1
|
Han W, Xiong N, Huang L. Probiotics and nanoparticle-mediated nutrient delivery in the management of transfusion-supported diseases. Front Cell Infect Microbiol 2025; 15:1575798. [PMID: 40292219 PMCID: PMC12021914 DOI: 10.3389/fcimb.2025.1575798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Bone marrow is vital for hematopoiesis, producing blood cells essential for oxygen transport, immune defense, and clotting. However, disorders like leukemia, lymphoma, aplastic anemia, and myelodysplastic syndromes can severely disrupt its function, leading to life-threatening complications. Traditional treatments, including chemotherapy and stem cell transplants, have significantly improved patient outcomes but are often associated with severe side effects and limitations, necessitating the exploration of safer, more targeted therapeutic strategies. Nanotechnology has emerged as a promising approach for addressing these challenges, particularly in the delivery of nutraceuticals-bioactive compounds derived from food sources with potential therapeutic benefits. Despite their promise, nutraceuticals often face clinical limitations due to poor bioavailability, instability, and inefficient delivery to target sites. Nanoparticles offer a viable solution by enhancing the stability, absorption, and targeted transport of nutraceuticals to bone marrow while minimizing systemic side effects. This study explores a range of bone marrow disorders, conventional treatment modalities, and the potential of nanoparticles to enhance nutraceutical-based therapies. By improving targeted delivery and therapeutic efficacy, nanoparticles could revolutionize bone marrow disease management, providing patients with more effective and less invasive treatment options. These advancements represent a significant step toward safer and more efficient therapeutic approaches, ultimately improving patient prognosis and overall health.
Collapse
Affiliation(s)
- Wendao Han
- Department of Blood Transfusion, Meizhou People’s Hospital, Meizhou Academy of
Medical Sciences, Meizhou, China
| | | | | |
Collapse
|
2
|
Shi D, Li Y, Tian M, Xue M, Wang J, An H. Nanomaterials-Based Drug Delivery Systems for Therapeutic Applications in Osteoporosis. Adv Biol (Weinh) 2025:e2400721. [PMID: 40195930 DOI: 10.1002/adbi.202400721] [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: 03/04/2025] [Indexed: 04/09/2025]
Abstract
The etiology of osteoporosis is rooted in the disruption of the intricate equilibrium between bone formation and bone resorption processes. Nevertheless, the conventional anti-osteoporotic medications and hormonal therapeutic regimens currently employed in clinical practice are associated with a multitude of adverse effects, thereby constraining their overall therapeutic efficacy and potential. Recently, nanomaterials have emerged as a promising alternative due to their minimal side effects, efficient drug delivery, and ability to enhance bone formation, aiding in restoring bone balance. This review delves into the fundamental principles of bone remodeling and the bone microenvironment, as well as current clinical treatment approaches for osteoporosis. It subsequently explores the research status of nanomaterial-based drug delivery systems for osteoporosis treatment, encompassing inorganic nanomaterials, organic nanomaterials, cell-mimicking carriers and exosomes mimics and emerging therapies targeting the osteoporosis microenvironment. Finally, the review discusses the potential of nanomedicine in treating osteoporosis and outlines the future trajectory of this burgeoning field. The aim is to provide a comprehensive reference for the application of nanomaterial-based drug delivery strategies in osteoporosis therapy, thereby fostering further advancements and innovations in this critical area of medical research.
Collapse
Affiliation(s)
- Donghong Shi
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
- State Key Laboratory of Reliability and Intelligence of Electrical Equipment, School of Electrical Engineering, Hebei University of Technology, Tianjin, 300130, P. R. China
| | - Yuling Li
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Meng Tian
- Hebei Tourism College, Hebei, Chengde, 067000, P. R. China
| | - Mengge Xue
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Jinping Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| | - Hailong An
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Innovation and Research Institute of Hebei University of Technology in Shijiazhuang, Hebei University of Technology, Tianjin, 300401, P. R. China
| |
Collapse
|
3
|
Wang Z, Xiang P, Xu Z, Gu M, Zhang R, Li Y, Chen H, He L, Yi C. The Role of Magnesium, Zinc, and Strontium in Osteoporotic Fracture Repair. Bioengineering (Basel) 2025; 12:201. [PMID: 40001719 PMCID: PMC11851939 DOI: 10.3390/bioengineering12020201] [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: 12/06/2024] [Revised: 02/13/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Osteoporotic fractures represent a significant public health challenge in the context of an aging global population, with the rising prevalence of osteoporosis intensifying the demand for effective fracture treatment. Restoring the structure and function of bone tissue damaged by osteoporosis-induced defects remains a critical issue in clinical practice. In recent years, bioactive metallic materials such as magnesium, zinc, and strontium have gained considerable attention due to their exceptional mechanical properties, biocompatibility, and biodegradability, positioning them as promising materials for osteoporotic fracture repair. This review systematically explored the biological mechanisms, application advancements, and associated challenges of magnesium, zinc, and strontium in fracture healing. Key topics included their roles in promoting osteoblast proliferation and differentiation, inhibiting osteoclast activity, and modulating the bone microenvironment. Additionally, this review examined the optimization strategies for their clinical application, such as their integration into bone scaffolds, the functionalization of conventional materials, and the synergistic effects between different metals. Finally, this review analyzed the current progress and unresolved issues in this field, offering a forward-looking perspective on the clinical potential of bioactive metallic materials in precision treatment of osteoporotic fractures.
Collapse
Affiliation(s)
- Zhen Wang
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.)
| | - Penghui Xiang
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.)
| | - Zhe Xu
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.)
| | - Meiqi Gu
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.)
| | - Rui Zhang
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.)
| | - Yifei Li
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.)
| | - Hua Chen
- Department of Orthopaedic Trauma, Chinese PLA General Hospital (301 Hospital), Beijing 100853, China
| | - Li He
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.)
| | - Chengla Yi
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.)
| |
Collapse
|
4
|
Benjamin AS, Nayak S. Iron oxide nanoparticles coated with bioactive materials: a viable theragnostic strategy to improve osteosarcoma treatment. DISCOVER NANO 2025; 20:18. [PMID: 39883285 PMCID: PMC11782756 DOI: 10.1186/s11671-024-04163-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 12/04/2024] [Indexed: 01/31/2025]
Abstract
Osteosarcoma (OS) is distinguished as a high-grade malignant tumor, characterized by rapid systemic metastasis, particularly to the lungs, resulting in very low survival rates. Understanding the complexities of tumor development and mutation is the need of the hour for the advancement of targeted therapies in cancer care. A significant innovation in this area is the use of nanotechnology, specifically nanoparticles, to tackle various challenges in cancer treatment. Iron oxide nanoparticles stand out in both therapeutic and diagnostic applications, offering a versatile platform for targeted drug delivery, hyperthermia, magneto-thermal therapy, and combinational therapy using modulation of ferroptosis pathways. These nanoparticles are easy to synthesize, non-toxic, biocompatible, and display enhanced circulation time within the system. They can also be easily conjugated to anti-cancer drugs, targeting agents, or genetic vectors that respond to specific stimuli or pH changes. The surface functionalization of these nanoparticles using bioactive molecules unveils a promising and effective nanoparticle system for assisting osteosarcoma therapy. This review will summarize the current conventional therapies for osteosarcoma and their disadvantages, the synthesis and modification of iron oxide nanoparticles documented in the literature, cellular targeting and uptake mechanism, with focus on their functionalization using natural biomaterials and application strategies towards management of osteosarcoma. The review also compiles the translational challenges and future prospects that must be addressed for clinical advancements of iron oxide based osteosarcoma treatment in the future.
