogenic differentiation in VSMCs, unveiling a novel mechanism beyond RANKL-RANK binding.
Collapse
MESH Headings
- RANK Ligand/metabolism
- RANK Ligand/genetics
- Animals
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Rats
- Humans
- Male
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Osteoprotegerin/metabolism
- Osteoprotegerin/genetics
- Parathyroid Hormone/metabolism
- Cells, Cultured
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Sara Fernández-Villabrille
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
| | - Julia Martín-Vírgala
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
| | - Beatriz Martín-Carro
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
| | - Francisco Baena-Huerta
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
| | - Nerea González-García
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
| | - Helena Gil-Peña
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
- AGC de la Infancia y Adolescencia, Hospital Universitario Central de Asturias (HUCA), Instituto de Investigación Sanitaria del Princiado de Asturias (ISPA), 33011 Oviedo, Spain
| | - Minerva Rodríguez-García
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
| | | | - José Luis Fernández-Martín
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Department of Medicine, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Cristina Alonso-Montes
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
| | - Manuel Naves-Díaz
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Department of Medicine, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Natalia Carrillo-López
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Department of Medicine, Universidad de Oviedo, 33006 Oviedo, Spain
| | - Sara Panizo
- Metabolismo Óseo, Vascular y Enfermedades Inflamatorias Crónicas, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain
- Redes de Investigación Cooperativa Orientadas a Resultados en Salud (RICORS), RICORS2040 (Kidney Disease), 33011 Oviedo, Spain
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, 33011 Oviedo, Spain
- Department of Medicine, Universidad de Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
27
|
Yu M, Qin K, Fan J, Zhao G, Zhao P, Zeng W, Chen C, Wang A, Wang Y, Zhong J, Zhu Y, Wagstaff W, Haydon RC, Luu HH, Ho S, Lee MJ, Strelzow J, Reid RR, He TC. The evolving roles of Wnt signaling in stem cell proliferation and differentiation, the development of human diseases, and therapeutic opportunities. Genes Dis 2024; 11:101026. [PMID: 38292186 PMCID: PMC10825312 DOI: 10.1016/j.gendis.2023.04.042] [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: 08/23/2022] [Revised: 03/18/2023] [Accepted: 04/12/2023] [Indexed: 02/01/2024] Open
Abstract
The evolutionarily conserved Wnt signaling pathway plays a central role in development and adult tissue homeostasis across species. Wnt proteins are secreted, lipid-modified signaling molecules that activate the canonical (β-catenin dependent) and non-canonical (β-catenin independent) Wnt signaling pathways. Cellular behaviors such as proliferation, differentiation, maturation, and proper body-axis specification are carried out by the canonical pathway, which is the best characterized of the known Wnt signaling paths. Wnt signaling has emerged as an important factor in stem cell biology and is known to affect the self-renewal of stem cells in various tissues. This includes but is not limited to embryonic, hematopoietic, mesenchymal, gut, neural, and epidermal stem cells. Wnt signaling has also been implicated in tumor cells that exhibit stem cell-like properties. Wnt signaling is crucial for bone formation and presents a potential target for the development of therapeutics for bone disorders. Not surprisingly, aberrant Wnt signaling is also associated with a wide variety of diseases, including cancer. Mutations of Wnt pathway members in cancer can lead to unchecked cell proliferation, epithelial-mesenchymal transition, and metastasis. Altogether, advances in the understanding of dysregulated Wnt signaling in disease have paved the way for the development of novel therapeutics that target components of the Wnt pathway. Beginning with a brief overview of the mechanisms of canonical and non-canonical Wnt, this review aims to summarize the current knowledge of Wnt signaling in stem cells, aberrations to the Wnt pathway associated with diseases, and novel therapeutics targeting the Wnt pathway in preclinical and clinical studies.
Collapse
Affiliation(s)
- Michael Yu
- School of Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Kevin Qin
- School of Medicine, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, The School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Guozhi Zhao
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Piao Zhao
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Zeng
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Neurology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523475, China
| | - Connie Chen
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Annie Wang
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yonghui Wang
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Clinical Laboratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai 200000, China
| | - Jiamin Zhong
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, The School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Orthopaedic Surgery, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Sherwin Ho
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael J. Lee
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Suture Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Suture Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
28
|
Mathold K, Nobin R, Brudin L, Carlsson M, Wanby P. Albumin-to-alkaline phosphatase ratio may be a better predictor of survival than sclerostin, dickkopf-1, osteopontin, osteoprotegerin and osteocalcin. Heliyon 2024; 10:e29639. [PMID: 38644839 PMCID: PMC11031828 DOI: 10.1016/j.heliyon.2024.e29639] [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: 08/18/2022] [Revised: 03/11/2024] [Accepted: 04/11/2024] [Indexed: 04/23/2024] Open
Abstract
Objectives The value of biochemical markers of bone turnover (BTMs) in predicting survival and disease remains unclear. In a prospective study we evaluated the novel biomarkers for bone turnover sclerostin, dickkopf-1 (DKK-1), osteopontin (OPN), osteoprotegerin (OPG) and osteocalcin (OC), as well as a traditional biomarker, alkaline phosphatase (ALP) in relation to risk of mortality, cardiovascular events and fractures. Participants and Methods:Routine blood tests and serum BTMs, including ALP, were analyzed in patients with hip fracture n = 97, stroke n = 71 and healthy volunteers n = 83 (mean age 86, 83 and 77, respectively), followed for 7 years. Hazard Ratios (HR) were calculated for mortality, cardiovascular events and fractures in relation to these biomarkers. After adding the albumin-to-ALP ratio (AAPR) a post hoc analysis was performed. Results 120 participants died during the study. In the entire group of patients and volunteers (n = 251) higher AAPR (HR 0.28, 95 % CI 0.14-0.59, p < 0.001) was associated with decreased mortality. OPN and OPG were associated with mortality risk only in the univariate statistical analysis. HR for high AAPR in relation to new cardiovascular events was borderline significant (HR 0.29, 95 % CI 0.08-1.06, p = 0.061). None of the examined biomarkers were associated with new fractures, nor with an increased risk of a new cardiovascular event. Conclusions AAPR may be a better predictor of mortality than the more novel BTMs, and higher AAPR could be associated with longer life expectancy. Further studies should determine the clinical usefulness of AAPR as a biomarker of mortality and cardiovascular disease.
