1
|
Yakut S, Tarakçı Gençer B, Yalçın MH, Aydın S, Yüksel H. Investigation of the effects of silymarin and vitamin C on kidney damage and aquaporin-2 downregulation in lithium-induced nephrogenic diabetes insipidus in rats. Drug Chem Toxicol 2025:1-11. [PMID: 39809261 DOI: 10.1080/01480545.2025.2450475] [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: 10/31/2024] [Revised: 12/13/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025]
Abstract
Although lithium (LIT) therapy is key in managing bipolar disorder long-term, prolonged use significantly contributes to acquired Nephrogenic Diabetes Insipidus (NDI). This study examined whether combining Silymarin (SIL) with Vitamin C (Vit C) enhances protection against lithium-induced nephrotoxicity in rats, comparing their individual antioxidant effects as well. Rats subjected to Li exposure were provided with a standard commercial diet supplemented with 80 mmol LiCl per kilogram for 28 days. Concurrently, SIL and Vit C were administered orally at dosages of 200 and 100 mg/kg body weight, respectively, throughout the 28 days. The study assessed levels of reactive oxygen species (ROS), glutathione (GSH), and malondialdehyde (MDA), as well as the enzyme activity of superoxide dismutase (SOD), to evaluate the protective effects of SIL and Vit C against oxidative stress. Aquaporin-2 (AQP2) levels in kidney tissues were evaluated using immunohistochemistry and ELISA. Serum and urine parameters (sodium, potassium, creatinine, blood urea nitrogen [BUN], and urea) and serum lithium levels were also measured. Lithium-induced nephrotoxicity showed increased renal toxicity markers and decreased antioxidant enzyme activity. SIL administration significantly reduced markers of kidney tissue toxicity, increased antioxidant enzyme activities, regulated the aforementioned physiological parameters in blood and urine, and downregulated AQP2 expression in the kidney. However, Vit C administration did not demonstrate a significant protective effect against lithium-induced renal toxicity. These findings indicate that SIL effectively protects against lithium-induced nephrotoxicity, whereas Vitamin C does not exhibit this protective effect.
Collapse
Affiliation(s)
- Seda Yakut
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Berrin Tarakçı Gençer
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Fırat University, Elâzığ, Turkey
| | - Mehmet Hanifi Yalçın
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Fırat University, Elâzığ, Turkey
| | - Süleyman Aydın
- Department of Biochemistry, Faculty of Medicine, Fırat University, Elâzığ, Turkey
| | - Hayati Yüksel
- Department of Pathology, Faculty of Veterinary Medicine, Bingöl University, Bingöl, Turkey
| |
Collapse
|
2
|
Cao C, Li J, Cui W, Dai J, Guan Z, Wang D, Zhao X. Metalomics Revealed that Changes of Serum Elements were Associated with Oxidative Stress-Induced Inflammation of Cortex in a Mouse Model of Autism. Biol Trace Elem Res 2024:10.1007/s12011-024-04501-0. [PMID: 39733022 DOI: 10.1007/s12011-024-04501-0] [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: 10/08/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder emerging during early childhood. However, the mechanism underlying the pathogenesis of ASD remains unclear. This study investigated the alterations of elements in serum and prefrontal cortex of BTBR T + tf/J (BTBR) mice and potential mechanisms. The male BTBR mice were used for experimental group and C57BL/6 J (C57) mice were used for control group (n = 15). After behavioral tests were monitored, serum and prefrontal cortex of mice were analyzed by ICP-MS. The results demonstrated that the level of copper (Cu) was increased, and the levels of calcium (Ca), magnesium (Mg), selenium (Se), cobalt (Co), iron (Fe) and zinc (Zn) were decreased in BTBR mice compared to C57 mice (p < 0.01). The levels of above differential elements in serum demonstrated positive correlations with those in prefrontal cortex. Meanwhile, differential elements in prefrontal cortex had correlations with the total distance traveled (open field test) and the number of marbles buried (marble burying test) in BTBR mice (p < 0.05 or p < 0.01). The abnormally changed elements in serum might cross blood-brain-barrier into the brain and lead to oxidative stress, causing inflammation. Furtherly, the levels of inflammation-related indicators including tumor necrosis factor-alpha (TNF-α), nuclear factor kappa-B (NF-κB), interleukin-6 (IL-6) and interleukin-1β (IL-1β) were increased in prefrontal cortex of BTBR mice (p < 0.01), which were consistent with the aforementioned results. Our study suggested that the abnormal elements in the serum of BTBR mice may cause oxidative stress and inflammation in prefrontal cortex, which might contribute to increase the understanding of ASD pathogenesis.
Collapse
Affiliation(s)
- Can Cao
- School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China
| | - Jian Li
- School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China
| | - Wenqi Cui
- School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China
| | - Jiaohua Dai
- School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China
| | - Zhiyu Guan
- School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China
| | - Dan Wang
- School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China.
| | - Xiujuan Zhao
- School of Public Health, Harbin Medical University, 194 Xuefu Road, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
3
|
Zhu N, Ni H, Guo S, Shen YQ, Chen Q. Bone complications of cancer treatment. Cancer Treat Rev 2024; 130:102828. [PMID: 39270364 DOI: 10.1016/j.ctrv.2024.102828] [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: 05/23/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024]
Abstract
With the advancements in conventional treatment modalities such as radiation, chemotherapy, and surgery, as well as the emergence of immunotherapy, the overall cure rate for solid tumor malignancies has experienced a significant increase. However, it is unfortunate that exposure to cancer treatments can have detrimental effects on the function of osteoblasts and osteoclasts, disturbing bone metabolic homeostasis in patients, as well as causing damage to bone marrow cells and other bone tissues. Consequently, certain tumor treatment options may pose a risk for subsequent bone diseases. Common bone disorders associated with cancer treatment include osteonecrosis, bone loss, and secondary bone tumors. (1)Cancer treatment-related osteonecrosis is primarily linked to the use of radiation therapy and certain chemicals, such as bisphosphonates, denosumab, antiangiogenic agents, and immunomodulators. It has been observed that high-dose radiation therapy is more likely to result in osteonecrosis. (2)Chemicals and hormones, particularly sex hormones, glucocorticoids, and thyroid hormones or thyrotropic hormones, are among the factors that can contribute to cancer treatment-related bone loss. (3)Secondary bone tumors differ from metastases originating from primary tumors, and radiotherapy plays a significant role in their development, while chemotherapy may also exert some influence. Radiogenic secondary bone tumors are predominantly malignant, with osteosarcoma being the most common type. Chemotherapy may be a risk factor for the relatively rare occurrence of secondary Ewing sarcoma of the bone. These treatment-related bone disorders have a considerable adverse impact on the prognosis of cancer patients. Hence, it is imperative to prioritize the bone health of patients undergoing cancer treatment and give it further attention.
Collapse
Affiliation(s)
- Nanxi Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hao Ni
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Shengzhao Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Ju R, Gao X, Zhang C, Tang W, Tian W, He M. Exogenous MSC based tissue regeneration: a review of immuno-protection strategies from biomaterial scaffolds. J Mater Chem B 2024; 12:8868-8882. [PMID: 39171946 DOI: 10.1039/d4tb00778f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Mesenchymal stem cell (MSC)-based tissue engineering holds great potential for regenerative medicine as a means of replacing damaged or lost tissues to restore their structure and function. However, the efficacy of MSC-based regeneration is frequently limited by the low survival rate and limited survival time of transplanted MSCs. Despite the inherent immune privileges of MSCs, such as low expression of major histocompatibility complex antigens, tolerogenic properties, local immunosuppressive microenvironment creation, and induction of immune tolerance, immune rejection remains a major obstacle to their survival and regenerative potential. Evidence suggests that immune protection strategies can enhance MSC therapeutic efficacy by prolonging their survival and maintaining their biological functions. Among various immune protection strategies, biomaterial-based scaffolds or cell encapsulation systems that mediate the interaction between transplanted MSCs and the host immune system or spatially isolate MSCs from the immune system for a specific time period have shown great promise. In this review, we provide a comprehensive overview of these biomaterial-based immune protection strategies employed for exogenous MSCs, highlighting the crucial role of modulating the immune microenvironment. Each strategy is critically examined, discussing its strengths, limitations, and potential applications in MSC-based tissue engineering. By elucidating the mechanisms behind immune rejection and exploring immune protection strategies, we aim to address the challenges faced by MSC-based tissue engineering and pave the way for enhancing the therapeutic outcomes of MSC therapies. The insights gained from this review will contribute to the development of more effective strategies to protect transplanted MSCs from immune rejection and enable their successful application in regenerative medicine.