Collapse
Affiliation(s)
- Amy Sarah Benjamin
- School of Advanced Sciences, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Sunita Nayak
- School of Biosciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
5
|
Kang Q, Wang W, Wu S, Hu G. B-doped nano-hydroxyapatite facilitates proliferation and differentiation of osteoblasts. J Orthop Surg Res 2025; 20:62. [PMID: 39825395 PMCID: PMC11748584 DOI: 10.1186/s13018-024-05414-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/25/2024] [Indexed: 01/20/2025] Open
Abstract
PURPOSE We aimed to explore the mechanism by which Boron-doped nano-hydroxyapatite (B-nHAp) facilitates the proliferation and differentiation of osteoblasts through controlled release of B. METHODS B-nHAp characterization was accomplished by means of X-ray diffraction, scanning electron microscopy, inductively coupled plasma mass spectrometry, and transmission electron microscopy. Human bone marrow mesenchymal stem cells (hBMSCs) were subjected to flow cytometry, alizarin red S staining, and cell counting kit-8 assay for proliferation and differentiation determination. Western blotting for protein expression detection together with real-time quantitative polymerase chain reaction for mRNA expression measurement were carried out on those related to hBMSC proliferation and differentiation. Immunofluorescence staining was conducted to determine the activity of the Wnt/β-catenin signaling pathway. RESULTS B-nHAp particles had structured configuration and uniform size, and a typical nHAp crystal structure. The B content in B-nHAp was in line with expectation. hBMSCs displayed stemness. B-nHAp significantly facilitated the proliferation of hBMSCs, and significantly more mineralized nodules formed in the B-nHAp group. B-nHAp significantly upregulated the expressions of marker molecules related to hBMSC proliferation and differentiation. B-nHAp boosted the activity of the Wnt/β-catenin signaling pathway. CONCLUSION B-nHAp modulates the Wnt/β-catenin signaling pathway to significantly enhance the proliferative and differential abilities of osteoblasts, potentially as an efficient material for bone repair.
Collapse
Affiliation(s)
- Qiao Kang
- Department of Maxillofacial Surgery, University Clinical Hospital No. 4, I.M. Sechenov First Moscow State Medical University, Moscow, Russia, 119048
| | - Wenhao Wang
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, 518033, China
| | - Shengtao Wu
- Department of Oral Surgery and Implantology, Carolinum, Goethe University, 60596, Frankfurt, Germany
| | - Ganggang Hu
- Xuzhou Medical University Affiliated Stomatology Hospital, Xuzhou, 221002, Jiangsu Province, China.
| |
Collapse
|
6
|
Zhou Z, Jin Z, Tian Y, Huangfu C, Fan Z, Liu D. CDK14 is regulated by IGF2BP2 and involved in osteogenic differentiation via Wnt/β-catenin signaling pathway in vitro. Life Sci 2024; 358:123148. [PMID: 39447733 DOI: 10.1016/j.lfs.2024.123148] [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: 06/18/2024] [Revised: 10/07/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024]
Abstract
AIMS Cyclin-dependent kinase (CDK) family proteins involve in various cellular processes via regulating the cell cycle; however, their expression during osteogenic differentiation and postmenopausal osteoporosis remains poorly understood. MAIN METHODS Using bioinformatics, we screened for CDK14 bound to Insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) and explored its expression in vitro with time-gradient model and in a mouse model of postmenopausal osteoporosis, building on prior research. Subsequently, we investigated its effect on osteoblast proliferation, cell cycle dynamics, and osteogenic differentiation by administering CDK14 siRNA and the covalent inhibitor FMF-04-159-2. Furthermore, we examined the interaction between IGF2BP2 and CDK14. Finally, we validated the regulatory role of CDK14 on the Wnt/β-catenin pathway. KEY FINDINGS Our findings demonstrate a time-dependent CDK14 expression patterns during osteogenic differentiation of MC3T3-E1 cell line, with an initial increase followed by gradual decline over time. Notably, CDK14 expression exhibited significant reduction in bone tissue of postmenopausal osteoporosis mouse model. CDK14 inhibition altered osteoblast cell cycle dynamics, significantly reduced cellular proliferation capacity, and impaired osteogenic differentiation ability. IGF2BP2 interacted with CDK14 mRNA, and stabilizing mRNA's structure and inhibiting its degradation. Additionally, CDK14 facilitated Low-density lipoprotein receptor-related protein 6 (LRP6) and Glycogen synthase kinase 3β (GSK3β) phosphorylation, thus regulating β-catenin levels. SIGNIFICANCE These findings provide further insight into the molecular mechanisms governing osteoblast proliferation, differentiation and osteoporosis.
Collapse
Affiliation(s)
- Zimo Zhou
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Zhuoru Jin
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Yicheng Tian
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Chenghao Huangfu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Zheng Fan
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| | - Da Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
7
|
Iwamoto N, Chiba K, Sato S, Tashiro S, Shiraishi K, Watanabe K, Ohki N, Okada A, Koga T, Kawashiri SY, Tamai M, Osaki M, Kawakami A. Preferable effect of CTLA4-Ig on both bone erosion and bone microarchitecture in rheumatoid arthritis revealed by HR-pQCT. Sci Rep 2024; 14:27673. [PMID: 39532911 PMCID: PMC11557861 DOI: 10.1038/s41598-024-77392-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
This exploratory study aimed to examine the impact of abatacept treatment on bone structure in patients with rheumatoid arthritis (RA) using high-resolution peripheral quantitative computed tomography (HR-pQCT). RA patients initiating either abatacept or newly introduced csDMARDs were enrolled in this prospective, non-randomized, two-group study. Bone structure in the 2nd and 3rd metacarpal heads was assessed using HR-pQCT at 0, 6, and 12 months after enrollment. Synovitis was evaluated using musculoskeletal ultrasound and MRI. The adjusted mean between-group differences (abatacept-csDMARDs group) were estimated using a mixed-effect model. Thirty-five patients (abatacept group: n = 15; csDMARDs group: n = 20) were analyzed. Changes in erosion volume, depth and width were numerically smaller in the abatacept group compared to the csDMARDs group (adjusted mean between-group differences: - 1.86 mm3, - 0.02 mm, and - 0.09 mm, respectively). Over a 12-month period, 5 erosions emerged in the csDMARDs group, while only 1 erosion appeared in the abatacept group. Compared to csDMARDs, abatacept better preserved bone microarchitecture; several components of bone microarchitecture were significantly worsened at 6 months in the csDMARDs group, but were not deteriorated at 6 months in the abatacept group. Changes in synovitis scores were similar between the two treatment groups. Our results indicate that abatacept prevented the progression of bone erosion including new occurrence, and also prevented worsening of bone strength independently with synovitis compared to csDMARDs including MTX. Thus, abatacept treatment may provide benefits not only in inhibiting the progress of bone erosion but also in preventing bone microarchitectural deterioration.