Collapse
Affiliation(s)
- K. Mathold
- Department of Primary Care, Kalmar, Sweden
| | - R. Nobin
- Department of Orthopedics, Kalmar, Sweden
| | - L. Brudin
- Department of Clinical Physiology, Kalmar and Department of Medical and Health Sciences, University of Linköping, Sweden
| | - M. Carlsson
- Department of Clinical Chemistry, Kalmar and Department of Medicine and Optometry, Linnaeus University, Sweden
| | - P. Wanby
- Department of Internal Medicine, Section of Endocrinology, Kalmar, Department of Medical and Health Sciences, University of Linköping and Department of Medicine and Optometry, Linnaeus University, Sweden
| |
Collapse
|
29
|
Biz C, Khamisy-Farah R, Puce L, Szarpak L, Converti M, Ceylan Hİ, Crimì A, Bragazzi NL, Ruggieri P. Investigating and Practicing Orthopedics at the Intersection of Sex and Gender: Understanding the Physiological Basis, Pathology, and Treatment Response of Orthopedic Conditions by Adopting a Gender Lens: A Narrative Overview. Biomedicines 2024; 12:974. [PMID: 38790936 PMCID: PMC11118756 DOI: 10.3390/biomedicines12050974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
In the biomedical field, the differentiation between sex and gender is crucial for enhancing the understanding of human health and personalizing medical treatments, particularly within the domain of orthopedics. This distinction, often overlooked or misunderstood, is vital for dissecting and treating musculoskeletal conditions effectively. This review delves into the sex- and gender-specific physiology of bones, cartilage, ligaments, and tendons, highlighting how hormonal differences impact the musculoskeletal system's structure and function, and exploring the physiopathology of orthopedic conditions from an epidemiological, molecular, and clinical perspective, shedding light on the discrepancies in disease manifestation across sexes. Examples such as the higher rates of deformities (adolescent idiopathic and adult degenerative scoliosis and hallux valgus) in females and osteoporosis in postmenopausal women illustrate the critical role of sex and gender in orthopedic health. Additionally, the review addresses the morbidity-mortality paradox, where women, despite appearing less healthy on frailty indexes, show lower mortality rates, highlighting the complex interplay between biological and social determinants of health. Injuries and chronic orthopedic conditions such osteoarthritis exhibit gender- and sex-specific prevalence and progression patterns, necessitating a nuanced approach to treatment that considers these differences to optimize outcomes. Moreover, the review underscores the importance of recognizing the unique needs of sexual minority and gender-diverse individuals in orthopedic care, emphasizing the impact of gender-affirming hormone therapy on aspects like bone health and perioperative risks. To foster advancements in sex- and gender-specific orthopedics, we advocate for the strategic disaggregation of data by sex and gender and the inclusion of "Sexual Orientation and Gender Identity" (SOGI) data in research and clinical practice. Such measures can enrich clinical insights, ensure tailored patient care, and promote inclusivity within orthopedic treatments, ultimately enhancing the precision and effectiveness of care for diverse patient populations. Integrating sex and gender considerations into orthopedic research and practice is paramount for addressing the complex and varied needs of patients. By embracing this comprehensive approach, orthopedic medicine can move towards more personalized, effective, and inclusive treatment strategies, thereby improving patient outcomes and advancing the field.
Collapse
Affiliation(s)
- Carlo Biz
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, 35128 Padova, Italy; (A.C.); (P.R.)
| | - Rola Khamisy-Farah
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
- Clalit Health Service, Akko 2412001, Israel
| | - Luca Puce
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, 16132 Genoa, Italy;
| | - Lukasz Szarpak
- Department of Clinical Research and Development, LUXMED Group, 02-676 Warsaw, Poland;
- Henry JN Taub Department of Emergency Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Manlio Converti
- Department of Mental Health, Local Health Unit ASL Napoli 2 Nord, 80027 Naples, Italy;
| | - Halil İbrahim Ceylan
- Department of Physical Education of Sports Teaching, Faculty of Kazim Karabekir Education, Atatürk University, Erzurum 25030, Turkey;
| | - Alberto Crimì
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, 35128 Padova, Italy; (A.C.); (P.R.)
| | - Nicola Luigi Bragazzi
- Laboratory for Industrial and Applied Mathematics (LIAM), Department of Mathematics and Statistics, York University, Toronto, ON M3J 1P3, Canada
- Department of Food and Drugs, University of Parma, 43125 Parma, Italy
| | - Pietro Ruggieri
- Orthopedics and Orthopedic Oncology, Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padova, 35128 Padova, Italy; (A.C.); (P.R.)
| |
Collapse
|
30
|
Liu R, Zhao Y, Su S, Kwabil A, Njoku PC, Yu H, Li X. Unveiling cancer dormancy: Intrinsic mechanisms and extrinsic forces. Cancer Lett 2024; 591:216899. [PMID: 38649107 DOI: 10.1016/j.canlet.2024.216899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/06/2024] [Accepted: 04/12/2024] [Indexed: 04/25/2024]
Abstract
Tumor cells disseminate in various distant organs at early stages of cancer progression. These disseminated tumor cells (DTCs) can stay dormant/quiescent without causing patient symptoms for years or decades. These dormant tumor cells survive despite curative treatments by entering growth arrest, escaping immune surveillance, and/or developing drug resistance. However, these dormant cells can reactivate to proliferate, causing metastatic progression and/or relapse, posing a threat to patients' survival. It's unclear how cancer cells maintain dormancy and what triggers their reactivation. What are better approaches to prevent metastatic progression and relapse through harnessing cancer dormancy? To answer these remaining questions, we reviewed the studies of tumor dormancy and reactivation in various types of cancer using different model systems, including the brief history of dormancy studies, the intrinsic characteristics of dormant cells, and the external cues at the cellular and molecular levels. Furthermore, we discussed future directions in the field and the strategies for manipulating dormancy to prevent metastatic progression and recurrence.
Collapse
Affiliation(s)
- Ruihua Liu
- School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010070, China; Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Yawei Zhao
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Shang Su
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Augustine Kwabil
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Prisca Chinonso Njoku
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Haiquan Yu
- School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia Autonomous Region, 010070, China.
| | - Xiaohong Li
- Department of Cell and Cancer Biology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
31
|
Wen W, Pang Y, Tian Y, Xu C, Wang J, Wu Y, Xie X. Osteogenic mesenchymal stem cells/progenitors in the periodontium. Oral Dis 2024; 30:914-920. [PMID: 36648363 DOI: 10.1111/odi.14507] [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: 09/29/2022] [Revised: 12/30/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023]
Abstract
Periodontitis is the major cause of tooth loss in adults and is mainly characterized by alveolar bone destruction. Elucidating the mesenchymal stem cell (MSC)/progenitor populations of alveolar bone formation will provide valuable insights into regenerative approaches to clinical practice, such as endogenous regeneration and stem-cell-based tissue engineering therapies. Classically, MSCs residing in the bone marrow, periosteum, periodontal ligament (PDL), and even the gingiva are considered to be osteogenic progenitors. Furthermore, the contributions of MSCs expressing specific markers, including Gli1, Axin2, PTHrP, LepR, and α-SMA, to alveolar bone formation have been studied using cell lineage tracing and gene knockout models. In this review, we describe the MSCs/progenitors of alveolar bone and the biological properties of different subpopulations of MSCs involved in alveolar bone development, remodeling, injury repair, and regeneration.