Collapse
Affiliation(s)
- Rongbai Ju
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xinhui Gao
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chi Zhang
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Wei Tang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Weidong Tian
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Min He
- Engineering Research Center of Oral Translational Medicine, Ministry of Education, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
- National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
5
|
Chang L, Luo Y, Li W, Liu F, Guo J, Dai B, Tong W, Qin L, Wang J, Xu J. A comparative study on the effects of biodegradable high-purity magnesium screw and polymer screw for fixation in epiphyseal trabecular bone. Regen Biomater 2024; 11:rbae095. [PMID: 39346687 PMCID: PMC11427752 DOI: 10.1093/rb/rbae095] [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: 03/06/2024] [Revised: 06/11/2024] [Accepted: 07/08/2024] [Indexed: 10/01/2024] Open
Abstract
With mechanical strength close to cortical bone, biodegradable and osteopromotive properties, magnesium (Mg)-based implants are promising biomaterials for orthopedic applications. However, during the degradation of such implants, there are still concerns on the potential adverse effects such as formation of cavities, osteolytic phenomena and chronic inflammation. Therefore, to transform Mg-based implants into clinical practice, the present study evaluated the local effects of high-purity Mg screws (HP-Mg, 99.99 wt%) by comparing with clinically approved polylactic acid (PLA) screws in epiphyseal trabecular bone of rabbits. After implantation of screws at the rabbit distal femur, bone microstructural, histomorphometric and biomechanical properties were measured at various time points (weeks 4, 8 and 16) using micro-CT, histology and histomorphometry, micro-indentation and scanning electron microscope. HP-Mg screws promoted peri-implant bone ingrowth with higher bone mass (BV/TV at week 4: 0.189 ± 0.022 in PLA group versus 0.313 ± 0.053 in Mg group), higher biomechanical properties (hardness at week 4: 35.045 ± 1.000 HV in PLA group versus 51.975 ± 2.565 HV in Mg group), more mature osteocyte LCN architecture, accelerated bone remodeling process and alleviated immunoreactive score (IRS of Ram11 at week 4: 5.8 ± 0.712 in PLA group versus 3.75 ± 0.866 in Mg group) as compared to PLA screws. Furthermore, we conducted finite element analysis to validate the superiority of HP-Mg screws as orthopedic implants by demonstrating reduced stress concentration and uniform stress distribution around the bone tunnel, which led to lower risks of trabecular microfractures. In conclusion, HP-Mg screws demonstrated greater osteogenic bioactivity and limited inflammatory response compared to PLA screws in the epiphyseal trabecular bone of rabbits. Our findings have paved a promising way for the clinical application of Mg-based implants.
Collapse
Affiliation(s)
- Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Ying Luo
- School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou, Guangdong, 510000, China
| | - Weirong Li
- Dongguan Eontec Co., Ltd, Dongguan, Guangdong, 510730, China
| | - Fangfei Liu
- Dongguan Eontec Co., Ltd, Dongguan, Guangdong, 510730, China
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Jiali Wang
- School of Biomedical Engineering, Sun Yat-Sen University, Guangzhou, Guangdong, 510000, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, 999077, China
| |
Collapse
|
6
|
Xu H, Wang L, Zhu X, Zhang H, Chen H, Zhang H. Jintiange capsule ameliorates glucocorticoid-induced osteonecrosis of the femoral head in rats by regulating the activity and differentiation of BMSCs. J Tradit Complement Med 2024; 14:568-580. [PMID: 39262662 PMCID: PMC11384076 DOI: 10.1016/j.jtcme.2024.03.013] [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: 09/12/2023] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 09/13/2024] Open
Abstract
Background and aim A surplus of glucocorticoids (GC) is a main cause of non-traumatic osteonecrosis of the femoral head (ONFH), and Jintiange (JTG), as one of the traditional Chinese medicines (TCM), also plays an instrumental role in the alleviation of bone loss simultaneously. Therefore, JTG was thought to be able to reverse GC-induced ONFH (GC-ONFH) to a certain extent. Experimental procedure In vivo, the effect of JTG on trabeculae in the subchondral bone of the femoral head was investigated using micro-computed tomography (micro-CT), TdT-mediated dUTP nick end labeling (TUNEL) and histological staining; in vitro, proliferation, viability, apoptosis, and senescence of purified bone mesenchymal stem cells (BMSCs) were examined to demonstrate the direct impact of JTG on these cells. Meanwhile after using a series of interventions, the function of JTG on BMSC differentiation could be assessed by measuring of osteogenic and adipogenic markers at levels of protein and mRNA. Results Our final results demonstrated that with the involvement of Wnt/β-catenin pathway, JTG was able to significantly promote osteogenesis, restrain adipogenesis, delay senescence in BMSCs, reduce osteoclast number, weaken apoptosis, and enhance proliferation of osteocytes, all of which could mitigate the progression of subchondral osteonecrosis. Conclusion According to the results of experiments in vitro and vivo, JTG was deemed to relieve the early GC-ONFH using the prevention of destruction of subchondral bone, which was contributed to regulating the differentiation of BMSCs and the number of osteoclasts.
Collapse
Affiliation(s)
- Hui Xu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Lin Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xunpeng Zhu
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Haigang Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongwei Chen
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hui Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
7
|
He X, Li Y, Zou D, Zu H, Li W, Zheng Y. An overview of magnesium-based implants in orthopaedics and a prospect of its application in spine fusion. Bioact Mater 2024; 39:456-478. [PMID: 38873086 PMCID: PMC11170442 DOI: 10.1016/j.bioactmat.2024.04.026] [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: 12/29/2023] [Revised: 04/22/2024] [Accepted: 04/22/2024] [Indexed: 06/15/2024] Open
Abstract
Due to matching biomechanical properties and significant biological activity, Mg-based implants present great potential in orthopedic applications. In recent years, the biocompatibility and therapeutic effect of magnesium-based implants have been widely investigated in trauma repair. In contrast, the R&D work of Mg-based implants in spinal fusion is still limited. This review firstly introduced the general background for Mg-based implants. Secondly, the mechanical properties and degradation behaviors of Mg and its traditional and novel alloys were reviewed. Then, different surface modification techniques of Mg-based implants were described. Thirdly, this review comprehensively summarized the biological pathways of Mg degradation to promote bone formation in neuro-musculoskeletal circuit, angiogenesis with H-type vessel formation, osteogenesis with osteoblasts activation and chondrocyte ossification as an integrated system. Fourthly, this review followed the translation process of Mg-based implants via updating the preclinical studies in fracture fixation, sports trauma repair and reconstruction, and bone distraction for large bone defect. Furthermore, the pilot clinical studies were involved to demonstrate the reliable clinical safety and satisfactory bioactive effects of Mg-based implants in bone formation. Finally, this review introduced the background of spine fusion surgeryand the challenges of biological matching cage development. At last, this review prospected the translation potential of a hybrid Mg-PEEK spine fusion cage design.
Collapse
Affiliation(s)
- Xuan He
- Department of Orthopaedics, Peking University Third Hospital, No.49 North Huayuan Road, Haidian, Beijing, PR China
| | - Ye Li
- Department of Rehabilitation Science, The Hong Kong Polytechnic University, Hong Kong SAR, PR China
| | - Da Zou
- Department of Orthopaedics, Peking University Third Hospital, No.49 North Huayuan Road, Haidian, Beijing, PR China
| | - Haiyue Zu
- Department of Orthopaedics, The First Affiliated Hospital of Suchow University, PR China
| | - Weishi Li
- Department of Orthopaedics, Peking University Third Hospital, No.49 North Huayuan Road, Haidian, Beijing, PR China
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University, Comprehensive Scientific Research Building, Beijing, PR China
| |
Collapse
|
8
|
He L, Chhantyal K, Chen Z, Zhu R, Zhang L. The association of combined vitamin C and D deficiency with bone mineral density and vertebral fracture. J Orthop Surg Res 2024; 19:460. [PMID: 39095815 PMCID: PMC11297575 DOI: 10.1186/s13018-024-04953-z] [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: 05/10/2024] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
PURPOSE Both vitamin C and D deficiencies are extremely common in clinical practice, especially in elderly population. Unfortunately, the role of vitamin C deficiency in osteoporosis related consequences is often neglected. The aim of the present study is to analyse if combined vitamin C and D deficiency would have an association with bone mineral density (BMD) and osteoporotic vertebral fracture (OVF). METHODS Ninety-nine post-menopausal female patients admitted in the department of spine surgery of third affiliated hospital of Sun Yat-sen University were enrolled in the study. The participants were divided into four groups; vitamin D deficiency alone (comparator group), vitamin C deficiency alone and combined vitamin C and D deficiency as experimental group. The levels of vitamin C, vitamin D, calcium, phosphorous, BMD and condition of OVF were analysed. RESULTS There were statistically significant differences between the groups in terms of vitamin C and D levels. In terms of lumbar BMD, significant differences were observed between vitamin D deficiency alone and combined vitamin C and D deficiency. Only the combined vitamin C and D deficiency had a significant negative association with lumbar BMD and T-score. Similarly, combined vitamin C and D deficiency had a significant positive association with lumbar osteoporosis. None of the groups had any significant association with OVF. Combined vitamin C and D deficiency was found to be significantly associated with lower lumbar BMD and osteoporosis. CONCLUSION Combined vitamin C and D deficiency results in lower bone mineral density and higher risk of osteoporosis. We believe that existence of deficiencies of both vitamins could have a synergistic effect. Therefore, we recommend that vitamin C and D should be routinely measured in clinical practice.