Collapse
Affiliation(s)
- Naoki Iwamoto
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan.
| | - Ko Chiba
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shuntaro Sato
- Clinical Research Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shigeki Tashiro
- Clinical Research Center, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Kazuteru Shiraishi
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Kounosuke Watanabe
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Nozomi Ohki
- Department of Radiological Sciences, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Akitomo Okada
- Department of Rheumatology, National Hospital Organization Nagasaki Medical Center, Kubara 2-1001-1, Omura, Nagasaki, 856-8562, Japan
| | - Tomohiro Koga
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Shin-Ya Kawashiri
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
- Center for Collaborative Medical Education and Development, Nagasaki University Institute of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Mami Tamai
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Makoto Osaki
- Department of Orthopedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| | - Atsushi Kawakami
- Division of Advanced Preventive Medical Sciences, Department of Immunology and Rheumatology, Nagasaki University Graduate School of Biomedical Sciences, 1-7-1 Sakamoto, Nagasaki, 852-8501, Japan
| |
Collapse
|
8
|
Wu Y, Sun B, Tang Y, Shen A, Lin Y, Zhao X, Li J, Monteiro MJ, Gu W. Bone targeted nano-drug and nano-delivery. Bone Res 2024; 12:51. [PMID: 39231955 PMCID: PMC11375042 DOI: 10.1038/s41413-024-00356-2] [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: 03/13/2024] [Revised: 07/04/2024] [Accepted: 07/16/2024] [Indexed: 09/06/2024] Open
Abstract
There are currently no targeted delivery systems to satisfactorily treat bone-related disorders. Many clinical drugs consisting of small organic molecules have a short circulation half-life and do not effectively reach the diseased tissue site. This coupled with repeatedly high dose usage that leads to severe side effects. With the advance in nanotechnology, drugs contained within a nano-delivery device or drugs aggregated into nanoparticles (nano-drugs) have shown promises in targeted drug delivery. The ability to design nanoparticles to target bone has attracted many researchers to develop new systems for treating bone related diseases and even repurposing current drug therapies. In this review, we shall summarise the latest progress in this area and present a perspective for future development in the field. We will focus on calcium-based nanoparticle systems that modulate calcium metabolism and consequently, the bone microenvironment to inhibit disease progression (including cancer). We shall also review the bone affinity drug family, bisphosphonates, as both a nano-drug and nano-delivery system for bone targeted therapy. The ability to target and release the drug in a controlled manner at the disease site represents a promising safe therapy to treat bone diseases in the future.
Collapse
Affiliation(s)
- Yilun Wu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Bing Sun
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Ying Tang
- Science and Technology Innovation Centre, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Aining Shen
- Shenzhen Bay Laboratory, Shenzhen, Guangdong, China
| | - Yanlin Lin
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Xiaohui Zhao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jingui Li
- School of Veterinary Medicine, Jiangsu Co-innovation Centre for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Michael J Monteiro
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia
| | - Wenyi Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, QLD, Australia.
| |
Collapse
|
9
|
Kolipaka R, Magesh I, Bharathy MA, Karthik S, Saranya I, Selvamurugan N. A potential function for MicroRNA-124 in normal and pathological bone conditions. Noncoding RNA Res 2024; 9:687-694. [PMID: 38577015 PMCID: PMC10990750 DOI: 10.1016/j.ncrna.2024.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/22/2024] [Accepted: 02/25/2024] [Indexed: 04/06/2024] Open
Abstract
Cells produce short single-stranded non-coding RNAs (ncRNAs) called microRNAs (miRNAs), which actively regulate gene expression at the posttranscriptional level. Several miRNAs have been observed to exert significant impacts on bone health and bone-related disorders. One of these, miR-124, is observed in bone microenvironments and is conserved across species. It affects bone cell growth and differentiation by activating different transcription factors and signaling pathways. In-depth functional analyses of miR-124 have revealed several physiological and pathological roles exerted through interactions with other ncRNAs. Deciphering these RNA-mediated signaling networks and pathways is essential for understanding the potential impacts of dysregulated miRNA functions on bone biology. In this review, we aim to provide a comprehensive analysis of miR-124's involvement in bone physiology and pathology. We highlight the importance of miR-124 in controlling transcription factors and signaling pathways that promote bone growth. This review reveals therapeutic implications for the treatment of bone-related diseases.
Collapse
Affiliation(s)
- Rushil Kolipaka
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Induja Magesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - M.R. Ashok Bharathy
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - S. Karthik
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - I. Saranya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - N. Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| |
Collapse
|
10
|
Scuruchi M, Avenoso A, Aliquò F, Pantano A, Campo GM, Campo S, D'Ascola A. miR-21 attenuated inflammation targeting MyD88 in human chondrocytes stimulated with Hyaluronan oligosaccharides. Arch Biochem Biophys 2024; 759:110112. [PMID: 39111613 DOI: 10.1016/j.abb.2024.110112] [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: 04/02/2024] [Revised: 07/11/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
Inflammation is the body's response to injuries, which depends on numerous regulatory factors. Among them, miRNAs have gained much attention for their role in regulating inflammatory gene expression at multiple levels. In particular, miR-21 is up-regulated during the inflammatory response and reported to be involved in the resolution of inflammation by down-regulating pro-inflammatory mediators, including MyD88. Herein, we evaluated the regulatory effects of miR-21 on the TLR-4/MyD88 pathway in an in vitro model of 6-mer HA oligosaccharides-induced inflammation in human chondrocytes. The exposition of chondrocytes to 6-mer HA induced the activation of the TLR4/MyD88 pathway, which culminates in NF-kB activation. Changes in miR-21, TLR-4, MyD88, NLRP3 inflammasome, IL-29, Caspase1, MMP-9, iNOS, and COX-2 mRNA expression of 6-mer HA-stimulated chondrocytes were examined by qRT-PCR. Protein amounts of TLR-4, MyD88, NLRP3 inflammasome, p-ERK1/2, p-AKT, IL-29, caspase1, MMP-9, p-NK-kB p65 subunit, and IKB-a have been evaluated by ELISA kits. NO and PGE2 levels have been assayed by colorimetric and ELISA kits, respectively. HA oligosaccharides induced a significant increase in the expression of the above parameters, including NF-kB activity. The use of a miR-21 mimic attenuated MyD88 expression levels and the downstream effectors. On the contrary, treatment with a miR-21 inhibitor induced opposite effects. Interestingly, the use of a MyD88 siRNA confirmed MyD88 as the target of miR-21 action. Our results suggest that miR-21 expression could increase in an attempt to reduce the inflammatory response, targeting MyD88.
Collapse
Affiliation(s)
- Michele Scuruchi
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy.
| | - Angela Avenoso
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98122, Messina, Italy.
| | - Federica Aliquò
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98122, Messina, Italy
| | - Alice Pantano
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Giuseppe M Campo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Salvatore Campo
- Department of Biomedical and Dental Sciences and Morphofunctional Images, University of Messina, 98122, Messina, Italy
| | - Angela D'Ascola
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| |
Collapse
|
11
|
Ganguly K, Luthfikasari R, Randhawa A, Dutta SD, Patil TV, Acharya R, Lim KT. Stimuli-Mediated Macrophage Switching, Unraveling the Dynamics at the Nanoplatforms-Macrophage Interface. Adv Healthc Mater 2024; 13:e2400581. [PMID: 38637323 DOI: 10.1002/adhm.202400581] [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: 02/15/2024] [Revised: 04/01/2024] [Indexed: 04/20/2024]
Abstract
Macrophages play an essential role in immunotherapy and tissue regeneration owing to their remarkable plasticity and diverse functions. Recent bioengineering developments have focused on using external physical stimuli such as electric and magnetic fields, temperature, and compressive stress, among others, on micro/nanostructures to induce macrophage polarization, thereby increasing their therapeutic potential. However, it is difficult to find a concise review of the interaction between physical stimuli, advanced micro/nanostructures, and macrophage polarization. This review examines the present research on physical stimuli-induced macrophage polarization on micro/nanoplatforms, emphasizing the synergistic role of fabricated structure and stimulation for advanced immunotherapy and tissue regeneration. A concise overview of the research advancements investigating the impact of physical stimuli, including electric fields, magnetic fields, compressive forces, fluid shear stress, photothermal stimuli, and multiple stimulations on the polarization of macrophages within complex engineered structures, is provided. The prospective implications of these strategies in regenerative medicine and immunotherapeutic approaches are highlighted. This review will aid in creating stimuli-responsive platforms for immunomodulation and tissue regeneration.