Collapse
Affiliation(s)
- Wen Wen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yu Pang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yuyang Tian
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Chunmei Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jun Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yafei Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xudong Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Med-X Center for Materials, Department of Periodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Buck HV, Stains JP. Osteocyte-mediated mechanical response controls osteoblast differentiation and function. Front Physiol 2024; 15:1364694. [PMID: 38529481 PMCID: PMC10961341 DOI: 10.3389/fphys.2024.1364694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/29/2024] [Indexed: 03/27/2024] Open
Abstract
Low bone mass is a pervasive global health concern, with implications for osteoporosis, frailty, disability, and mortality. Lifestyle factors, including sedentary habits, metabolic dysfunction, and an aging population, contribute to the escalating prevalence of osteopenia and osteoporosis. The application of mechanical load to bone through physical activity and exercise prevents bone loss, while sufficient mechanical load stimulates new bone mass acquisition. Osteocytes, cells embedded within the bone, receive mechanical signals and translate these mechanical cues into biological signals, termed mechano-transduction. Mechano-transduction signals regulate other bone resident cells, such as osteoblasts and osteoclasts, to orchestrate changes in bone mass. This review explores the mechanisms through which osteocyte-mediated response to mechanical loading regulates osteoblast differentiation and bone formation. An overview of bone cell biology and the impact of mechanical load will be provided, with emphasis on the mechanical cues, mechano-transduction pathways, and factors that direct progenitor cells toward the osteoblast lineage. While there are a wide range of clinically available treatments for osteoporosis, the majority act through manipulation of the osteoclast and may have significant disadvantages. Despite the central role of osteoblasts to the deposition of new bone, few therapies directly target osteoblasts for the preservation of bone mass. Improved understanding of the mechanisms leading to osteoblastogenesis may reveal novel targets for translational investigation.
Collapse
Affiliation(s)
| | - Joseph Paul Stains
- School of Medicine, University of Maryland, Baltimore, MD, United States
| |
Collapse
|
33
|
Stringer F, Sims NA, Sachithanandan N, Aleksova J. Severe Osteoporosis With Pathogenic LRP5 Variant. JCEM CASE REPORTS 2024; 2:luae021. [PMID: 38404691 PMCID: PMC10888517 DOI: 10.1210/jcemcr/luae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Indexed: 02/27/2024]
Abstract
A 24-year-old female patient was diagnosed with osteoporosis after presenting with numerous fractures throughout her childhood and adolescence. Risk factors included chronic constipation, severe vitamin D deficiency, and long-term high-dose steroid use for severe eczema. Metabolic bone disorder clinical exome screening (limited panel of metabolic bone disorders and gastrointestinal disorders) was undertaken and revealed a class 4 likely pathogenic variant in the LRP5 gene known to cause osteoporosis. Optimal treatment for patients with this variant is not well defined. A literature review of the condition and potential treatment options is discussed.
Collapse
Affiliation(s)
- Felicity Stringer
- Department of Endocrinology, St Vincent's Hospital Melbourne, Fitzroy, Melbourne, VIC 3065, Australia
| | - Natalie A Sims
- St Vincent's Institute of Medical Research, Fitzroy, Melbourne, VIC 3065, Australia
- Melbourne Medical School, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Nirupa Sachithanandan
- Department of Endocrinology, St Vincent's Hospital Melbourne, Fitzroy, Melbourne, VIC 3065, Australia
- Melbourne Medical School, The University of Melbourne, Parkville, VIC 3052, Australia
| | - Jasna Aleksova
- Department of Endocrinology, St Vincent's Hospital Melbourne, Fitzroy, Melbourne, VIC 3065, Australia
- Department of Medicine, Monash University, Clayton, VIC 3168, Australia
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, VIC 3168, Australia
| |
Collapse
|
34
|
Arya PN, Saranya I, Selvamurugan N. Crosstalk between Wnt and bone morphogenetic protein signaling during osteogenic differentiation. World J Stem Cells 2024; 16:102-113. [PMID: 38455105 PMCID: PMC10915952 DOI: 10.4252/wjsc.v16.i2.102] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/04/2024] [Accepted: 01/22/2024] [Indexed: 02/26/2024] Open
Abstract
Mesenchymal stem cells (MSCs) originate from many sources, including the bone marrow and adipose tissue, and differentiate into various cell types, such as osteoblasts and adipocytes. Recent studies on MSCs have revealed that many transcription factors and signaling pathways control osteogenic development. Osteogenesis is the process by which new bones are formed; it also aids in bone remodeling. Wnt/β-catenin and bone morphogenetic protein (BMP) signaling pathways are involved in many cellular processes and considered to be essential for life. Wnt/β-catenin and BMPs are important for bone formation in mammalian development and various regulatory activities in the body. Recent studies have indicated that these two signaling pathways contribute to osteogenic differentiation. Active Wnt signaling pathway promotes osteogenesis by activating the downstream targets of the BMP signaling pathway. Here, we briefly review the molecular processes underlying the crosstalk between these two pathways and explain their participation in osteogenic differentiation, emphasizing the canonical pathways. This review also discusses the crosstalk mechanisms of Wnt/BMP signaling with Notch- and extracellular-regulated kinases in osteogenic differentiation and bone development.
Collapse
Affiliation(s)
- Pakkath Narayanan Arya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Iyyappan Saranya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India.
| |
Collapse
|
35
|
Yun C, Kim SH, Kim KM, Yang MH, Byun MR, Kim JH, Kwon D, Pham HTM, Kim HS, Kim JH, Jung YS. Advantages of Using 3D Spheroid Culture Systems in Toxicological and Pharmacological Assessment for Osteogenesis Research. Int J Mol Sci 2024; 25:2512. [PMID: 38473760 DOI: 10.3390/ijms25052512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Bone differentiation is crucial for skeletal development and maintenance. Its dysfunction can cause various pathological conditions such as rickets, osteoporosis, osteogenesis imperfecta, or Paget's disease. Although traditional two-dimensional cell culture systems have contributed significantly to our understanding of bone biology, they fail to replicate the intricate biotic environment of bone tissue. Three-dimensional (3D) spheroid cell cultures have gained widespread popularity for addressing bone defects. This review highlights the advantages of employing 3D culture systems to investigate bone differentiation. It highlights their capacity to mimic the complex in vivo environment and crucial cellular interactions pivotal to bone homeostasis. The exploration of 3D culture models in bone research offers enhanced physiological relevance, improved predictive capabilities, and reduced reliance on animal models, which have contributed to the advancement of safer and more effective strategies for drug development. Studies have highlighted the transformative potential of 3D culture systems for expanding our understanding of bone biology and developing targeted therapeutic interventions for bone-related disorders. This review explores how 3D culture systems have demonstrated promise in unraveling the intricate mechanisms governing bone homeostasis and responses to pharmacological agents.