Collapse
Affiliation(s)
- Lei He
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, 510630, China
- Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
- Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Kishor Chhantyal
- Emergency department, Doncaster and Bassetlaw Teaching Hospitals NHS Foundation Trust, Doncaster, England
| | - Zihao Chen
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, 510630, China
- Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
- Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Ruijue Zhu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, 510630, China
- Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
- Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China
| | - Liangming Zhang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, 510630, China.
- Guangdong Provincial Center for Quality Control of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China.
- Guangdong Provincial Center for Engineering and Technology Research of Minimally Invasive Spine Surgery, No. 600 Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
9
|
Zheng L, An Y, Tong W, Chen Z, Wang Y, Zhang H, Zhang S, Chen X, Liu W, Wang X, Xu J, Qin L. Canonical pathways for validating steroid-associated osteonecrosis in mice. Bone 2024; 183:117094. [PMID: 38582289 DOI: 10.1016/j.bone.2024.117094] [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: 01/06/2024] [Revised: 03/19/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
The present study aimed to establish and evaluate a preclinical model of steroid-associated osteonecrosis (SAON) in mice. Sixteen 24-week-old male C57BL/6 mice were used to establish SAON by two intraperitoneal injections of lipopolysaccharide (LPS), followed by three subcutaneous injections of methylprednisolone (MPS). Each injection was conducted on working day, with an interval of 24 h. Six cycles of injections were conducted. Additional twelve mice (age- and gender-matched) were used as normal controls. At 2 and 6 weeks after completing induction, bilateral femora and bilateral tibiae were collected for histological examination, micro-CT scanning, and bulk RNA sequencing. All mice were alive until sacrificed at the indicated time points. The typical SAON lesion was identified by histological evaluation at week 2 and week 6 with increased lacunae and TUNEL+ osteocytes. Micro-CT showed significant bone degeneration at week 6 in SAON model. Histology and histomorphometry showed significantly lower Runx2+ area, mineralizing surface (MS/BS), mineral apposition rate (MAR), bone formation rate (BFR/BS), type H vessels, Ki67+ (proliferating) cells, and higher marrow fat fraction, osteoclast number and TNFα+ areas in SAON group. Bulk RNA-seq revealed changed canonical signaling pathways regulating cell cycle, angiogenesis, osteogenesis, and osteoclastogenesis in the SAON group. The present study successfully established SAON in mice with a combination treatment of LPS and MPS, which could be considered a reliable and reproducible animal model to study the pathophysiology and molecular mechanism of early-stage SAON and to develop potential therapeutic approaches for the prevention and treatment of SAON.
Collapse
Affiliation(s)
- Lizhen Zheng
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong; Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong.
| | - Yuanming An
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong; The Sir Yue-Kong Pao Cancer Centre, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong; Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Hong Kong
| | - Ziyi Chen
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Yaofeng Wang
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences, Hong Kong
| | - Haozhi Zhang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Shi'an Zhang
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Xin Chen
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Weiyang Liu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Xinluan Wang
- Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Hong Kong; Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong; Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Hong Kong.
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong; Joint Laboratory of Chinese Academic of Science and Hong Kong for Biomaterials, The Chinese University of Hong Kong, Hong Kong; Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, PR China.
| |
Collapse
|
10
|
Zheng L, Zhao S, Li Y, Xu J, Yan W, Guo B, Xu J, Jiang L, Zhang Y, Wei H, Jiang Q. Engineered MgO nanoparticles for cartilage-bone synergistic therapy. SCIENCE ADVANCES 2024; 10:eadk6084. [PMID: 38457498 PMCID: PMC10923500 DOI: 10.1126/sciadv.adk6084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
The emerging therapeutic strategies for osteoarthritis (OA) are shifting toward comprehensive approaches that target periarticular tissues, involving both cartilage and subchondral bone. This shift drives the development of single-component therapeutics capable of acting on multiple tissues and cells. Magnesium, an element essential for maintaining skeletal health, shows promise in treating OA. However, the precise effects of magnesium on cartilage and subchondral bone are not yet clear. Here, we investigated the therapeutic effect of Mg2+ on OA, unveiling its protective effects on both cartilage and bone at the cellular and animal levels. The beneficial effect on the cartilage-bone interaction is primarily mediated by the PI3K/AKT pathway. In addition, we developed poly(lactic-co-glycolic acid) (PLGA) microspheres loaded with nano-magnesium oxide modified with stearic acid (SA), MgO&SA@PLGA, for intra-articular injection. These microspheres demonstrated remarkable efficacy in alleviating OA in rat models, highlighting their translational potential in clinical applications.
Collapse
Affiliation(s)
- Liming Zheng
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Sheng Zhao
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Yixuan Li
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics and Traumatology, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Wenjin Yan
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Baosheng Guo
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| | - Jianbin Xu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
| | - Lifeng Jiang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine; Orthopedics Research Institute of Zhejiang University; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, Zhejiang, 310000, PR China
| | - Yifeng Zhang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, PR China
| | - Hui Wei
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University; State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Qing Jiang
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, 321 Zhongshan Road; State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University; Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation; Institute of Medical 3D Printing, Nanjing University; Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing 210008, Jiangsu, PR China
| |
Collapse
|
11
|
Shi Z, Yang F, Hu Y, Pang Q, Shi L, Du T, Cao Y, Song B, Yu X, Cao Z, Ye Z, Liu C, Yu R, Chen X, Zhu Y, Pang Q. An oxidized dextran-composite self-healing coated magnesium scaffold reduces apoptosis to induce bone regeneration. Carbohydr Polym 2024; 327:121666. [PMID: 38171658 DOI: 10.1016/j.carbpol.2023.121666] [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: 09/29/2023] [Revised: 11/15/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024]
Abstract
Self-healing coatings have shown promise in controlling the degradation of scaffolds and addressing coating detachment issues. However, developing a self-healing coating for magnesium (Mg) possessing multiple biological functions in infectious environments remains a significant challenge. In this study, a self-healing coating was developed for magnesium scaffolds using oxidized dextran (OD), 3-aminopropyltriethoxysilane (APTES), and nano-hydroxyapatite (nHA) doped micro-arc oxidation (MHA), named OD-MHA/Mg. The results demonstrated that the OD-MHA coating effectively addresses coating detachment issues and controls the degradation of Mg in an infectious environment through self-healing mechanisms. Furthermore, the OD-MHA/Mg scaffold exhibits antibacterial, antioxidant, and anti-apoptotic properties, it also promotes bone repair by upregulating the expression of osteogenesis genes and proteins. The findings of this study indicate that the OD-MHA coated Mg scaffold possessing multiple biological functions presents a promising approach for addressing infectious bone defects. Additionally, the study showcases the potential of polysaccharides with multiple biological functions in facilitating tissue healing even in challenging environments.
Collapse
Affiliation(s)
- Zewen Shi
- Department of Orthopedics, Ningbo No. 2 Hospital, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315211, China; Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fang Yang
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yiwei Hu
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Qian Pang
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Lin Shi
- Department of Orthopedics, Ningbo No. 2 Hospital, Ningbo 315000, China
| | - Tianyu Du
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Yuhao Cao
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Baiyang Song
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Xueqiang Yu
- Department of Radiology, Ningbo No. 2 Hospital, Ningbo 315000, China
| | - Zhaoxun Cao
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhewei Ye
- Department of Orthopaedics, Wuhan Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chen Liu
- Ningbo Branch of Chinese Academy of Ordnance Science, Ningbo 315100, China
| | - Rongyao Yu
- Department of Orthopedics, Ningbo No. 2 Hospital, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315211, China
| | - Xianjun Chen
- Department of Orthopedics, Ningbo No. 2 Hospital, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315211, China.
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo 315211, China.
| | - Qingjiang Pang
- Department of Orthopedics, Ningbo No. 2 Hospital, Ningbo 315000, China; Health Science Center, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
12
|
Cheng W, Yang H, Xiao L, Yang G, Lu Q, Kaplan DL. Nanosized Silk-Magnesium Complexes for Promotion of Angiogenic and Osteogenic Activities. ACS APPLIED MATERIALS & INTERFACES 2024; 16:9880-9889. [PMID: 38359078 DOI: 10.1021/acsami.3c18195] [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: 02/17/2024]
Abstract
Injectable hydrogels with osteogenic and angiogenetic properties are of interest in bone tissue engineering. Since the bioactivity of ions is concentration-dependent, nanosized silk-magnesium (Mg) complexes were previously developed and assembled into hydrogels with angiogenic capabilities but failed to control both osteogenic and angiogenetic activities effectively. Here, nanosized silk particles with different sizes were obtained by using ultrasonic treatment to control silk-Mg coordination and particle formation, resulting in silk-Mg hydrogels with different types of bioactivity. Fourier transform infrared and X-ray diffraction results revealed that different coordination intensities were present in the different complexes as a basis for the differences in activities. Slow Mg ion release was controlled by these nanosized silk-Mg complexes through degradation. With the same amount of Mg ions, the different silk-Mg complexes exhibited different angiogenic and osteogenic capacities. Complexes with both angiogenic and osteogenic capacities were developed by optimizing the sizes of the silk particles, resulting in faster and improved quality of bone formed in vivo than complexes with the same composition of silk and Mg but only angiogenic or osteogenic capacities. The biological selectivity of silk-Mg complexes should facilitate applications in tissue regeneration.