Collapse
Affiliation(s)
- Keya Ganguly
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Institute of Forest Science, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Rachmi Luthfikasari
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Aayushi Randhawa
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Sayan Deb Dutta
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Tejal V Patil
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Rumi Acharya
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Ki-Taek Lim
- Department of Biosystems Engineering, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Interdisciplinary Program in Smart Agriculture, Kangwon National University, Chuncheon, 24341, Republic of Korea
| |
Collapse
|
12
|
Chen J, Hao Z, Li H, Wang J, Chen T, Wang Y, Shi G, Wang J, Wang Z, Zhang Z, Li J. Osteoporotic osseointegration: therapeutic hallmarks and engineering strategies. Theranostics 2024; 14:3859-3899. [PMID: 38994021 PMCID: PMC11234277 DOI: 10.7150/thno.96516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/05/2024] [Indexed: 07/13/2024] Open
Abstract
Osteoporosis is a systemic skeletal disease caused by an imbalance between bone resorption and formation. Current treatments primarily involve systemic medication and hormone therapy. However, these systemic treatments lack directionality and are often ineffective for locally severe osteoporosis, with the potential for complex adverse reactions. Consequently, treatment strategies using bioactive materials or external interventions have emerged as the most promising approaches. This review proposes twelve microenvironmental treatment targets for osteoporosis-related pathological changes, including local accumulation of inflammatory factors and reactive oxygen species (ROS), imbalance of mitochondrial dynamics, insulin resistance, disruption of bone cell autophagy, imbalance of bone cell apoptosis, changes in neural secretions, aging of bone cells, increased local bone tissue vascular destruction, and decreased regeneration. Additionally, this review examines the current research status of effective or potential biophysical and biochemical stimuli based on these microenvironmental treatment targets and summarizes the advantages and optimal parameters of different bioengineering stimuli to support preclinical and clinical research on osteoporosis treatment and bone regeneration. Finally, the review addresses ongoing challenges and future research prospects.
Collapse
Affiliation(s)
- Jiayao Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Zhuowen Hao
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Hanke Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Jianping Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Tianhong Chen
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan 430060, P.R. China
| | - Guang Shi
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Junwu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Zepu Wang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Zheyuan Zhang
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| | - Jingfeng Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan 430071, P.R. China
| |
Collapse
|
13
|
Wen C, Xu X, Zhang Y, Xia J, Liang Y, Xu L. Bone Targeting Nanoparticles for the Treatment of Osteoporosis. Int J Nanomedicine 2024; 19:1363-1383. [PMID: 38371454 PMCID: PMC10871045 DOI: 10.2147/ijn.s444347] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/30/2024] [Indexed: 02/20/2024] Open
Abstract
Osteoporosis (OP) affects millions of people worldwide, especially postmenopausal women and the elderly. Although current available anti-OP agents can show promise in slowing down bone resorption, most are not specifically delivered to the hard tissue, causing significant toxicity. A bone-targeted nanodrug delivery system can reduce side effects and precisely deliver drug candidates to the bone. This review focuses on the progress of bone-targeted nanoparticles in OP therapy. We enumerate the existing OP medications, types of bone-targeted nanoparticles and categorize pairs of the most common bone-targeting functional groups. Finally, we summarize the potential use of bone-targeted nanoparticles in OP treatment. Ongoing research into the development of targeted ligands and nanocarriers will continue to expand the possibilities of OP-targeted therapies into clinical application.
Collapse
Affiliation(s)
- Caining Wen
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Xiao Xu
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Yuanmin Zhang
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Jiang Xia
- Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong SAR, People’s Republic of China
| | - Yujie Liang
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, People’s Republic of China
- Engineering Research Center of Intelligent Rehabilitation, College of Rehabilitation Medicine, Jining Medical University, Jining, Shandong, People’s Republic of China
| | - Limei Xu
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, People’s Republic of China
| |
Collapse
|
14
|
Liu R, Liu S, Wu S, Xia M, Liu W, Wang L, Dong M, Niu W. Milk-Derived Small Extracellular Vesicles Promote Osteogenic Differentiation and Inhibit Inflammation via microRNA-21. Int J Mol Sci 2023; 24:13873. [PMID: 37762176 PMCID: PMC10531249 DOI: 10.3390/ijms241813873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/02/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic apical periodontitis (CAP) is a disease with characteristics of inflammation and bone loss. In this study, our objective was to examine the function of small extracellular vesicles (sEVs) obtained from milk in encouraging osteogenic differentiation and inhibiting inflammation by miR-21 in CAP. The expression of miR-21 was detected using qRT-PCR in human CAP samples. The impact of miR-21 on the process of osteogenic differentiation was investigated using CCK-8, qRT-PCR, immunofluorescence staining, and Western blot analysis. The evaluation of RAW 264.7 cell polarization and the assessment of inflammatory factor expression were conducted through qRT-PCR. The influence of sEVs on MC3T3-E1 cells and RAW 264.7 cells was examined, with a particular emphasis on the involvement of miR-21. In human CAP samples, a decrease in miR-21 expression was observed. MiR-21 increased the expression of osteogenesis-related genes and M2 polarization genes while decreasing the expression of M1 polarization genes and inflammatory cytokines. Treatment with milk-derived sEVs also promoted osteogenesis and M2 polarization while inhibiting M1 polarization and inflammation. Conversely, the addition of miR-21 inhibitors resulted in opposite effects. Our results indicated that sEVs derived from milk had a positive effect on bone formation and activation of anti-inflammatory (M2) macrophages and simultaneously reduced inflammation by regulating miR-21 in CAP.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ming Dong
- School of Stomatology, Dalian Medical University, Dalian 116044, China
| | - Weidong Niu
- School of Stomatology, Dalian Medical University, Dalian 116044, China
| |
Collapse
|
15
|
Anwar A, Sapra L, Gupta N, Ojha RP, Verma B, Srivastava RK. Fine-tuning osteoclastogenesis: An insight into the cellular and molecular regulation of osteoclastogenesis. J Cell Physiol 2023. [PMID: 37183350 DOI: 10.1002/jcp.31036] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/16/2023]
Abstract
Osteoclasts, the bone-resorbing cells, are essential for the bone remodeling process and are involved in the pathophysiology of several bone-related diseases. The extensive corpus of in vitro research and crucial mouse model studies in the 1990s demonstrated the key roles of monocyte/macrophage colony-stimulating factor, receptor activator of nuclear factor kappa B ligand (RANKL) and integrin αvβ3 in osteoclast biology. Our knowledge of the molecular mechanisms by which these variables control osteoclast differentiation and function has significantly advanced in the first decade of this century. Recent developments have revealed a number of novel insights into the fundamental mechanisms governing the differentiation and functional activity of osteoclasts; however, these mechanisms have not yet been adequately documented. Thus, in the present review, we discuss various regulatory factors including local and hormonal factors, innate as well as adaptive immune cells, noncoding RNAs (ncRNAs), etc., in the molecular regulation of the intricate and tightly regulated process of osteoclastogenesis. ncRNAs have a critical role as epigenetic controllers of osteoclast physiologic activities, including differentiation and bone resorption. The primary ncRNAs, which include micro-RNAs, circular RNAs, and long noncoding RNAs, form a complex network that affects gene transcription activities associated with osteoclast biological activity. Greater knowledge of the involvement of ncRNAs in osteoclast biological activities will contribute to the treatment and management of several skeletal diseases such as osteoporosis, osteoarthritis, rheumatoid arthritis, etc. Moreover, we further outline potential therapies targeting these regulatory pathways of osteoclastogenesis in distinct bone pathologies.