Collapse
Affiliation(s)
- Chawon Yun
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Sou Hyun Kim
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Kyung Mok Kim
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Min Hye Yang
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| | - Mi Ran Byun
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Joung-Hee Kim
- Department of Medical Beauty Care, Dongguk University Wise, Gyeongju 38066, Republic of Korea
| | - Doyoung Kwon
- Jeju Research Institute of Pharmaceutical Sciences, College of Pharmacy, Jeju National University, Jeju 63243, Republic of Korea
| | - Huyen T M Pham
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Hyo-Sop Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Jae-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Republic of Korea
| | - Young-Suk Jung
- Department of Pharmacy, Research Institute for Drug Development, College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
36
|
Balachander GM, Nilawar S, Meka SRK, Ghosh LD, Chatterjee K. Unravelling microRNA regulation and miRNA-mRNA regulatory networks in osteogenesis driven by 3D nanotopographical cues. Biomater Sci 2024; 12:978-989. [PMID: 38189225 DOI: 10.1039/d3bm01597a] [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: 01/09/2024]
Abstract
Three-dimensional (3D) culturing of cells is being adopted for developing tissues for various applications such as mechanistic studies, drug testing, tissue regeneration, and animal-free meat. These approaches often involve cost-effective differentiation of stem or progenitor cells. One approach is to exploit architectural cues on a 3D substrate to drive cellular differentiation, which has been shown to be effective in various studies. Although extensive gene expression data from such studies have shown that gene expression patterns might differ, the gene regulatory networks controlling the expression of genes are rarely studied. In this study, we profiled genes and microRNAs (miRNAs) via next-generation sequencing (NGS) in human mesenchymal stem cells (hMSCs) driven toward osteogenesis via architectural cues in 3D matrices (3D conditions) and compared with cells in two-dimensional (2D) culture driven toward osteogenesis via soluble osteoinductive factors (OF conditions). The total number of differentially expressed genes was smaller in 3D compared to OF conditions. A distinct set of genes was observed under these conditions that have been shown to control osteogenic differentiation via different pathways. Small RNA sequencing revealed a core set of miRNAs to be differentially expressed under these conditions, similar to those that have been previously implicated in osteogenesis. We also observed a distinct regulation of miRNAs in these samples that can modulate gene expression, suggesting supplementary gene regulatory networks operative under different stimuli. This study provides insights into studying gene regulatory networks for identifying critical nodes to target for enhanced cellular differentiation and reveal the differences in physical and biochemical cues to drive cell fates.
Collapse
Affiliation(s)
- Gowri Manohari Balachander
- School of Biomedical Engineering, Indian Institute of Technology (BHU) Varanasi, Varanasi-221005, India.
| | - Sagar Nilawar
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India.
| | - Sai Rama Krishna Meka
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India.
| | - Lopamudra Das Ghosh
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India.
| | - Kaushik Chatterjee
- Department of Materials Engineering, Indian Institute of Science, Bangalore-560012, India.
| |
Collapse
|
37
|
Kavinda MD, Lee MH, Kang CH, Choi YH, Kim GY. 2,4'-Dihydroxybenzophenone Exerts Bone Formation and Antiosteoporotic Activity by Stimulating the β-Catenin Signaling Pathway. ACS Pharmacol Transl Sci 2024; 7:395-405. [PMID: 38357289 PMCID: PMC10863440 DOI: 10.1021/acsptsci.3c00251] [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: 09/25/2023] [Revised: 12/13/2023] [Accepted: 01/03/2024] [Indexed: 02/16/2024]
Abstract
2,4'-Dihydroxybenzophenone (DHP) is an organic compound derived from Garcinia xanthochymus, but there have been no reports on its biochemical functions and bioavailability. In this study, we evaluated whether DHP affects osteoblast differentiation and activation in MC3T3-E1 preosteoblast cells, as well as antiosteoporotic activity in zebrafish larvae. Nontoxic concentrations of DHP-treated MC3T3-E1 preosteoblast cells increased alkaline phosphatase (ALP) activation and mineralization in a concentration-dependent manner, accompanied by higher expression of osteoblast-specific markers, including Runt-related transcription factor 2 (RUNX2), osterix, and ALP. Consistent with the data in MC3T3-E1 preosteoblast cells, DHP upregulated osteoblast-specific marker genes in zebrafish larvae and simultaneously enhanced vertebral formation. We also revealed that DHP increased the phosphorylation of glycogen synthase kinase-3β (GSK-3β) at Ser9 and the total expression of β-catenin in the cytosol and markedly increased the localization of β-catenin into the nucleus. Furthermore, DHP restored the prednisolone (PDS)-induced marked decrease in ALP activity and mineralization, as well as osteoblast-specific marker expression. In PDS-treated zebrafish, DHP also alleviated PDS-induced osteoporosis by restoring vertebral formation and osteoblast-related gene expression. Taken together, these results suggest that DHP is a potential osteoanabolic candidate for treating osteoporosis by stimulating osteoblast differentiation.
Collapse
Affiliation(s)
| | - Mi-Hwa Lee
- Nakdonggang
National Institute of Biological Resources, Sanju 37242, Republic of Korea
| | - Chang-Hee Kang
- Nakdonggang
National Institute of Biological Resources, Sanju 37242, Republic of Korea
| | - Yung Hyun Choi
- Department
of Biochemistry, College of Korean Medicine, Dong-Eui University, Busan 47227, Republic
of Korea
| | - Gi-Young Kim
- Department
of Marine Life Science, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
38
|
Xiao B, Liu Y, Chandrasiri I, Adjei-Sowah E, Mereness J, Yan M, Benoit DSW. Bone-Targeted Nanoparticle Drug Delivery System-Mediated Macrophage Modulation for Enhanced Fracture Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2305336. [PMID: 37797180 PMCID: PMC10922143 DOI: 10.1002/smll.202305336] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/03/2023] [Indexed: 10/07/2023]
Abstract
Despite decades of progress, developing minimally invasive bone-specific drug delivery systems (DDS) to improve fracture healing remains a significant clinical challenge. To address this critical therapeutic need, nanoparticle (NP) DDS comprised of poly(styrene-alt-maleic anhydride)-b-poly(styrene) (PSMA-b-PS) functionalized with a peptide that targets tartrate-resistant acid phosphatase (TRAP) and achieves preferential fracture accumulation has been developed. The delivery of AR28, a glycogen synthase kinase-3 beta (GSK3β) inhibitor, via the TRAP binding peptide-NP (TBP-NP) expedites fracture healing. Interestingly, however, NPs are predominantly taken up by fracture-associated macrophages rather than cells typically associated with fracture healing. Therefore, the underlying mechanism of healing via TBP-NP is comprehensively investigated herein. TBP-NPAR28 promotes M2 macrophage polarization and enhances osteogenesis in preosteoblast-macrophage co-cultures in vitro. Longitudinal analysis of TBP-NPAR28 -mediated fracture healing reveals distinct spatial distributions of M2 macrophages, an increased M2/M1 ratio, and upregulation of anti-inflammatory and downregulated pro-inflammatory genes compared to controls. This work demonstrates the underlying therapeutic mechanism of bone-targeted NP DDS, which leverages macrophages as druggable targets and modulates M2 macrophage polarization to enhance fracture healing, highlighting the therapeutic benefit of this approach for fractures and bone-associated diseases.