Collapse
Affiliation(s)
- Weinan Cheng
- Department of Sports Medicine, Shanghai Jiao Tong University Affiliated Shanghai Sixth People's Hospital, Shanghai 200233, People's Republic of China
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China
| | - Huaxiang Yang
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China
- National Engineering Laboratory for Modern Silk & Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou 215123, People's Republic of China
| | - Liying Xiao
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China
| | - Gongwen Yang
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China
| | - Qiang Lu
- State Key Laboratory of Radiation Medicine and Radiation Protection, Institutes for Translational Medicine, Soochow University, Suzhou 215123, People's Republic of China
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
13
|
Sadeghi O, Khademi Z, Saneei P, Hassanzadeh-Keshteli A, Daghaghzadeh H, Tavakkoli H, Adibi P, Esmaillzadeh A. Dietary Magnesium Intake Is Inversely Associated With Ulcerative Colitis: A Case-Control Study. CROHN'S & COLITIS 360 2024; 6:otae009. [PMID: 38464345 PMCID: PMC10923208 DOI: 10.1093/crocol/otae009] [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: 08/19/2023] [Indexed: 03/12/2024] Open
Abstract
Background Ulcerative colitis (UC) causes long-lasting inflammation and ulcers in the gut. Limited observational data are available linking dietary magnesium intake and UC. In the present study, we aimed to investigate the association between dietary magnesium intake and UC in adults. Methods The current population-based case-control study was performed on 109 UC patients and 218 age (±2 years) and sex-matched controls. The diagnosis of UC was made according to the standard criteria by a gastroenterology specialist. Dietary intakes were assessed using a validated self-administrated 106-item dish-based Food Frequency Questionnaire (FFQ). We also used a pretested questionnaire to collect data on potential confounders. Results Individuals in the top tertile of magnesium intake were less likely to have UC compared with those in the bottom tertile. A significant inverse relationship was found between dietary magnesium intake and UC (odds ratio [OR]: 0.32, 95% confidence interval [CI]: 0.18-0.59) in the crude model. This relationship was also observed when we took several potential confounding into account (OR: 0.30, 95% CI: 0.14-0.68). Conclusions Adherence to a magnesium-rich diet may have a role in preventing UC. However, further studies are needed to confirm our findings.
Collapse
Affiliation(s)
- Omid Sadeghi
- Nutrition and Food Security Research Center, Department of Community Nutrition, Student Research Committee, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zeinab Khademi
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Parvane Saneei
- Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ammar Hassanzadeh-Keshteli
- Department of Medicine, University of Alberta, Edmonton, Canada
- Integrative Functional Gastroenterology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamed Daghaghzadeh
- Integrative Functional Gastroenterology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamid Tavakkoli
- Integrative Functional Gastroenterology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Peyman Adibi
- Integrative Functional Gastroenterology Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ahmad Esmaillzadeh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular–Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Gong C, Yang J, Zhang X, Wei Z, Wang X, Huang X, Yu L, Guo W. Functionalized Magnesium Phosphate Cement Induces In Situ Vascularized Bone Regeneration via Surface Lyophilization of Chondroitin Sulfate. Biomedicines 2023; 12:74. [PMID: 38255182 PMCID: PMC10812989 DOI: 10.3390/biomedicines12010074] [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: 09/27/2023] [Revised: 12/15/2023] [Accepted: 12/24/2023] [Indexed: 01/24/2024] Open
Abstract
Bone defect repair poses significant challenges in orthopedics, thereby increasing the demand for bone substitutes. Magnesium phosphate cements (MPCs) are widely used for bone defect repair because of their excellent mechanical properties and biodegradability. However, high crystallinity and uncontrolled magnesium ion (Mg2+) release limit the surface bioactivity of MPCs in bone regeneration. Here, we fabricate chondroitin sulfate (CS) as a surface coating via the lyophilization method, namely CMPC. We find that the CS coating is uniformly distributed and improves the mechanical properties of MPC through anionic electrostatic adsorption, while mediating degradation-related controlled ion release of Mg2+. Using a combination of in vitro and in vivo analyses, we show that the CS coating maintained cytocompatibility while increasing the cell adhesion area of MC3T3-E1s. Furthermore, we display accelerated osteogenesis and angiogenesis of CMPC, which are related to appropriate ion concentration of Mg2+. Our findings reveal that the preparation of a lyophilized CS coating is an effective method to promote surface bioactivity and mediate Mg2+ concentration dependent osteogenesis and angiogenesis, which have great potential in bone regeneration.
Collapse
Affiliation(s)
- Changtian Gong
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (C.G.); (J.Y.); (X.Z.); (Z.W.); (X.W.); (X.H.); (L.Y.)
- Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jian Yang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (C.G.); (J.Y.); (X.Z.); (Z.W.); (X.W.); (X.H.); (L.Y.)
| | - Xiping Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (C.G.); (J.Y.); (X.Z.); (Z.W.); (X.W.); (X.H.); (L.Y.)
| | - Zhun Wei
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (C.G.); (J.Y.); (X.Z.); (Z.W.); (X.W.); (X.H.); (L.Y.)
| | - Xingyu Wang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (C.G.); (J.Y.); (X.Z.); (Z.W.); (X.W.); (X.H.); (L.Y.)
| | - Xinghan Huang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (C.G.); (J.Y.); (X.Z.); (Z.W.); (X.W.); (X.H.); (L.Y.)
| | - Ling Yu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (C.G.); (J.Y.); (X.Z.); (Z.W.); (X.W.); (X.H.); (L.Y.)
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan 430060, China; (C.G.); (J.Y.); (X.Z.); (Z.W.); (X.W.); (X.H.); (L.Y.)
| |
Collapse
|
15
|
Park Y, Sato T, Lee J. Functional and analytical recapitulation of osteoclast biology on demineralized bone paper. Nat Commun 2023; 14:8092. [PMID: 38062034 PMCID: PMC10703810 DOI: 10.1038/s41467-023-44000-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Osteoclasts are the primary target for osteoporosis drug development. Recent animal studies revealed the crucial roles of osteoblasts in regulating osteoclastogenesis and the longer lifespans of osteoclasts than previously thought with fission and recycling. However, existing culture platforms are limited to replicating these newly identified cellular processes. We report a demineralized bone paper (DBP)-based osteoblast culture and osteoclast assay platform that replicates osteoclast fusion, fission, resorption, and apoptosis with high fidelity and analytical power. An osteoid-inspired DBP supports rapid and structural mineral deposition by osteoblasts. Coculture osteoblasts and bone marrow monocytes under biochemical stimulation recapitulate osteoclast differentiation and function. The DBP-based bone model allows longitudinal quantitative fluorescent monitoring of osteoclast responses to bisphosphonate drug, substantiating significantly reducing their number and lifespan. Finally, we demonstrate the feasibility of humanizing the bone model. The DBP-based osteo assay platforms are expected to advance bone remodeling-targeting drug development with improved prediction of clinical outcomes.
Collapse
Affiliation(s)
- Yongkuk Park
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Tadatoshi Sato
- Department of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Jungwoo Lee
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01003, USA.
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
16
|
Lin ZY, Liang YY, Wang R, Hu B, He WJ, Li JK, Ding ZA, Lin ZY, Zhang S. Dietary magnesium is able to influence the relationship between vitamin C and estimated glomerular filtration rate: A cross-sectional study. Food Sci Nutr 2023; 11:4773-4780. [PMID: 37576036 PMCID: PMC10420853 DOI: 10.1002/fsn3.3456] [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: 01/07/2023] [Revised: 05/08/2023] [Accepted: 05/13/2023] [Indexed: 08/15/2023] Open
Abstract
The estimated glomerular filtration rate (eGFR) is a comprehensive index that is widely used to assess renal function. Although studies have confirmed a correlation between eGFR and dietary vitamin C, the impact of varying levels of vitamin C on eGFR remains unclear. Additionally, the interaction between dietary magnesium intake and vitamin C concentration on eGFR is not well understood. As such, the objective of this study was to investigate the relationship between dietary magnesium intake and vitamin C in relation to eGFR. This study analyzed the data of consecutive NHANES from 2005 to 2018. We included 17,633 participants aged 20 or older and used multiple linear regression analysis to evaluate the relationship between dietary vitamin C and eGFR. Dietary Mg intake from experimental data was dichotomized into a low dietary Mg intake group (≤254 mg/day) and a normal dietary Mg intake group (>254 mg/day). To evaluate the impact of dietary magnesium intake on eGFR, a multivariable linear regression was conducted utilizing an interaction test between dietary vitamin C and eGFR. We discovered a positive association between dietary vitamin C content and eGFR. The relationship between dietary vitamin C levels and eGFR differed between individuals with low Mg intake and those with normal Mg intake (β: 2.96 95% CI:1.63 ~ 4.29 vs. β: 1.05 95% CI: -0.15 to 2.25), and the positive association of high dietary vitamin C content with eGFR was stronger in the low Mg intake group. Furthermore, we observed that dietary magnesium intake significantly altered the positive association between dietary vitamin C and eGFR (interaction value of 0.020). Our experimental study revealed that the interaction between dietary magnesium and dietary vitamin C can significantly impact eGFR. This finding carries significant implications for the treatment of diseases resulting from abnormal eGFR, as well as the selection of clinically relevant drugs.