Collapse
Affiliation(s)
- Aleena Anwar
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Navita Gupta
- Department of Allied Health Sciences, Chitkara School of Health Sciences, Chitkara University, Chandigarh, Punjab, India
| | - Rudra P Ojha
- Department of Zoology, Nehru Gram Bharati University, Prayagraj, Uttar Pradesh, India
| | - Bhupendra Verma
- 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
|
16
|
Rouco H, García-García P, Briffault E, Diaz-Rodriguez P. Modulating osteoclasts with nanoparticles: A path for osteoporosis management? WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023:e1885. [PMID: 37037204 DOI: 10.1002/wnan.1885] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/27/2023] [Accepted: 02/21/2023] [Indexed: 04/12/2023]
Abstract
Osteoclasts are the cells responsible for the bone resorption process during bone remodeling. In a healthy situation, this process results from an equilibrium between new matrix formation by osteoblast and matrix resorption by osteoclast. Osteoporosis (OP) is a systemic bone disease characterized by a decreased bone mass density and alterations in bone microarchitecture, increasing fracture predisposition. Despite the variety of available therapies for OP management there is a growing gap in its treatment associated to the low patients´ adherence owing to concerns related with long-term efficacy or safety. This makes the development of new and safe treatments necessary. Among the newly developed strategies, the use of synthetic and natural nanoparticles to modulate osteoclasts differentiation, activity, apoptosis or crosstalk with osteoblasts have arisen. Synthetic nanoparticles exert their therapeutic effect either by loading antiresorptive drugs or including molecules for osteoclasts gene regulation. Moreover, this control over osteoclasts can be improved by their targeting to bone extracellular matrix or osteoclast membranes. Furthermore, natural nanoparticles, also known as extracellular vesicles, have been identified to play a key role in bone homeostasis. Consequently, these systems have been widely studied to control osteoblasts and osteoclasts under variable environments. Additionally, the ability to bioengineer extracellular vesicles has allowed to obtain biomimetic systems with desirable characteristics as drug carriers for osteoclasts. The analyzed information reveals the possibility of modulating osteoclasts by different mechanisms through nanoparticles decreasing bone resorption. These findings suggest that controlling osteoclast activity using nanoparticles has the potential to improve osteoporosis management. This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
- Helena Rouco
- School of Pharmacy, University of Nottingham, Nottingham, UK
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, I+D Farma Group (GI-1645), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Patricia García-García
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, I+D Farma Group (GI-1645), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Institute of Biomedical Technologies (ITB), La Laguna, Spain
| | - Erik Briffault
- Department of Chemical Engineering and Pharmaceutical Technology, Universidad de La Laguna, La Laguna, Spain
| | - Patricia Diaz-Rodriguez
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, I+D Farma Group (GI-1645), Facultad de Farmacia, Instituto de Materiales (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Institute of Biomedical Technologies (ITB), Universidad de La Laguna, La Laguna, Spain
| |
Collapse
|
17
|
Sikora M, Śmieszek A, Pielok A, Marycz K. MiR-21-5p regulates the dynamic of mitochondria network and rejuvenates the senile phenotype of bone marrow stromal cells (BMSCs) isolated from osteoporotic SAM/P6 mice. Stem Cell Res Ther 2023; 14:54. [PMID: 36978118 PMCID: PMC10053106 DOI: 10.1186/s13287-023-03271-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Progression of senile osteoporosis is associated with deteriorated regenerative potential of bone marrow-derived mesenchymal stem/stromal cells (BMSCs). According to the recent results, the senescent phenotype of osteoporotic cells strongly correlates with impaired regulation of mitochondria dynamics. Moreover, due to the ageing of population and growing osteoporosis incidence, more efficient methods concerning BMSCs rejuvenation are intensely investigated. Recently, miR-21-5p was reported to play a vital role in bone turnover, but its therapeutic mechanisms in progenitor cells delivered from senile osteoporotic patients remain unclear. Therefore, the goal of this paper was to investigate for the first time the regenerative potential of miR-21-5p in the process of mitochondrial network regulation and stemness restoration using the unique model of BMSCs isolated from senile osteoporotic SAM/P6 mice model. METHODS BMSCs were isolated from healthy BALB/c and osteoporotic SAM/P6 mice. We analysed the impact of miR-21-5p on the expression of crucial markers related to cells' viability, mitochondria reconstruction and autophagy progression. Further, we established the expression of markers vital for bone homeostasis, as well as defined the composition of extracellular matrix in osteogenic cultures. The regenerative potential of miR-21 in vivo was also investigated using a critical-size cranial defect model by computed microtomography and SEM-EDX imaging. RESULTS MiR-21 upregulation improved cells' viability and drove mitochondria dynamics in osteoporotic BMSCs evidenced by the intensification of fission processes. Simultaneously, miR-21 enhanced the osteogenic differentiation of BMSCs evidenced by increased expression of Runx-2 but downregulated Trap, as well as improved calcification of extracellular matrix. Importantly, the analyses using the critical-size cranial defect model indicated on a greater ratio of newly formed tissue after miR-21 application, as well as upregulated content of calcium and phosphorus within the defect site. CONCLUSIONS Our results demonstrate that miR-21-5p regulates the fission and fusion processes of mitochondria and facilitates the stemness restoration of senile osteoporotic BMSCs. At the same time, it enhances the expression of RUNX-2, while reduces TRAP accumulation in the cells with deteriorated phenotype. Therefore, miR-21-5p may bring a novel molecular strategy for senile osteoporosis diagnostics and treatment.
Collapse
Affiliation(s)
- Mateusz Sikora
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences Wroclaw, Norwida 27B St, 50-375, Wrocław, Poland
| | - Agnieszka Śmieszek
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences Wroclaw, Norwida 27B St, 50-375, Wrocław, Poland
| | - Ariadna Pielok
- Department of Experimental Biology, The Faculty of Biology and Animal Science, University of Environmental and Life Sciences Wroclaw, Norwida 27B St, 50-375, Wrocław, Poland
| | - Krzysztof Marycz
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, One Shields Avenue, Davis, CA, 95616-8739, USA.
- International Institute of Translational Medicine, Jesionowa 11 Street, 55-124, Malin, Poland.
| |
Collapse
|
18
|
Intravaia JT, Graham T, Kim HS, Nanda HS, Kumbar SG, Nukavarapu SP. Smart Orthopedic Biomaterials and Implants. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023; 25:100439. [PMID: 36642994 PMCID: PMC9835562 DOI: 10.1016/j.cobme.2022.100439] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Musculoskeletal injuries including bone defects continue to present a significant challenge in orthopedic surgery due to suboptimal healing. Bone reconstruction strategies focused on the use of biological grafts and bone graft substitutes in the form of biomaterials-based 3D structures in fracture repair. Recent advances in biomaterials science and engineering have resulted in the creation of intricate 3D bone-mimicking structures that are mechanically stable, biodegradable, and bioactive to support bone regeneration. Current efforts are focused on improving the biomaterial and implant physicochemical properties to promote interactions with the host tissue and osteogenesis. The "smart" biomaterials and their 3D structures are designed to actively interact with stem/progenitor cells and the extracellular matrix (ECM) to influence the local environment towards osteogenesis and de novo tissue formation. This article will summarize such smart biomaterials and the methodologies to apply either internal or external stimuli to control the tissue healing microenvironment. A particular emphasis is also made on the use of smart biomaterials and strategies to create functional bioactive implants for bone defect repair and regeneration.