Collapse
Affiliation(s)
- Baixue Xiao
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Yuxuan Liu
- Materials Science Program, University of Rochester, Rochester, NY, 14623, USA
| | - Indika Chandrasiri
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Emmanuela Adjei-Sowah
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Jared Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Ming Yan
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
| | - Danielle S W Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14623, USA
- Materials Science Program, University of Rochester, Rochester, NY, 14623, USA
- Department of Chemical Engineering, University of Rochester, Rochester, NY, 14623, USA
- Department of Bioengineering, Phil and Penny Knight Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR, 97403, USA
| |
Collapse
|
39
|
Hughes L, Centner C. Idiosyncratic bone responses to blood flow restriction exercise: new insights and future directions. J Appl Physiol (1985) 2024; 136:283-297. [PMID: 37994414 PMCID: PMC11212818 DOI: 10.1152/japplphysiol.00723.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023] Open
Abstract
Applying blood flow restriction (BFR) during low-load exercise induces beneficial adaptations of the myotendinous and neuromuscular systems. Despite the low mechanical tension, BFR exercise facilitates a localized hypoxic environment and increase in metabolic stress, widely regarded as the primary stimulus for tissue adaptations. First evidence indicates that low-load BFR exercise is effective in promoting an osteogenic response in bone, although this has previously been postulated to adapt primarily during high-impact weight-bearing exercise. Besides studies investigating the acute response of bone biomarkers following BFR exercise, first long-term trials demonstrate beneficial adaptations in bone in both healthy and clinical populations. Despite the increasing number of studies, the physiological mechanisms are largely unknown. Moreover, heterogeneity in methodological approaches such as biomarkers of bone metabolism measured, participant and study characteristics, and time course of measurement renders it difficult to formulate accurate conclusions. Furthermore, incongruity in the methods of BFR application (e.g., cuff pressure) limits the comparability of datasets and thus hinders generalizability of study findings. Appropriate use of biomarkers, effective BFR application, and befitting study design have the potential to progress knowledge on the acute and chronic response of bone to BFR exercise and contribute toward the development of a novel strategy to protect or enhance bone health. Therefore, the purpose of the present synthesis review is to 1) evaluate current mechanistic evidence; 2) discuss and offer explanations for similar and contrasting data findings; and 3) create a methodological framework for future mechanistic and applied research.
Collapse
Affiliation(s)
- Luke Hughes
- Department of Sport Exercise & Rehabilitation, Northumbria University, Newcastle upon Tyne, United Kingdom
| | - Christoph Centner
- Department of Sport and Sport Science, University of Freiburg, Freiburg, Germany
- Praxisklinik Rennbahn, Muttenz, Switzerland
| |
Collapse
|
40
|
Yılmaz D, Mathavan N, Wehrle E, Kuhn GA, Müller R. Mouse models of accelerated aging in musculoskeletal research for assessing frailty, sarcopenia, and osteoporosis - A review. Ageing Res Rev 2024; 93:102118. [PMID: 37935249 DOI: 10.1016/j.arr.2023.102118] [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: 07/03/2023] [Revised: 10/01/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Musculoskeletal aging encompasses the decline in bone and muscle function, leading to conditions such as frailty, osteoporosis, and sarcopenia. Unraveling the underlying molecular mechanisms and developing effective treatments are crucial for improving the quality of life for those affected. In this context, accelerated aging models offer valuable insights into these conditions by displaying the hallmarks of human aging. Herein, this review focuses on relevant mouse models of musculoskeletal aging with particular emphasis on frailty, osteoporosis, and sarcopenia. Among the discussed models, PolgA mice in particular exhibit hallmarks of musculoskeletal aging, presenting early-onset frailty, as well as reduced bone and muscle mass that closely resemble human musculoskeletal aging. Ultimately, findings from these models hold promise for advancing interventions targeted at age-related musculoskeletal disorders, effectively addressing the challenges posed by musculoskeletal aging and associated conditions in humans.
Collapse
Affiliation(s)
- Dilara Yılmaz
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | - Esther Wehrle
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland; AO Research Institute Davos, Davos Platz, Switzerland
| | - Gisela A Kuhn
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
41
|
Qin K, Yu M, Fan J, Wang H, Zhao P, Zhao G, Zeng W, Chen C, Wang Y, Wang A, Schwartz Z, Hong J, Song L, Wagstaff W, Haydon RC, Luu HH, Ho SH, Strelzow J, Reid RR, He TC, Shi LL. Canonical and noncanonical Wnt signaling: Multilayered mediators, signaling mechanisms and major signaling crosstalk. Genes Dis 2024; 11:103-134. [PMID: 37588235 PMCID: PMC10425814 DOI: 10.1016/j.gendis.2023.01.030] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/01/2022] [Accepted: 01/29/2023] [Indexed: 08/18/2023] Open
Abstract
Wnt signaling plays a major role in regulating cell proliferation and differentiation. The Wnt ligands are a family of 19 secreted glycoproteins that mediate their signaling effects via binding to Frizzled receptors and LRP5/6 coreceptors and transducing the signal either through β-catenin in the canonical pathway or through a series of other proteins in the noncanonical pathway. Many of the individual components of both canonical and noncanonical Wnt signaling have additional functions throughout the body, establishing the complex interplay between Wnt signaling and other signaling pathways. This crosstalk between Wnt signaling and other pathways gives Wnt signaling a vital role in many cellular and organ processes. Dysregulation of this system has been implicated in many diseases affecting a wide array of organ systems, including cancer and embryological defects, and can even cause embryonic lethality. The complexity of this system and its interacting proteins have made Wnt signaling a target for many therapeutic treatments. However, both stimulatory and inhibitory treatments come with potential risks that need to be addressed. This review synthesized much of the current knowledge on the Wnt signaling pathway, beginning with the history of Wnt signaling. It thoroughly described the different variants of Wnt signaling, including canonical, noncanonical Wnt/PCP, and the noncanonical Wnt/Ca2+ pathway. Further description involved each of its components and their involvement in other cellular processes. Finally, this review explained the various other pathways and processes that crosstalk with Wnt signaling.