Collapse
Affiliation(s)
- Zheng-Yang Lin
- Department of Clinical Medicine The Second Clinical School of Guangzhou Medical University Guangzhou China
| | - Yong-Yi Liang
- Department of Preventive Medicine School of Public Health, Guangzhou Medical University Guangzhou China
| | - Ru Wang
- Department of Medical Imaging The Second Clinical School of Guangzhou Medical University Guangzhou China
| | - Biao Hu
- Department of Clinical Medicine The Second Clinical School of Guangzhou Medical University Guangzhou China
| | - Wen-Ju He
- Department of Clinical Medicine The Second Clinical School of Guangzhou Medical University Guangzhou China
| | - Jun-Kui Li
- Department of Clinical Medicine The Second Clinical School of Guangzhou Medical University Guangzhou China
| | - Zi-Ang Ding
- Department of Clinical Medicine The First Clinical School of Guangzhou Medical University Guangzhou China
| | - Zhuo-Yuan Lin
- Department of Urology The Second Affiliated Hospital of Guangzhou Medical University Guangzhou Medical University Guangzhou China
| | - Shi Zhang
- Department of Gastrointestinal Surgery The Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
| |
Collapse
|
17
|
Wu N, Gao H, Wang X, Pei X. Surface Modification of Titanium Implants by Metal Ions and Nanoparticles for Biomedical Application. ACS Biomater Sci Eng 2023; 9:2970-2990. [PMID: 37184344 DOI: 10.1021/acsbiomaterials.2c00722] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Implant surface modification can improve osseointegration and reduce peri-implant inflammation. Implant surfaces are modified with metals because of their excellent mechanical properties and significant functions. Metal surface modification is divided into metal ions and nanoparticle surface modification. These two methods function by adding a finishing metal to the surface of the implant, and both play a role in promoting osteogenic, angiogenic, and antibacterial properties. Based on this, the nanostructural surface changes confer stronger antibacterial and cellular affinity to the implant surface. The current paper reviews the forms, mechanisms, and applications of nanoparticles and metal ion modifications to provide a foundation for the surface modification of implants.
Collapse
Affiliation(s)
- Nan Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongyu Gao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xu Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
18
|
Liu Z, Tang Q, Liu RT, Yu MZ, Peng H, Zhang CQ, Zhu ZZ, Wei XJ. Laponite intercalated biomimetic multilayer coating prevents glucocorticoids induced orthopedic implant failure. Bioact Mater 2023; 22:60-73. [PMID: 36203962 PMCID: PMC9519439 DOI: 10.1016/j.bioactmat.2022.09.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/18/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022] Open
|
19
|
Cai W, Wakamatsu K, Zucca FA, Wang Q, Yang K, Mohamadzadehonarvar N, Srivastava P, Tanaka H, Holly G, Casella L, Ito S, Zecca L, Chen X. DOPA pheomelanin is increased in nigral neuromelanin of Parkinson's disease. Prog Neurobiol 2023; 223:102414. [PMID: 36746222 DOI: 10.1016/j.pneurobio.2023.102414] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 01/29/2023] [Accepted: 02/02/2023] [Indexed: 02/05/2023]
Abstract
Neuromelanin (NM) in dopaminergic neurons of human substantia nigra (SN) has a melanic component that consists of pheomelanin and eumelanin moieties and has been proposed as a key factor contributing to dopaminergic neuron vulnerability in Parkinson's disease (PD). While eumelanin is considered as an antioxidant, pheomelanin and related oxidative stress are associated with compromised drug and metal ion binding and melanoma risk. Using postmortem SN from patients with PD or Alzheimer's disease (AD) and unaffected controls, we identified increased L-3,4-dihydroxyphenylalanine (DOPA) pheomelanin and increased ratios of dopamine (DA) pheomelanin markers to DA in PD SN compared to controls. Eumelanins derived from both DOPA and DA were reduced in PD group. In addition, we report an increase in DOPA pheomelanin relative to DA pheomelanin in PD SN. In AD SN, we observed unaltered melanin markers despite reduced DOPA compared to controls. Furthermore, synthetic DOPA pheomelanin induced neuronal cell death in vitro while synthetic DOPA eumelanin showed no significant effect on cell viability. Our findings provide insights into the different roles of pheomelanin and eumelanin in PD pathophysiology. We anticipate our study will lead to further investigations on pheomelanin and eumelanin individually as biomarkers and possibly therapeutic targets for PD.
Collapse
Affiliation(s)
- Waijiao Cai
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Institutes of Integrative Medicine, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Shanghai, China
| | - Kazumasa Wakamatsu
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, Japan
| | - Fabio A Zucca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Qing Wang
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, USA
| | - Kai Yang
- Institutes of Integrative Medicine, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Shanghai, China
| | - Niyaz Mohamadzadehonarvar
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, USA
| | - Pranay Srivastava
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, USA
| | - Hitomi Tanaka
- Department of Medical Technology, School of Health Sciences, Gifu University of Medical Science, Seki, Japan
| | - Gabriel Holly
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Luigi Casella
- Department of Chemistry, University of Pavia, Pavia, Italy
| | - Shosuke Ito
- Institute for Melanin Chemistry, Fujita Health University, Toyoake, Japan
| | - Luigi Zecca
- Institute of Biomedical Technologies, National Research Council of Italy, Segrate, Milan, Italy
| | - Xiqun Chen
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, USA; Aligning Science Across Parkinson's Collaborative Research Network, Chevy Chase, USA.
| |
Collapse
|
20
|
Bian Y, Hu T, Lv Z, Xu Y, Wang Y, Wang H, Zhu W, Feng B, Liang R, Tan C, Weng X. Bone tissue engineering for treating osteonecrosis of the femoral head. EXPLORATION (BEIJING, CHINA) 2023; 3:20210105. [PMID: 37324030 PMCID: PMC10190954 DOI: 10.1002/exp.20210105] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 05/12/2022] [Indexed: 06/16/2023]
Abstract
Osteonecrosis of the femoral head (ONFH) is a devastating and complicated disease with an unclear etiology. Femoral head-preserving surgeries have been devoted to delaying and hindering the collapse of the femoral head since their introduction in the last century. However, the isolated femoral head-preserving surgeries cannot prevent the natural progression of ONFH, and the combination of autogenous or allogeneic bone grafting often leads to many undesired complications. To tackle this dilemma, bone tissue engineering has been widely developed to compensate for the deficiencies of these surgeries. During the last decades, great progress has been made in ingenious bone tissue engineering for ONFH treatment. Herein, we comprehensively summarize the state-of-the-art progress made in bone tissue engineering for ONFH treatment. The definition, classification, etiology, diagnosis, and current treatments of ONFH are first described. Then, the recent progress in the development of various bone-repairing biomaterials, including bioceramics, natural polymers, synthetic polymers, and metals, for treating ONFH is presented. Thereafter, regenerative therapies for ONFH treatment are also discussed. Finally, we give some personal insights on the current challenges of these therapeutic strategies in the clinic and the future development of bone tissue engineering for ONFH treatment.
Collapse
Affiliation(s)
- Yixin Bian
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Tingting Hu
- State Key Laboratory of Chemical Resource EngineeringBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijingChina
| | - Zehui Lv
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Yiming Xu
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Yingjie Wang
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Han Wang
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Wei Zhu
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Bin Feng
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| | - Ruizheng Liang
- State Key Laboratory of Chemical Resource EngineeringBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijingChina
| | - Chaoliang Tan
- Department of ChemistryCity University of Hong KongKowloonHong Kong SARChina
| | - Xisheng Weng
- Department of Orthopedic SurgeryState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Science and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
21
|
Feng Z, Jin M, Liang J, Kang J, Yang H, Guo S, Sun X. Insight into the effect of biomaterials on osteogenic differentiation of mesenchymal stem cells: A review from a mitochondrial perspective. Acta Biomater 2023; 164:1-14. [PMID: 36972808 DOI: 10.1016/j.actbio.2023.03.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/02/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023]
Abstract
Bone damage may be triggered by a variety of factors, and the damaged area often requires a bone graft. Bone tissue engineering can serve as an alternative strategy for repairing large bone defects. Mesenchymal stem cells (MSCs), the progenitor cells of connective tissue, have become an important tool for tissue engineering due to their ability to differentiate into a variety of cell types. The precise regulation of the growth and differentiation of the stem cells used for bone regeneration significantly affects the efficiency of this type of tissue engineering. During the process of osteogenic induction, the dynamics and function of localized mitochondria are altered. These changes may also alter the microenvironment of the therapeutic stem cells and result in mitochondria transfer. Mitochondrial regulation not only affects the induction/rate of differentiation, but also influences its direction, determining the final identity of the differentiated cell. To date, bone tissue engineering research has mainly focused on the influence of biomaterials on phenotype and nuclear genotype, with few studies investigating the role of mitochondria. In this review, we provide a comprehensive summary of researches into the role of mitochondria in MSCs differentiation and critical analysis regarding smart biomaterials that are able to "programme" mitochondria modulation was proposed. STATEMENT OF SIGNIFICANCE: : • This review proposed the precise regulation of the growth and differentiation of the stem cells used to seed bone regeneration. • This review addressed the dynamics and function of localized mitochondria during the process of osteogenic induction and the effect of mitochondria on the microenvironment of stem cells. • This review summarized biomaterials which affect the induction/rate of differentiation, but also influences its direction, determining the final identity of the differentiated cell through the regulation of mitochondria.