Collapse
Affiliation(s)
| | - Trevon Graham
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Hyun S. Kim
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Himansu S. Nanda
- Terasaki Institute, University of California, Los Angeles, CA, USA
- Mechanical Engineering, IIITDM, Jabalpur, MP, India
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Syam P. Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
19
|
Lei C, Song JH, Li S, Zhu YN, Liu MY, Wan MC, Mu Z, Tay FR, Niu LN. Advances in materials-based therapeutic strategies against osteoporosis. Biomaterials 2023; 296:122066. [PMID: 36842238 DOI: 10.1016/j.biomaterials.2023.122066] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 02/16/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023]
Abstract
Osteoporosis is caused by the disruption in homeostasis between bone formation and bone resorption. Conventional management of osteoporosis involves systematic drug administration and hormonal therapy. These treatment strategies have limited curative efficacy and multiple adverse effects. Biomaterials-based therapeutic strategies have recently emerged as promising alternatives for the treatment of osteoporosis. The present review summarizes the current status of biomaterials designed for managing osteoporosis. The advantages of biomaterials-based strategies over conventional systematic drug treatment are presented. Different anti-osteoporotic delivery systems are concisely addressed. These materials include injectable hydrogels and nanoparticles, as well as anti-osteoporotic bone tissue engineering materials. Fabrication techniques such as 3D printing, electrostatic spinning and artificial intelligence are appraised in the context of how the use of these adjunctive techniques may improve treatment efficacy. The limitations of existing biomaterials are critically analyzed, together with deliberation of the future directions in biomaterials-based therapies. The latter include discussion on the use of combination strategies to enhance therapeutic efficacy in the osteoporosis niche.
Collapse
Affiliation(s)
- Chen Lei
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jing-Han Song
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Song Li
- School of Stomatology, Xinjiang Medical University. Urumqi 830011, China
| | - Yi-Na Zhu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Ming-Yi Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Mei-Chen Wan
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhao Mu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Franklin R Tay
- The Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Li-Na Niu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| |
Collapse
|
20
|
Magnetic Hydroxyapatite Composite Nanoparticles for Augmented Differentiation of MC3T3-E1 Cells for Bone Tissue Engineering. Mar Drugs 2023; 21:md21020085. [PMID: 36827126 PMCID: PMC9960960 DOI: 10.3390/md21020085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/16/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
Progressive aging harms bone tissue structure and function and, thus, requires effective therapies focusing on permanent tissue regeneration rather than partial cure, beginning with regenerative medicine. Due to advances in tissue engineering, stimulating osteogenesis with biomimetic nanoparticles to create a regenerative niche has gained attention for its efficacy and cost-effectiveness. In particular, hydroxyapatite (HAP, Ca10(PO4)6(OH)2) has gained significant interest in orthopedic applications as a major inorganic mineral of native bone. Recently, magnetic nanoparticles (MNPs) have also been noted for their multifunctional potential for hyperthermia, MRI contrast agents, drug delivery, and mechanosensitive receptor manipulation to induce cell differentiation, etc. Thus, the present study synthesizes HAP-decorated MNPs (MHAP NPs) via the wet chemical co-precipitation method. Synthesized MHAP NPs were evaluated against the preosteoblast MC3T3-E1 cells towards concentration-dependent cytotoxicity, proliferation, morphology staining, ROS generation, and osteogenic differentiation. The result evidenced that MHAP NPs concentration up to 10 µg/mL was non-toxic even with the time-dependent proliferation studies. As nanoparticle concentration increased, FACS apoptosis assay and ROS data showed a significant rise in apoptosis and ROS generation. The MC3T3-E1 cells cocultured with 5 µg/mL MHAP NPs showed significant osteogenic differentiation potential. Thus, MHAP NPs synthesized with simple wet chemistry could be employed in bone regenerative therapy.
Collapse
|
21
|
Yang J, Wu J, Guo Z, Zhang G, Zhang H. Iron Oxide Nanoparticles Combined with Static Magnetic Fields in Bone Remodeling. Cells 2022; 11:cells11203298. [PMID: 36291164 PMCID: PMC9600888 DOI: 10.3390/cells11203298] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/18/2022] [Indexed: 11/27/2022] Open
Abstract
Iron oxide nanoparticles (IONPs) are extensively used in bone-related studies as biomaterials due to their unique magnetic properties and good biocompatibility. Through endocytosis, IONPs enter the cell where they promote osteogenic differentiation and inhibit osteoclastogenesis. Static magnetic fields (SMFs) were also found to enhance osteoblast differentiation and hinder osteoclastic differentiation. Once IONPs are exposed to an SMF, they become rapidly magnetized. IONPs and SMFs work together to synergistically enhance the effectiveness of their individual effects on the differentiation and function of osteoblasts and osteoclasts. This article reviewed the individual and combined effects of different types of IONPs and different intensities of SMFs on bone remodeling. We also discussed the mechanism underlying the synergistic effects of IONPs and SMFs on bone remodeling.
Collapse
Affiliation(s)
- Jiancheng Yang
- Department of Spine Surgery, People’s Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen 518109, China
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Jiawen Wu
- Department of Spine Surgery, People’s Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen 518109, China
| | - Zengfeng Guo
- Department of Spine Surgery, People’s Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen 518109, China
| | - Gejing Zhang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Hao Zhang
- Department of Spine Surgery, People’s Hospital of Longhua, Affiliated Hospital of Southern Medical University, Shenzhen 518109, China
- Correspondence: ; Tel.: +86-13823352822
| |
Collapse
|
22
|
Esteves M, Abreu R, Fernandes H, Serra-Almeida C, Martins PAT, Barão M, Cristóvão AC, Saraiva C, Ferreira R, Ferreira L, Bernardino L. MicroRNA-124-3p-enriched small extracellular vesicles as a therapeutic approach for Parkinson's disease. Mol Ther 2022; 30:3176-3192. [PMID: 35689381 PMCID: PMC9552816 DOI: 10.1016/j.ymthe.2022.06.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 05/10/2022] [Accepted: 06/06/2022] [Indexed: 12/09/2022] Open
Abstract
Parkinson's disease is a neurodegenerative disease characterized by the loss of dopaminergic neurons in the substantia nigra with no effective cure available. MicroRNA-124 has been regarded as a promising therapeutic entity for Parkinson's disease due to its pro-neurogenic and neuroprotective roles. However, its efficient delivery to the brain remains challenging. Here, we used umbilical cord blood mononuclear cell-derived extracellular vesicles as a biological vehicle to deliver microRNA (miR)-124-3p and evaluate its therapeutic effects in a mouse model of Parkinson's disease. In vitro, miR-124-3p-loaded small extracellular vesicles induced neuronal differentiation in subventricular zone neural stem cell cultures and protected N27 dopaminergic cells against 6-hydroxydopamine-induced toxicity. In vivo, intracerebroventricularly administered small extracellular vesicles were detected in the subventricular zone lining the lateral ventricles and in the striatum and substantia nigra, the brain regions most affected by the disease. Most importantly, although miR-124-3p-loaded small extracellular vesicles did not increase the number of new neurons in the 6-hydroxydopamine-lesioned striatum, the formulation protected dopaminergic neurons in the substantia nigra and striatal fibers, which fully counteracted motor behavior symptoms. Our findings reveal a novel promising therapeutic application of small extracellular vesicles as delivery agents for miR-124-3p in the context of Parkinson's disease.