Collapse
Affiliation(s)
- Kevin Qin
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Michael Yu
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL 60064, USA
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jiaming Fan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, The School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hongwei Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Piao Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Guozhi Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Departments of Orthopaedic Surgery and Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Interventional Neurology, The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, Guangdong 523475, China
| | - Connie Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Yonghui Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Department of Clinical Laboratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai 200000, China
| | - Annie Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Zander Schwartz
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- School of Biomedical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Jeffrey Hong
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lily Song
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - William Wagstaff
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Rex C. Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Hue H. Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Sherwin H. Ho
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Jason Strelzow
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
- Laboratory of Craniofacial Biology and Development, Department of Surgery Section of Plastic Surgery, The University of Chicago Medical Center, Chicago, IL 60637, USA
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL 60637, USA
| |
Collapse
|
42
|
Moraes de Lima Perini M, Pugh JN, Scott EM, Bhula K, Chirgwin A, Reul ON, Berbari NF, Li J. Primary cilia in osteoblasts and osteocytes are required for skeletal development and mechanotransduction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.15.570609. [PMID: 38318207 PMCID: PMC10843151 DOI: 10.1101/2023.12.15.570609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Primary cilia have been involved in the development and mechanosensation of various tissue types, including bone. In this study, we explored the mechanosensory role of primary cilia in bone growth and adaptation by examining two cilia specific genes, IFT88 and MKS5, required for proper cilia assembly and function. To analyze the role of primary cilia in osteoblasts, Osx1-GFP:Cre mice were bred with IFT88 LoxP/LoxP to generate mice with a conditional knockout of primary cilia in osteoblasts. A significant decrease in body weight was observed in both male (p=0.0048) and female (p=0.0374) conditional knockout (cKO) mice compared to the wild type (WT) controls. The femurs of cKO mice were significantly shorter than that of the WT mice of both male (p=0.0003) and female (p=0.0019) groups. Histological analysis revealed a significant difference in MAR (p=0.0005) and BFR/BS (p<0.0001) between female cKO and WT mice. The BFR/BS of male cKO mice was 58.03% lower compared to WT mice. To further investigate the role of primary cilia in osteocytes, Dmp1-8kb-Cre mice were crossed with MKS5 LoxP/LoxP to generate mice with defective cilia in osteocytes. In vivo axial ulnar loading was performed on 16-week-old mice for 3 consecutive days. The right ulnae were loaded for 120 cycles/day at a frequency of 2Hz with a peak force of 2.9N for female mice and 3.2N for male mice. Load-induced bone formation was measured using histomorphometry. The relative values of MS/BS, MAR and BFR/BS (loaded ulnae minus nonloaded ulnae) in male MKS5 cKO mice were decreased by 24.88%, 46.27% and 48.24%, respectively, compared to the controls. In the female groups, the rMS/BS was 52.5% lower, the rMAR was 27.58% lower, and the rBFR/BS was 41.54% lower in MKS5 cKO mice than the WT group. Histological analysis indicated that MKS5 cKO mice showed significantly decreased response to mechanical loading compared to the controls. Taken together, these data highlight a critical role of primary cilia in bone development and mechanotransduction, suggesting that the presence of primary cilia in osteoblasts play an important role in skeletal development, and primary cilia in osteocytes mediate mechanically induced bone formation.
Collapse
|
43
|
Sanaei R, Pagel CN, Ayodele BA, Lozanovski B, Beths T, Leary M, Shidid D, Kastrati E, Elambasseril J, Bühner U, Williamson T, Ryan S, Brandt M. Reducing the prosthesis modulus by inclusion of an open space lattice improves osteogenic response in a sheep model of extraarticular defect. Front Bioeng Biotechnol 2023; 11:1301454. [PMID: 38130824 PMCID: PMC10733966 DOI: 10.3389/fbioe.2023.1301454] [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: 09/25/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Introduction: Stress shielding is a common complication following endoprosthetic reconstruction surgery. The resulting periprosthetic osteopenia often manifests as catastrophic fractures and can significantly limit future treatment options. It has been long known that bone plates with lower elastic moduli are key to reducing the risk of stress shielding in orthopedics. Inclusion of open space lattices in metal endoprostheses is believed to reduce the prosthesis modulus potentially improving stress shielding. However, no in vivo data is currently available to support this assumption in long bone reconstruction. This manuscript aims to address this hypothesis using a sheep model of extraarticular bone defect. Methods: Initially, CT was used to create a virtual resection plan of the distal femoral metaphyses and to custom design endoprostheses specific to each femur. The endoprostheses comprised additively manufactured Ti6Al4V-ELI modules that either had a solid core with a modulus of ∼120 GPa (solid implant group) or an open space lattice core with unit cells that had a modulus of 3-6 GPa (lattice implant group). Osteotomies were performed using computer-assisted navigation followed by implantations. The periprosthetic, interfacial and interstitial regions of interest were evaluated by a combination of micro-CT, back-scattered scanning electron microscopy (BSEM), as well as epifluorescence and brightfield microscopy. Results: In the periprosthetic region, mean pixel intensity (a proxy for tissue mineral density in BSEM) in the caudal cortex was found to be higher in the lattice implant group. This was complemented by BSEM derived porosity being lower in the lattice implant group in both caudal and cranial cortices. In the interfacial and interstitial regions, most pronounced differences were observed in the axial interfacial perimeter where the solid implant group had greater bone coverage. In contrast, the lattice group had a greater coverage in the cranial interfacial region. Conclusion: Our findings suggest that reducing the prosthesis modulus by inclusion of an open-space lattice in its design has a positive effect on bone material and morphological parameters particularly within the periprosthetic regions. Improved mechanics appears to also have a measurable effect on the interfacial osteogenic response and osteointegration.
Collapse
Affiliation(s)
- Reza Sanaei
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Charles Neil Pagel
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Babatunde A. Ayodele
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Bill Lozanovski
- RMIT Centre for Additive Manufacturing, RMIT University, Carlton, VIC, Australia
| | - Thierry Beths
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Martin Leary
- RMIT Centre for Additive Manufacturing, RMIT University, Carlton, VIC, Australia
| | - Darpan Shidid
- RMIT Centre for Additive Manufacturing, RMIT University, Carlton, VIC, Australia
| | - Endri Kastrati
- RMIT Centre for Additive Manufacturing, RMIT University, Carlton, VIC, Australia
- Stryker Australia Pty Ltd., St Leonards, NSW, Australia
| | - Joe Elambasseril
- RMIT Centre for Additive Manufacturing, RMIT University, Carlton, VIC, Australia
| | | | - Tom Williamson
- RMIT Centre for Additive Manufacturing, RMIT University, Carlton, VIC, Australia
- Stryker Australia Pty Ltd., St Leonards, NSW, Australia
| | - Stewart Ryan
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, VIC, Australia
| | - Milan Brandt
- RMIT Centre for Additive Manufacturing, RMIT University, Carlton, VIC, Australia
| |
Collapse
|
44
|
Mahajan A, Bhattacharyya S. Immunomodulation by mesenchymal stem cells during osteogenic differentiation: Clinical implications during bone regeneration. Mol Immunol 2023; 164:143-152. [PMID: 38011783 DOI: 10.1016/j.molimm.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/08/2023] [Accepted: 11/12/2023] [Indexed: 11/29/2023]
Abstract
Critical bone defects resulting in delayed and non-union are a major concern in the field of orthopedics. Over the past decade, mesenchymal stem cells (MSCs) have become a promising frontier for bone repair and regeneration owing to their high expansion rate and osteogenic differentiation potential ex vivo. MSCs have also long been associated with their ability to modulate immune response in the recipients. These can even skew the immune response towards pro-inflammatory or anti-inflammatory type by sensing their local microenvironment. MSCs adopt anti-inflammatory phenotype at bone injury site and secrete various immunomodulatory factors such as IDO, NO, TGFβ1 and PGE-2 which have redundant role in osteoblast differentiation and bone formation. As such, several studies have also sought to decipher the immunomodulatory effects of osteogenically differentiated MSCs. The present review discusses the immunomodulatory status of MSCs during their osteogenic differentiation and summarizes few mechanisms that cause immunosuppression by osteogenically differentiated MSCs and its implication during bone healing.