Collapse
Affiliation(s)
- Ziyi Feng
- Department of Plastic Surgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110002 Liaoning Province, China
| | - Meiqi Jin
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang, 110122, Liaoning Province, China
| | - Junzhi Liang
- Center of Reproductive Medicine, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping, Shenyang, 110004 Liaoning Province, China
| | - Junning Kang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping, Shenyang, 110004 Liaoning Province, China
| | - Huazhe Yang
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang, 110122, Liaoning Province, China.
| | - Shu Guo
- Department of Plastic Surgery, The First Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110002 Liaoning Province, China.
| | - Xiaoting Sun
- School of Forensic Medicine, China Medical University, No.77, Puhe Road, Shenyang, 110122, Liaoning Province, China.
| |
Collapse
|
22
|
Magnesium surface-activated 3D printed porous PEEK scaffolds for in vivo osseointegration by promoting angiogenesis and osteogenesis. Bioact Mater 2023; 20:16-28. [PMID: 35633876 PMCID: PMC9123089 DOI: 10.1016/j.bioactmat.2022.05.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/01/2022] [Accepted: 05/07/2022] [Indexed: 12/14/2022] Open
Abstract
Polyetheretherketone (PEEK) has been an alternative material for titanium in bone defect repair, but its clinical application is limited by its poor osseointegration. In this study, a porous structural design and activated surface modification were used to enhance the osseointegration capacity of PEEK materials. Porous PEEK scaffolds were manufactured via fused deposition modeling and a polydopamine (PDA) coating chelated with magnesium ions (Mg2+) was utilized on the surface. After surface modification, the hydrophilicity of PEEK scaffolds was significantly enhanced, and bioactive Mg2+ could be released. In vitro results showed that the activated surface could promote cell proliferation and adhesion and contribute to osteoblast differentiation and mineralization; the released Mg2+ promoted angiogenesis and might contribute to the formation of osteogenic H-type vessels. Furthermore, porous PEEK scaffolds were implanted in rabbit femoral condyles for in vivo evaluation of osseointegration. The results showed that the customized three-dimensional porous structure facilitated vascular ingrowth and bone ingrowth within the PEEK scaffolds. The PDA coating enhanced the interfacial osseointegration of porous PEEK scaffolds and the released Mg2+ accelerated early bone ingrowth by promoting early angiogenesis during the coating degradation process. This study provides an efficient solution for enhancing the osseointegration of PEEK materials, which has high potential for translational clinical applications. PEEK materials were modified by structural porosification and surface activation simultaneously. Bioactive Mg2+ released by surface-activated porous PEEK scaffolds enhanced angiogenesis and osteogenesis. Customized three-dimensional porous structure of PEEK scaffolds facilitated vascular ingrowth and bone ingrowth. Surface-activated porous PEEK scaffolds achieved satisfactory osseointegration in vivo.
Collapse
|
23
|
Zan R, Shen S, Huang Y, Yu H, Liu Y, Yang S, Zheng B, Gong Z, Wang W, Zhang X, Suo T, Liu H. Research hotspots and trends of biodegradable magnesium and its alloys. SMART MATERIALS IN MEDICINE 2023; 4:468-479. [DOI: 10.1016/j.smaim.2023.01.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
24
|
Hou Y, Zhang R, Cheng H, Wang Y, Zhang Q, Zhang L, Wang L, Li R, Wu X, Li B. Mg2+-doped carbon dots synthesized based on Lycium ruthenicum in cell imaging and promoting osteogenic differentiation in vitro. Colloids Surf A Physicochem Eng Asp 2023. [DOI: 10.1016/j.colsurfa.2022.130264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
25
|
Gholizadeh‐Moghaddam M, Ghasemi‐Tehrani H, Askari G, Jaripur M, Clark CCT, Rouhani MH. Effect of magnesium supplementation in improving hyperandrogenism, hirsutism, and sleep quality in women with polycystic ovary syndrome: A randomized, placebo-controlled clinical trial. Health Sci Rep 2023; 6:e1013. [PMID: 36620514 PMCID: PMC9798963 DOI: 10.1002/hsr2.1013] [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: 08/17/2022] [Revised: 10/29/2022] [Accepted: 12/12/2022] [Indexed: 12/31/2022] Open
Abstract
Background and Aims Polycystic ovary syndrome (PCOS) is one of the common endocrine disorders in women, which causes numerous symptoms in women. The relationship of many micronutrients with this syndrome has been investigated. This study was conducted to examine the effects of magnesium supplementation on hyperandrogenism, hirsutism, and sleep quality in women with PCOS. Methods In this parallel randomized clinical trial, 64 women with PCOS were randomly assigned to the magnesium group (n = 32) or placebo group (n = 32) for 10 weeks. Patients in the magnesium group received one 250 mg magnesium oxide tablet, per day. Hyperandrogenism, hirsutism, and sleep quality were measured at the beginning and end of the study. This randomized clinical trial was registered at https://www.IRCT.ir (IRCT20130903014551N8). Results Magnesium supplementation had no significant effect on hyperandrogenism (p = 0.51 for dehydroepiandrosterone sulfates, p = 0.27 for testosterone), hirsutism (p = 0.23), and sleep quality (p = 0.85) compared with placebo. Conclusions The present study showed that a single dose of magnesium supplementation elicited no beneficial effects on the mentioned symptoms in polycystic women. It is possible that the positive effects of magnesium observed in the former studies were due to the synergistic effects of other vitamins or minerals. More studies are needed in this area.
Collapse
Affiliation(s)
- Mahsa Gholizadeh‐Moghaddam
- Nutrition and Food Security Research Center, Department of Community NutritionIsfahan University of Medical SciencesIsfahanIran
| | - Hatav Ghasemi‐Tehrani
- Department of Obstetrics and GynecologyIsfahan University of Medical SciencesIsfahanIran
| | - Gholamreza Askari
- Nutrition and Food Security Research Center, Department of Community NutritionIsfahan University of Medical SciencesIsfahanIran
| | - Mahsima Jaripur
- Nutrition and Food Security Research Center, Department of Community NutritionIsfahan University of Medical SciencesIsfahanIran
| | | | - Mohammad Hossein Rouhani
- Nutrition and Food Security Research Center, Department of Community NutritionIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
26
|
Wang Z, Liu B, Yin B, Zheng Y, Tian Y, Wen P. Comprehensive review of additively manufactured biodegradable magnesium implants for repairing bone defects from biomechanical and biodegradable perspectives. Front Chem 2022; 10:1066103. [PMID: 36523749 PMCID: PMC9745192 DOI: 10.3389/fchem.2022.1066103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 11/21/2022] [Indexed: 10/21/2023] Open
Abstract
Bone defect repair is a complicated clinical problem, particularly when the defect is relatively large and the bone is unable to repair itself. Magnesium and its alloys have been introduced as versatile biomaterials to repair bone defects because of their excellent biocompatibility, osteoconductivity, bone-mimicking biomechanical features, and non-toxic and biodegradable properties. Therefore, magnesium alloys have become a popular research topic in the field of implants to treat critical bone defects. This review explores the popular Mg alloy research topics in the field of bone defects. Bibliometric analyses demonstrate that the degradation control and mechanical properties of Mg alloys are the main research focus for the treatment of bone defects. Furthermore, the additive manufacturing (AM) of Mg alloys is a promising approach for treating bone defects using implants with customized structures and functions. This work reviews the state of research on AM-Mg alloys and the current challenges in the field, mainly from the two aspects of controlling the degradation rate and the fabrication of excellent mechanical properties. First, the advantages, current progress, and challenges of the AM of Mg alloys for further application are discussed. The main mechanisms that lead to the rapid degradation of AM-Mg are then highlighted. Next, the typical methods and processing parameters of laser powder bed fusion fabrication on the degradation characteristics of Mg alloys are reviewed. The following section discusses how the above factors affect the mechanical properties of AM-Mg and the recent research progress. Finally, the current status of research on AM-Mg for bone defects is summarized, and some research directions for AM-Mg to drive the application of clinical orthopedic implants are suggested.