Collapse
Affiliation(s)
- Marta Esteves
- Health Sciences Research Centre (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Ricardo Abreu
- Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht University, Maastricht 6200, the Netherlands; CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Cantanhede, Portugal
| | - Hugo Fernandes
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Cantanhede, Portugal; Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Catarina Serra-Almeida
- Health Sciences Research Centre (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Patrícia A T Martins
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Cantanhede, Portugal
| | - Marta Barão
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Cantanhede, Portugal
| | - Ana Clara Cristóvão
- Health Sciences Research Centre (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal; Neurosov, UBImedical, EM506, University of Beira Interior, Covilhã, Portugal
| | - Cláudia Saraiva
- Health Sciences Research Centre (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Raquel Ferreira
- Health Sciences Research Centre (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal
| | - Lino Ferreira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Cantanhede, Portugal; Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Liliana Bernardino
- Health Sciences Research Centre (CICS-UBI), Faculty of Health Sciences, University of Beira Interior, 6200-506 Covilhã, Portugal.
| |
Collapse
|
23
|
Wang J, Cui Y, Liu H, Li S, Sun S, Xu H, Peng C, Wang Y, Wu D. MicroRNA-loaded biomaterials for osteogenesis. Front Bioeng Biotechnol 2022; 10:952670. [PMID: 36199361 PMCID: PMC9527286 DOI: 10.3389/fbioe.2022.952670] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
The large incidence of bone defects in clinical practice increases not only the demand for advanced bone transplantation techniques but also the development of bone substitute materials. A variety of emerging bone tissue engineering materials with osteogenic induction ability are promising strategies for the design of bone substitutes. MicroRNAs (miRNAs) are a class of non-coding RNAs that regulate intracellular protein expression by targeting the non-coding region of mRNA3′-UTR to play an important role in osteogenic differentiation. Several miRNA preparations have been used to promote the osteogenic differentiation of stem cells. Therefore, multiple functional bone tissue engineering materials using miRNA as an osteogenic factor have been developed and confirmed to have critical efficacy in promoting bone repair. In this review, osteogenic intracellular signaling pathways mediated by miRNAs are introduced in detail to provide a clear understanding for future clinical treatment. We summarized the biomaterials loaded with exogenous cells engineered by miRNAs and biomaterials directly carrying miRNAs acting on endogenous stem cells and discussed their advantages and disadvantages, providing a feasible method for promoting bone regeneration. Finally, we summarized the current research deficiencies and future research directions of the miRNA-functionalized scaffold. This review provides a summary of a variety of advanced miRNA delivery system design strategies that enhance bone regeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Dankai Wu
- *Correspondence: Yanbing Wang, ; Dankai Wu,
| |
Collapse
|
24
|
Biswas L, Niveria K, Verma AK. Paradoxical role of reactive oxygen species in bone remodelling: implications in osteoporosis and possible nanotherapeutic interventions. EXPLORATION OF MEDICINE 2022. [DOI: 10.37349/emed.2022.00102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Osteoporosis is a metabolic bone disorder that affects both sexes and is the most common cause of fractures. Osteoporosis therapies primarily inhibit osteoclast activity, and are seldom designed to trigger new bone growth thereby frequently causing severe systemic adverse effects. Physiologically, the intracellular redox state depends on the ratio of pro-oxidants, oxidizing agents (reactive oxygen species, ROS) and antioxidants. ROS is the key contributor to oxidative stress in osteoporosis as changes in redox state are responsible for dynamic bone remodeling and bone regeneration. Imbalances in ROS generation vs. antioxidant systems play a pivotal role in pathogenesis of osteoporosis, stimulating osteoblasts and osteocytes towards osteoclastogenesis. ROS prevents mineralization and osteogenesis, causing increased turnover of bone loss. Alternatively, antioxidants either directly or indirectly, contribute to activation of osteoblasts leading to differentiation and mineralization, thereby reducing osteoclastogenesis. Owing to the unpredictability of immune responsiveness and reported adverse effects, despite promising outcomes from drugs against oxidative stress, treatment in clinics targeting osteoclast has been limited. Nanotechnology-mediated interventions have gained remarkable superiority over other treatment modalities in regenerative medicine. Nanotherapeutic approaches exploit the antioxidant properties of nanoparticles for targeted drug delivery to trigger bone repair, by enhancing their osteogenic and anti-osteoclastogenic potentials to influence the biocompatibility, mechanical properties and osteoinductivity. Therefore, exploiting nanotherapeutics for maintaining the differentiation and proliferation of osteoblasts and osteoclasts is quintessential.
Collapse
Affiliation(s)
- Largee Biswas
- 1Nanobiotech lab, Department of Zoology, Kirori Mal College, University of Delhi, Delhi 110007, India
| | - Karishma Niveria
- 1Nanobiotech lab, Department of Zoology, Kirori Mal College, University of Delhi, Delhi 110007, India
| | - Anita Kamra Verma
- 1Nanobiotech lab, Department of Zoology, Kirori Mal College, University of Delhi, Delhi 110007, India 2Fellow, Delhi School of Public Health, Institution of Eminence, University of Delhi, Delhi 110007, India
| |
Collapse
|
25
|
Tian H, Chen F, Wang Y, Liu Y, Ma G, Zhao Y, Ma Y, Tian T, Ma R, Yu Y, Wang D. Nur77 Prevents Osteoporosis by Inhibiting the NF-κB Signalling Pathway and Osteoclast Differentiation. J Cell Mol Med 2022; 26:2163-2176. [PMID: 35181992 PMCID: PMC8995449 DOI: 10.1111/jcmm.17238] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/24/2022] [Accepted: 01/31/2022] [Indexed: 01/16/2023] Open
Abstract
Inflammation is a major risk factor for osteoporosis, and reducing inflammatory levels is important for the prevention of osteoporosis. Although nuclear receptor 77 (Nur77) protects against inflammation in a variety of diseases, its role in osteoporosis is unknown. Therefore, the main purpose of this study was to investigate the osteoprotective and anti‐inflammatory effects of Nur77. The microCT and haematoxylin and eosin staining results indicated that knockout of Nur77 accelerated femoral bone loss in mice. The enzyme‐linked immunosorbent assay (ELISA) results showed that knockout of Nur77 increased the serum levels of hsCRP and IL‐6. The expression levels of NF‐κB, IL‐6, TNF‐α and osteoclastogenesis factors (TRAP, NFATC1, Car2, Ctsk) in the femurs of Nur77 knockout mice were increased significantly. Furthermore, in vitro, shNur77 promoted the differentiation of RAW264.7 cells into osteoclasts by activating NF‐κB, which was confirmed by PDTC treatment. Mechanistically, Nur77 inhibited osteoclast differentiation by inducing IκB‐α and suppressing IKK‐β. In RAW264.7 cells, overexpression of Nur77 alleviated inflammation induced by siIκB‐α, while siIKK‐β alleviated inflammation induced by shNur77. Consistent with the in vivo studies, we found that compared with control group, older adults with high serum hsCRP levels were more likely to suffer from osteoporosis (OR = 1.76, p < 0.001). Our data suggest that Nur77 suppresses osteoclast differentiation by inhibiting the NF‐κB signalling pathway, strongly supporting the notion that Nur77 has the potential to prevent and treat osteoporosis.
Collapse
Affiliation(s)
- Huanlian Tian
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.,Department of Health Statistics, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Feng Chen
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yingfang Wang
- Department of General Practice Medicine, Nanyang Centre Hospital, Nanyang, Henan, China
| | - Yixuan Liu
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Guojing Ma
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yuhong Zhao
- Department of Clinical Epidemiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yanan Ma
- Department of Health Statistics, School of Public Health, China Medical University, Shenyang, Liaoning, China
| | - Tingting Tian
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ruze Ma
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yang Yu
- Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China
| | - Difei Wang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
26
|
Deng Y, Wei W, Tang P. Applications of Calcium-Based Nanomaterials in Osteoporosis Treatment. ACS Biomater Sci Eng 2022; 8:424-443. [PMID: 35080365 DOI: 10.1021/acsbiomaterials.1c01306] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
With rapidly aging populations worldwide, osteoporosis has become a serious global public health problem. Caused by disordered systemic bone remodeling, osteoporosis manifests as progressive loss of bone mass and microarchitectural deterioration of bone tissue, increasing the risk of fractures and eventually leading to osteoporotic fragility fractures. As fracture risk increases, antiosteoporosis treatments transition from nonpharmacological management to pharmacological intervention, and finally to the treatment of fragility fractures. Calcium-based nanomaterials (CBNMs) have unique advantages in osteoporosis treatment because of several characteristics including similarity to natural bone, excellent biocompatibility, easy preparation and functionalization, low pH-responsive disaggregation, and inherent pro-osteogenic properties. By combining additional ingredients, CBNMs can play multiple roles to construct antiosteoporotic biomaterials with different forms. This review covers recent advances in CBNMs for osteoporosis treatment. For ease of understanding, CBNMs for antiosteoporosis treatment can be classified as locally applied CBNMs, such as implant coatings and filling materials for osteoporotic bone regeneration, and systemically administered CBNMs for antiosteoporosis treatment. Locally applied CBNMs for osteoporotic bone regeneration develop faster than the systemically administered CBNMs, an important consideration given the serious outcomes of fragility fractures. Nevertheless, many innovations in construction strategies and preparation methods have been applied to build systemically administered CBNMs. Furthermore, with increasing interest in delaying osteoporosis progression and avoiding fragility fracture occurrence, research into systemic administration of CBNMs for antiosteoporosis treatment will have more development prospects. Deep understanding of the CBNM preparation process and optimizing CBNM properties will allow for increased application of CBNMs in osteoporosis treatments in the future.