Collapse
Affiliation(s)
- Aditi Mahajan
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Shalmoli Bhattacharyya
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
45
|
Abdulrahman SJ, Abdulhadi MA, Turki Jalil A, Falah D, Merza MS, Almulla AF, Ali A, Ali RT. Conjugated linoleic acid and glucosamine supplements may prevent bone loss in aging by regulating the RANKL/RANK/OPG pathway. Mol Biol Rep 2023; 50:10579-10588. [PMID: 37932498 DOI: 10.1007/s11033-023-08839-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 09/25/2023] [Indexed: 11/08/2023]
Abstract
The skeleton is a living organ that undergoes constant changes, including bone formation and resorption. It is affected by various diseases, such as osteoporosis, osteopenia, and osteomalacia. Nowadays, several methods are applied to protect bone health, including the use of hormonal and non-hormonal medications and supplements. However, certain drugs like glucocorticoids, thiazolidinediones, heparin, anticonvulsants, chemotherapy, and proton pump inhibitors can endanger bone health and cause bone loss. New studies are exploring the use of supplements, such as conjugated linoleic acid (CLA) and glucosamine, with fewer side effects during treatment. Various mechanisms have been proposed for the effects of CLA and glucosamine on bone structure, both direct and indirect. One mechanism that deserves special attention is the regulatory effect of RANKL/RANK/OPG on bone turnover. The RANKL/RANK/OPG pathway is considered a motive for osteoclast maturation and bone resorption. The cytokine system, consisting of the receptor activator of the nuclear factor (NF)-kB ligand (RANKL), its receptor RANK, and its decoy receptor, osteoprotegerin (OPG), plays a vital role in bone turnover. Over the past few years, researchers have observed the impact of CLA and glucosamine on the RANKL/RANK/OPG mechanism of bone turnover. However, no comprehensive study has been published on these supplements and their mechanism. To address this gap in knowledge, we have critically reviewed their potential effects. This review aims to assist in developing efficient treatment strategies and focusing future studies on these supplements.
Collapse
Affiliation(s)
| | - Mohanad Ali Abdulhadi
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq
| | | | - Dumooa Falah
- National University of Science and Technology, Dhi Qar, Iraq
| | - Muna S Merza
- Prosthetic dental Techniques Department, Al-Mustaqbal University College, Babylon, 51001, Iraq
| | - Abbas F Almulla
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Ahmed Ali
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Ronak Taher Ali
- College of Medical Technology, Al-Kitab University, Kirkuk, Iraq
| |
Collapse
|
46
|
Chatterjee S, Ghosh S, Datey A, Mahish C, Chattopadhyay S, Chattopadhyay S. Chikungunya virus perturbs the Wnt/β-catenin signaling pathway for efficient viral infection. J Virol 2023; 97:e0143023. [PMID: 37861335 PMCID: PMC10688348 DOI: 10.1128/jvi.01430-23] [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: 09/13/2023] [Accepted: 09/16/2023] [Indexed: 10/21/2023] Open
Abstract
IMPORTANCE Being obligate parasites, viruses use various host cell machineries in effectively replicating their genome, along with virus-encoded enzymes. In order to carry out infection and pathogenesis, viruses are known to manipulate fundamental cellular processes in cells and interfere with host gene expression. Several viruses interact with the cellular proteins involved in the Wnt/β-catenin pathway; however, reports regarding the involvement of protein components of the Wnt/β-catenin pathway in Chikungunya virus (CHIKV) infection are scarce. Additionally, there are currently no remedies or vaccines available for CHIKV. This is the first study to report that modulation of the Wnt/β-catenin pathway is crucial for effective CHIKV infection. These investigations deepen the understanding of the underlying mechanisms of CHIKV infection and offer new avenue for developing effective countermeasures to efficiently manage CHIKV infection.
Collapse
Affiliation(s)
- Sanchari Chatterjee
- Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Soumyajit Ghosh
- Institute of Life Sciences, Bhubaneswar, India
- Regional Centre for Biotechnology, Faridabad, India
| | - Ankita Datey
- Institute of Life Sciences, Bhubaneswar, India
- School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT) University, Bhubaneswar, India
| | - Chandan Mahish
- National Institute of Science Education and Research, an OCC of Homi Bhaba National Institute, Bhubaneswar, Odisha, India
| | - Subhasis Chattopadhyay
- National Institute of Science Education and Research, an OCC of Homi Bhaba National Institute, Bhubaneswar, Odisha, India
| | | |
Collapse
|
47
|
Khotib J, Marhaeny HD, Miatmoko A, Budiatin AS, Ardianto C, Rahmadi M, Pratama YA, Tahir M. Differentiation of osteoblasts: the links between essential transcription factors. J Biomol Struct Dyn 2023; 41:10257-10276. [PMID: 36420663 DOI: 10.1080/07391102.2022.2148749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/12/2022] [Indexed: 11/27/2022]
Abstract
Osteoblasts, cells derived from mesenchymal stem cells (MSCs) in the bone marrow, are cells responsible for bone formation and remodeling. The differentiation of osteoblasts from MSCs is triggered by the expression of specific genes, which are subsequently controlled by pro-osteogenic pathways. Mature osteoblasts then differentiate into osteocytes and are embedded in the bone matrix. Dysregulation of osteoblast function can cause inadequate bone formation, which leads to the development of bone disease. Various key molecules are involved in the regulation of osteoblastogenesis, which are transcription factors. Previous studies have heavily examined the role of factors that control gene expression during osteoblastogenesis, both in vitro and in vivo. However, the systematic relationship of these transcription factors remains unknown. The involvement of ncRNAs in this mechanism, particularly miRNAs, lncRNAs, and circRNAs, has been shown to influence transcriptional factor activity in the regulation of osteoblast differentiation. Here, we discuss nine essential transcription factors involved in osteoblast differentiation, including Runx2, Osx, Dlx5, β-catenin, ATF4, Ihh, Satb2, and Shn3. In addition, we summarize the role of ncRNAs and their relationship to these essential transcription factors in order to improve our understanding of the transcriptional regulation of osteoblast differentiation. Adequate exploration and understanding of the molecular mechanisms of osteoblastogenesis can be a critical strategy in the development of therapies for bone-related diseases.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Junaidi Khotib
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Honey Dzikri Marhaeny
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Andang Miatmoko
- Department of Pharmaceutical Science, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Aniek Setiya Budiatin
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Mahardian Rahmadi
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Yusuf Alif Pratama
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya, Indonesia
| | - Muhammad Tahir
- Department of Pharmaceutical Science, Kulliyah of Pharmacy, International Islamic University Malaysia, Pahang, Malaysia
| |
Collapse
|
48
|
Wang K, Hu S. The synergistic effects of polyphenols and intestinal microbiota on osteoporosis. Front Immunol 2023; 14:1285621. [PMID: 37936705 PMCID: PMC10626506 DOI: 10.3389/fimmu.2023.1285621] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/11/2023] [Indexed: 11/09/2023] Open
Abstract
Osteoporosis is a common metabolic disease in middle-aged and elderly people. It is characterized by a reduction in bone mass, compromised bone microstructure, heightened bone fragility, and an increased susceptibility to fractures. The dynamic imbalance between osteoblast and osteoclast populations is a decisive factor in the occurrence of osteoporosis. With the increase in the elderly population in society, the incidence of osteoporosis, disability, and mortality have gradually increased. Polyphenols are a fascinating class of compounds that are found in both food and medicine and exhibit a variety of biological activities with significant health benefits. As a component of food, polyphenols not only provide color, flavor, and aroma but also act as potent antioxidants, protecting our cells from oxidative stress and reducing the risk of chronic disease. Moreover, these natural compounds exhibit anti-inflammatory properties, which aid in immune response regulation and potentially alleviate symptoms of diverse ailments. The gut microbiota can degrade polyphenols into more absorbable metabolites, thereby increasing their bioavailability. Polyphenols can also shape the gut microbiota and increase its abundance. Therefore, studying the synergistic effect between gut microbiota and polyphenols may help in the treatment and prevention of osteoporosis. By delving into how gut microbiota can enhance the bioavailability of polyphenols and how polyphenols can shape the gut microbiota and increase its abundance, this review offers valuable information and references for the treatment and prevention of osteoporosis.