Collapse
Affiliation(s)
- Zhengguang Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Bingchuan Liu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Bangzhao Yin
- Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, China
| | - Yun Tian
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Peking University Third Hospital, Beijing, China
| | - Peng Wen
- Department of Mechanical Engineering, Tsinghua University, Beijing, China
| |
Collapse
|
27
|
Li X, Dai B, Guo J, Zhu Y, Xu J, Xu S, Yao Z, Chang L, Li Y, He X, Chow DHK, Zhang S, Yao H, Tong W, Ngai T, Qin L. Biosynthesized Bandages Carrying Magnesium Oxide Nanoparticles Induce Cortical Bone Formation by Modulating Endogenous Periosteal Cells. ACS NANO 2022; 16:18071-18089. [PMID: 36108267 DOI: 10.1021/acsnano.2c04747] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Bone grafting is frequently conducted to treat bone defects caused by trauma and tumor removal, yet with significant medical and socioeconomic burdens. Space-occupying bone substitutes remain challenging in the control of osteointegration, and meanwhile activation of endogenous periosteal cells by using non-space-occupying implants to promote new bone formation becomes another therapeutic strategy. Here, we fabricated a magnesium-based artificial bandage with optimal micropatterns for activating periosteum-associated biomineralization. Collagen was self-assembled on the surface of magnesium oxide nanoparticles embedded electrospun fibrous membranes as a hierarchical bandage structure to facilitate the integration with periosteum in situ. After the implantation on the surface of cortical bone in vivo, magnesium ions were released to generate a pro-osteogenic immune microenvironment by activating the endogenous periosteal macrophages into M2 phenotype and, meanwhile, promote blood vessel formation and neurite outgrowth. In a cortical bone defect model, magnesium-based artificial bandage guided the surrounding newly formed bone tissue to cover the defected area. Taken together, our study suggests that the strategy of stimulating bone formation can be achieved with magnesium delivery to periosteum in situ and the proposed periosteal bandages act as a bioactive media for accelerating bone healing.
Collapse
Affiliation(s)
- Xu Li
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Bingyang Dai
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Yuwei Zhu
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Shunxiang Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Zhi Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Liang Chang
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Ye Li
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Xuan He
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Dick Ho Kiu Chow
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Shian Zhang
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Hao Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| | - To Ngai
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong999077, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong999077, China
- Innovative Orthopedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong999077, China
| |
Collapse
|
28
|
Zhao M, Chen G, Zhang S, Chen B, Wu Z, Zhang C. A bioactive poly(ether-ether-ketone) nanocomposite scaffold regulates osteoblast/osteoclast activity for the regeneration of osteoporotic bone. J Mater Chem B 2022; 10:8719-8732. [PMID: 36239238 DOI: 10.1039/d2tb01387h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Due to the lower regeneration capacity of the osteoporotic bone, the treatment of osteoporotic defects is extremely challenging in clinics. In this study, strontium-doped bioactive glass nanoparticles loaded with sodium alendronate (ALN), namely A-SrBG, were incorporated into the poly(ether-ether-ketone) matrix to fabricate a bioactive composite scaffold (ASP), which was expected to both inhibit bone resorption and promote bone regeneration. The results showed that such a composite scaffold with interconnected macropores (200-400 μm) could release Ca2+, Sr2+, and ALN in vitro. The proliferation, alkaline phosphatase (ALP) activity, expression of osteogenesis-related genes, and formation of calcified nodules of rat bone marrow stromal cells (rBMSCs) were clearly evidenced, and the reduction in the proliferation, tartrate-resistant acid phosphatase (TRAP) activity, cell fusion, and expression of osteoclastogenesis-related genes of osteoclasts was observed as well. In the presence of the ASP scaffold, enhanced osteogenesis along with inhibiting osteoclastogenesis was observed by modulating the osteoprotegerin (OPG)/receptor activator for nuclear factor κB ligand (RANKL) ratio. The efficacy of the composite scaffold in the regeneration of osteoporotic critical-sized cranial defect in a rat model was evaluated. Therefore, the bioactive composite scaffold with excellent biocompatibility and osteogenic potential could be a promising material for the repair of osteoporotic bone defects.
Collapse
Affiliation(s)
- Mengen Zhao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| | - Guo Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Shixiong Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| | - Bin Chen
- Division of Orthopaedics and Traumatology, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Zhaoying Wu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| | - Chao Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, 518107, China.
| |
Collapse
|
29
|
Wang Z, Wang W, Zhang X, Cao F, Zhang T, Bhakta Pokharel D, Chen D, Li J, Yang J, Xiao C, Ren Y, Qin G, Zhao D. Modulation of Osteogenesis and Angiogenesis Activities Based on Ionic Release from Zn-Mg Alloys. MATERIALS (BASEL, SWITZERLAND) 2022; 15:ma15207117. [PMID: 36295204 PMCID: PMC9608845 DOI: 10.3390/ma15207117] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 05/12/2023]
Abstract
The enhancement of osteogenesis and angiogenesis remains a great challenge for the successful regeneration of engineered tissue. Biodegradable Mg and Zn alloys have received increasing interest as potential biodegradable metallic materials, partially due to the biological functions of Mg2+ and Zn2+ with regard to osteogenesis and angiogenesis, respectively. In the present study, novel biodegradable Zn-xMg (x = 0.2, 0.5, 1.0 wt.%) alloys were designed and fabricated, and the effects of adding different amounts of Mg to the Zn matrix were investigated. The osteogenesis and angiogenesis beneficial effects of Zn2+ and Mg2+ release during the biodegradation were characterized, demonstrating coordination with the bone regeneration process in a dose-dependent manner. The results show that increased Mg content leads to a higher amount of released Mg2+ while decreasing the Zn2+ concentration in the extract. The osteogenesis of pre-osteoblasts was promoted in Zn-0.5Mg and Zn-1Mg due to the higher concentration of Mg2+. Moreover, pure Zn extract presented the highest activity in angiogenesis, owing to the highest concentration of Zn2+ release (6.415 μg/mL); the proliferation of osteoblast cells was, however, inhibited under such a high Zn2+ concentration. Although the concentration of Zn ion was decreased in Zn-0.5Mg and Zn-1Mg compared with pure Zn, the angiogenesis was not influenced when the concentration of Mg in the extract was sufficiently increased. Hence, Mg2+ and Zn2+ in Zn-Mg alloys show a dual modulation effect. The Zn-0.5Mg alloy was indicated to be a promising implant candidate due to demonstrating the appropriate activity in regulating osteogenesis and angiogenesis. The present work evaluates the effect of the Mg content in Zn-based alloys on biological activities, and the results provide guidance regarding the Zn-Mg composition in designs for orthopedic application.
Collapse
Affiliation(s)
- Ziming Wang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Weidan Wang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Xiuzhi Zhang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
- Correspondence: (X.Z.); (D.Z.)
| | - Fang Cao
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Tianwei Zhang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
- School of Mechanical Engineering, Dalian Jiaotong University, Dalian 116028, China
| | - Durga Bhakta Pokharel
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang 110016, China
| | - Di Chen
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Junlei Li
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Jiahui Yang
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Chi Xiao
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
| | - Yuping Ren
- School of Materials Science and Engineering, Northeastern University, Shenyang 110819, China
| | - Gaowu Qin
- School of Materials Science and Engineering, Northeastern University, Shenyang 110819, China
| | - Dewei Zhao
- Department of Orthopaedics, Affiliated Zhongshan Hospital of Dalian University, Dalian 116001, China
- Correspondence: (X.Z.); (D.Z.)
| |
Collapse
|
30
|
Yuan Z, Wan Z, Gao C, Wang Y, Huang J, Cai Q. Controlled magnesium ion delivery system for in situ bone tissue engineering. J Control Release 2022; 350:360-376. [PMID: 36002052 DOI: 10.1016/j.jconrel.2022.08.036] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/17/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022]
Abstract
Magnesium cation (Mg2+) has been an emerging therapeutic agent for inducing vascularized bone regeneration. However, the therapeutic effects of current magnesium (Mg) -containing biomaterials are controversial due to the concentration- and stage-dependent behavior of Mg2+. Here, we first provide an overview of biochemical mechanism of Mg2+ in various concentrations and suggest that 2-10 mM Mg2+in vitro may be optimized. This review systematically summarizes and discusses several types of controlled Mg2+ delivery systems based on polymer-Mg composite scaffolds and Mg-containing hydrogels, as well as their design philosophy and several parameters that regulate Mg2+ release. Given that the continuous supply of Mg2+ may prevent biomineral deposition in the later stage of bone regeneration and maturation, we highlight the controlled delivery of Mg2+ based dual- or multi-ions system, especially for the hierarchical therapeutic ion release system, which shows enhanced biomineralization. Finally, the remaining challenges and perspectives of Mg-containing biomaterials for future in situ bone tissue engineering are discussed as well.