Collapse
Affiliation(s)
- Yuan Deng
- Department of Orthopedics, Fourth Medical Center, General Hospital of Chinese PLA, Beijing 100000, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering Institute of Process Engineering Chinese Academy of Sciences No. 1 Bei-Er-Tiao, Beijing 100190, P. R. China
| | - Peifu Tang
- Department of Orthopedics, Fourth Medical Center, General Hospital of Chinese PLA, Beijing 100000, China
| |
Collapse
|
27
|
Bai RJ, Li YS, Zhang FJ. Osteopontin, a bridge links osteoarthritis and osteoporosis. Front Endocrinol (Lausanne) 2022; 13:1012508. [PMID: 36387862 PMCID: PMC9649917 DOI: 10.3389/fendo.2022.1012508] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent joint disease characterized by degradation of articular cartilage, inflammation, and changes in periarticular and subchondral bone of joints. Osteoporosis (OP) is another systemic skeletal disease characterized by low bone mass and bone mineral density (BMD) accompanied by microarchitectural deterioration in bone tissue and increased bone fragility and fracture risk. Both OA and OP are mainly affected on the elderly people. Recent studies have shown that osteopontin (OPN) plays a vital role in bone metabolism and homeostasis. OPN involves these biological activities through participating in the proliferation, migration, differentiation, and adhesion of several bone-related cells, including chondrocytes, synoviocytes, osteoclasts, osteoblasts, and marrow mesenchymal stem cells (MSCs). OPN has been demonstrated to be closely related to the occurrence and development of many bone-related diseases, such as OA and OP. This review summarizes the role of OPN in regulating inflammation activity and bone metabolism in OA and OP. Furthermore, some drugs that targeted OPN to treat OA and OP are also summarized in the review. However, the complex mechanism of OPN in regulating OA and OP is not fully elucidated, which drives us to explore the depth effect of OPN on these two bone diseases.
Collapse
Affiliation(s)
- Rui-Jun Bai
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yu-Sheng Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
- *Correspondence: Yu-Sheng Li, ; Fang-Jie Zhang,
| | - Fang-Jie Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan, China
- Department of Emergency Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Yu-Sheng Li, ; Fang-Jie Zhang,
| |
Collapse
|
28
|
Marycz K, Śmieszek A, Kornicka-Garbowska K, Pielok A, Janeczek M, Lipińska A, Nikodem A, Filipiak J, Sobierajska P, Nedelec JM, Wiglusz RJ. Novel Nanohydroxyapatite (nHAp)-Based Scaffold Doped with Iron Oxide Nanoparticles (IO), Functionalized with Small Non-Coding RNA (miR-21/124) Modulates Expression of Runt-Related Transcriptional Factor 2 and Osteopontin, Promoting Regeneration of Osteoporotic Bone in Bilateral Cranial Defects in a Senescence-Accelerated Mouse Model (SAM/P6). PART 2. Int J Nanomedicine 2021; 16:6049-6065. [PMID: 34511905 PMCID: PMC8418301 DOI: 10.2147/ijn.s316240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/28/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose Healing of osteoporotic defects is challenging and requires innovative approaches to elicit molecular mechanisms promoting osteoblasts-osteoclasts coupling and bone homeostasis. Methods Cytocompatibility and biocompatibility of previously characterised nanocomposites, i.e Ca5(PO4)3OH/Fe3O4 (later called nHAp/IO) functionalised with microRNAs (nHAp/IO@miR-21/124) was tested. In vitro studies were performed using a direct co-culture system of MC3T3-E1 pre-osteoblast and 4B12 pre-osteoclasts. The analysis included determination of nanocomposite influence on cultures morphology (confocal imaging), viability and metabolic activity (Alamar Blue assay). Pro-osteogenic signals were identified at mRNA, miRNA and protein level with RT-qPCR, Western blotting and immunocytochemistry. Biocompatibility of biomaterials was tested using bilateral cranial defect performed on a senescence-accelerated mouse model, ie SAM/P6 and Balb/c. The effect of biomaterial on the process of bone healing was monitored using microcomputed tomography. Results The nanocomposites promoted survival and metabolism of bone cells, as well as enhanced functional differentiation of pre-osteoblasts MC3T3-E1 in co-cultures with pre-osteoclasts. Differentiation of MC3T3-E1 driven by nHAp/IO@miR-21/124 nanocomposite was manifested by improved extracellular matrix differentiation and up-regulation of pro-osteogenic transcripts, ie late osteogenesis markers. The nanocomposite triggered bone healing in a cranial defect model in SAM/P6 mice and was replaced by functional bone in Balb/c mice. Conclusion This study demonstrates that the novel nanocomposite nHAp/IO can serve as a platform for therapeutic miRNA delivery. Obtained nanocomposite elicit pro-osteogenic signals, decreasing osteoclasts differentiation, simultaneously improving osteoblasts metabolism and their transition toward pre-osteocytes and bone mineralisation. The proposed scaffold can be an effective interface for in situ regeneration of osteoporotic bone, especially in elderly patients.
Collapse
Affiliation(s)
- Krzysztof Marycz
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Wroclaw, 50-375, Poland.,International Institute of Translational Medicine, Malin, 55-124, Poland
| | - Agnieszka Śmieszek
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Wroclaw, 50-375, Poland
| | - Katarzyna Kornicka-Garbowska
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Wroclaw, 50-375, Poland.,International Institute of Translational Medicine, Malin, 55-124, Poland
| | - Ariadna Pielok
- Department of Experimental Biology, Wroclaw University of Environmental and Life Sciences, Wroclaw, 50-375, Poland
| | - Maciej Janeczek
- Department of Biostructure and Animal Physiology, Wroclaw University of and Life Sciences, Wroclaw, 51-631, Poland
| | - Anna Lipińska
- Department of Biostructure and Animal Physiology, Wroclaw University of and Life Sciences, Wroclaw, 51-631, Poland
| | - Anna Nikodem
- Department of Mechanics, Materials and Biomedical Engineering, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wrocław, Poland
| | - Jarosław Filipiak
- Department of Mechanics, Materials and Biomedical Engineering, Wroclaw University of Science and Technology, Wybrzeze Wyspianskiego 27, 50-370 Wrocław, Poland
| | - Paulina Sobierajska
- Institute of Low Temperature and Structure Research, Polish Academy of Sciences, Okolna 2, 50-422 Wroclaw, Poland
| | - Jean-Marie Nedelec
- Universite Clermont Auvergne, Clermont Auvergne INP, CNRS, ICCF, Clermont-Ferrand, France
| | - Rafał J Wiglusz
- International Institute of Translational Medicine, Malin, 55-124, Poland.,Institute of Low Temperature and Structure Research, Polish Academy of Sciences, Okolna 2, 50-422 Wroclaw, Poland
| |
Collapse
|