Collapse
Affiliation(s)
- Keyu Wang
- The Orthopaedic Center, The Affiliated Wenling Hospital of Wenzhou Medical University (The First People’s Hospital of Wenling), Wenling, Zhejiang, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan, China
| | - Siwang Hu
- The Orthopaedic Center, The Affiliated Wenling Hospital of Wenzhou Medical University (The First People’s Hospital of Wenling), Wenling, Zhejiang, China
| |
Collapse
|
49
|
Zhou L, Zhao S, Xing X. Effects of different signaling pathways on odontogenic differentiation of dental pulp stem cells: a review. Front Physiol 2023; 14:1272764. [PMID: 37929208 PMCID: PMC10622672 DOI: 10.3389/fphys.2023.1272764] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/02/2023] [Indexed: 11/07/2023] Open
Abstract
Dental pulp stem cells (DPSCs) are a type of mesenchymal stem cells that can differentiate into odontoblast-like cells and protect the pulp. The differentiation of DPSCs can be influenced by biomaterials or growth factors that activate different signaling pathways in vitro or in vivo. In this review, we summarized six major pathways involved in the odontogenic differentiation of DPSCs, Wnt signaling pathways, Smad signaling pathways, MAPK signaling pathways, NF-kB signaling pathways, PI3K/AKT/mTOR signaling pathways, and Notch signaling pathways. Various factors can influence the odontogenic differentiation of DPSCs through one or more signaling pathways. By understanding the interactions between these signaling pathways, we can expand our knowledge of the mechanisms underlying the regeneration of the pulp-dentin complex.
Collapse
Affiliation(s)
| | | | - Xianghui Xing
- Department of Pediatric Dentistry, Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
50
|
Fassio A, Atzeni F, Rossini M, D’Amico V, Cantatore F, Chimenti MS, Crotti C, Frediani B, Giusti A, Peluso G, Rovera G, Scolieri P, Raimondo V, Gatti D. Osteoimmunology of Spondyloarthritis. Int J Mol Sci 2023; 24:14924. [PMID: 37834372 PMCID: PMC10573470 DOI: 10.3390/ijms241914924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/15/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
The mechanisms underlying the development of bone damage in the context of spondyloarthritis (SpA) are not completely understood. To date, a considerable amount of evidence indicates that several developmental pathways are crucially involved in osteoimmunology. The present review explores the biological mechanisms underlying the relationship between inflammatory dysregulation, structural progression, and osteoporosis in this diverse family of conditions. We summarize the current knowledge of bone biology and balance and the foundations of bone regulation, including bone morphogenetic protein, the Wnt pathway, and Hedgehog signaling, as well as the role of cytokines in the development of bone damage in SpA. Other areas surveyed include the pathobiology of bone damage and systemic bone loss (osteoporosis) in SpA and the effects of pharmacological treatment on focal bone damage. Lastly, we present data relative to a survey of bone metabolic assessment in SpA from Italian bone specialist rheumatology centers. The results confirm that most of the attention to bone health is given to postmenopausal subjects and that the aspect of metabolic bone health may still be underrepresented. In our opinion, it may be the time for a call to action to increase the interest in and focus on the diagnosis and management of SpA.
Collapse
Affiliation(s)
- Angelo Fassio
- Dipartimento di Medicina, Università di Verona, 37124 Verona, Italy; (M.R.); (D.G.)
| | - Fabiola Atzeni
- Unità Operativa Complessa di Reumatologia Azienda Ospedaliero Universitaria Policlinico “G. Martino” di Messina, 35128 Messina, Italy; (F.A.); (V.D.)
| | - Maurizio Rossini
- Dipartimento di Medicina, Università di Verona, 37124 Verona, Italy; (M.R.); (D.G.)
| | - Valeria D’Amico
- Unità Operativa Complessa di Reumatologia Azienda Ospedaliero Universitaria Policlinico “G. Martino” di Messina, 35128 Messina, Italy; (F.A.); (V.D.)
| | - Francesco Cantatore
- Unità Operativa Complessa di Reumatologia Universitaria, Polic. “Riuniti” di Foggia, 71122 Foggia, Italy;
| | - Maria Sole Chimenti
- Dipartimento di Medicina dei Sistemi, Reumatologia, Allergologia e Immunologia Clinica Università di Roma Tor Vergata, 00133 Rome, Italy;
| | - Chiara Crotti
- UOC Osteoporosi e Malattie Metaboliche dell’Osso Dipartimento di Reumatologia e Scienze Mediche ASST-G. Pini-CTO, 20122 Milan, Italy;
| | - Bruno Frediani
- Department of Medical, Surgical and Neuroscience Sciences, Rheumatology University of Siena, 53100 Siena, Italy;
| | - Andrea Giusti
- SSD Malattie Reumatologiche e del Metabolismo Osseo, Dipartimento delle Specialità Mediche, ASL3, 16132 Genova, Italy;
| | - Giusy Peluso
- UOC di Reumatologia-Fondazione Policlinico Universitario Agostino Gemelli-IRCSS, 00168 Rome, Italy;
| | - Guido Rovera
- Ospedale S. Andrea, Divisione Reumatologia, 13100 Vercelli, Italy;
| | - Palma Scolieri
- Ambulatorio di Reumatologia Ospedale Nuovo Regina Margherita ASL ROMA1, 00153 Rome, Italy;
| | | | - Davide Gatti
- Dipartimento di Medicina, Università di Verona, 37124 Verona, Italy; (M.R.); (D.G.)
| | | |
Collapse
|