Collapse
Affiliation(s)
- Zuoying Yuan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Zhuo Wan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China
| | - Chenyuan Gao
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yue Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China; Beijing Innovation Centre for Engineering Science and Advanced Technology, Peking University, Beijing 100871, China.
| | - Qing Cai
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China..
| |
Collapse
|
31
|
Zheng L, Zhuang Z, Li Y, Shi T, Fu K, Yan W, Zhang L, Wang P, Li L, Jiang Q. Bone targeting antioxidative nano-iron oxide for treating postmenopausal osteoporosis. Bioact Mater 2022; 14:250-261. [PMID: 35310348 PMCID: PMC8897644 DOI: 10.1016/j.bioactmat.2021.11.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/07/2021] [Accepted: 11/07/2021] [Indexed: 12/15/2022] Open
Affiliation(s)
- Liming Zheng
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Zaikai Zhuang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Yixuan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Tianshu Shi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Kai Fu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Wenjin Yan
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Lei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
| | - Peng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, 210008, PR China
- Corresponding author. State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| | - Lan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, 210008, PR China
- Corresponding author. State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China
- Jiangsu Engineering Research Center for 3D Bioprinting, Nanjing, 210008, PR China
- Corresponding author. State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| |
Collapse
|
32
|
Li Z, Wang H, Zhang K, Yang B, Xie X, Yang Z, Kong L, Shi P, Zhang Y, Ho YP, Zhang ZY, Li G, Bian L. Bisphosphonate-based hydrogel mediates biomimetic negative feedback regulation of osteoclastic activity to promote bone regeneration. Bioact Mater 2022; 13:9-22. [PMID: 35224288 PMCID: PMC8844702 DOI: 10.1016/j.bioactmat.2021.11.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/04/2021] [Accepted: 11/04/2021] [Indexed: 12/13/2022] Open
Abstract
The intricate dynamic feedback mechanisms involved in bone homeostasis provide valuable inspiration for the design of smart biomaterial scaffolds to enhance in situ bone regeneration. In this work, we assembled a biomimetic hyaluronic acid nanocomposite hydrogel (HA-BP hydrogel) by coordination bonds with bisphosphonates (BPs), which are antiosteoclastic drugs. The HA-BP hydrogel exhibited expedited release of the loaded BP in response to an acidic environment. Our in vitro studies showed that the HA-BP hydrogel inhibits mature osteoclastic differentiation of macrophage-like RAW264.7 cells via the released BP. Furthermore, the HA-BP hydrogel can support the initial differentiation of primary macrophages to preosteoclasts, which are considered essential during bone regeneration, whereas further differentiation to mature osteoclasts is effectively inhibited by the HA-BP hydrogel via the released BP. The in vivo evaluation showed that the HA-BP hydrogel can enhance the in situ regeneration of bone. Our work demonstrates a promising strategy to design biomimetic biomaterial scaffolds capable of regulating bone homeostasis to promote bone regeneration. HA-BP hydrogel can mediate the expedited release of BP in response to the acidic microenvironment created by osteoclasts. HA-BP hydrogel supports preosteoclastic differentiation, but inhibits the further osteoclastic maturation. The implantation of HA-BP hydrogel in critical-sized bone defects significantly promotes in situ bone regeneration in vivo.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Haixing Wang
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Kunyu Zhang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
| | - Boguang Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Xian Xie
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Zhengmeng Yang
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Lingchi Kong
- Department of Orthopaedic Surgery, Affiliated Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai, 200233, China
| | - Peng Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Yuan Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Yi-Ping Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
| | - Zhi-Yong Zhang
- Translational Research Centre of Regenerative Medicine and 3D Printing of Guangzhou Medical University, Guangdong Province Engineering Research Center for Biomedical Engineering, State Key Laboratory of Respiratory Disease, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou City, Guangdong Province, 510150, China
- Corresponding author.
| | - Gang Li
- Department of Orthopaedic and Traumatology, The Chinese University of Hong Kong, Sha Tin, New Territories, 999077, Hong Kong, China
- Corresponding author.
| | - Liming Bian
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, China
- Corresponding author. School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
33
|
Zhu T, Jiang M, Zhang M, Cui L, Yang X, Wang X, Liu G, Ding J, Chen X. Construction and validation of steroid-induced rabbit osteonecrosis model. MethodsX 2022; 9:101713. [PMID: 35601954 PMCID: PMC9120059 DOI: 10.1016/j.mex.2022.101713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 04/21/2022] [Indexed: 12/04/2022] Open
Abstract
Osteonecrosis is a common orthopedic disease in clinic, resulting in joint collapse if appropriate treatment is not given in time. The clinical usage of high-dose steroid is one of the common causes of osteonecrosis. In several studies, the intravenous injection of steroid with or without lipopolysaccharide is the most commonly used strategy to construct osteonecrosis animal model. However, the injection dose, frequency, and interval of steroid and validation of successful model construction lack generally accepted protocol, and the survival and model formation rates are unsatisfactory. We have optimized the construction protocol of osteonecrosis animal model based on the previously reported ones and established a mature animal model of osteonecrosis for future studies.A rabbit model of osteonecrosis was constructed by multiple injections of high-dose methylprednisolone. The multidisciplinary biomedical examinations demonstrated the successful construction of osteonecrosis model in the rabbit.
Collapse
|
34
|
Zhu WY, Guo J, Yang WF, Tao ZY, Lan X, Wang L, Xu J, Qin L, Su YX. Biodegradable magnesium implant enhances angiogenesis and alleviates medication-related osteonecrosis of the jaw in rats. J Orthop Translat 2022; 33:153-161. [PMID: 35415073 PMCID: PMC8965768 DOI: 10.1016/j.jot.2022.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/26/2022] [Accepted: 03/12/2022] [Indexed: 01/01/2023] Open
Abstract
Background Medication-related osteonecrosis of the jaw (MRONJ) is a serious complication associated with antiresorptive and antiangiogenic medications, of which impaired angiogenesis is a key pathological alteration. Since Magnesium (Mg)-based implants possess proangiogenic effects, we hypothesized that the biodegradable Mg implant could alleviate the development of MRONJ via enhancing angiogenesis. Methods MRONJ model was established and divided into the Veh + Ti group (Vehicle-treated rat, with Titanium (Ti) implant), BP + Ti group (Bisphosphonate (BP)-treated rat, with Ti implant), BP + Mg group (BP-treated rat, with Mg implant), BP + Mg + SU5416 group (BP-treated rat, with Mg implant and vascular endothelial growth factor (VEGF) receptor-2 inhibitor), BP + Mg + BIBN group (BP-treated rat, with Mg implant and calcitonin gene-related peptide (CGRP) receptor antagonist), and BP + Mg + SU5416+BIBN group (BP-treated rat, with Mg implant and VEGF receptor-2 inhibitor and CGRP receptor antagonist). The occurrence of MRONJ, alveolar bone necrosis, new bone formation and vessel formation were assessed by histomorphometry, immunohistochemistry, and micro-CT analysis. Results Eight weeks after surgery, the BP + Mg group had significantly reduced occurrence of MRONJ-like lesion and histological osteonecrosis, increased bone microstructural parameters, and increased expressions of VEGFA and CGRP, than the BP + Ti group. By simultaneously blocking VEGF receptor-2 and CGRP receptor, the vessel volume and new bone formation in the BP + Mg group were significantly decreased, meanwhile the occurrence of MRONJ-like lesion and histological bone necrosis were significantly increased. Conclusion Biodegradable Mg implant could alleviate the development of MRONJ-like lesion, possibly via upregulating VEGF- and CGRP-mediated angiogenesis. Mg-based implants have the translational potential to be developed as a novel internal fixation device for patients with the risk of MRONJ. The Translational potential of this article This work reports a biodegradable Mg implant which ameliorates the development of MRONJ-like lesions possibly due to its angiogenic property. Mg-based implants have the potential to be developed as a novel internal fixation device for patients at the risk of MRONJ.
Collapse
Affiliation(s)
- Wang-yong Zhu
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Jiaxin Guo
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Wei-fa Yang
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Zhuo-ying Tao
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Xinmiao Lan
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Leilei Wang
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Jiankun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yu-xiong Su
- Division of Oral and Maxillofacial Surgery, Faculty of Dentistry, The University of Hong Kong, Hong Kong Special Administrative Region
- Corresponding author.
| |
Collapse
|
35
|
Dai B, Xu J, Li X, Huang L, Hopkins C, Wang H, Yao H, Mi J, Zheng L, Wang J, Tong W, Chow DHK, Li Y, He X, Hu P, Chen Z, Zu H, Li Y, Yao Y, Jiang Q, Qin L. Macrophages in epididymal adipose tissue secrete osteopontin to regulate bone homeostasis. Nat Commun 2022; 13:427. [PMID: 35058428 PMCID: PMC8776868 DOI: 10.1038/s41467-021-27683-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 11/04/2021] [Indexed: 12/21/2022] Open
Abstract
Epididymal white adipose tissue (eWAT) secretes an array of cytokines to regulate the metabolism of organs and tissues in high-fat diet (HFD)-induced obesity, but its effects on bone metabolism are not well understood. Here, we report that macrophages in eWAT are a main source of osteopontin, which selectively circulates to the bone marrow and promotes the degradation of the bone matrix by activating osteoclasts, as well as modulating bone marrow-derived macrophages (BMDMs) to engulf the lipid droplets released from adipocytes in the bone marrow of mice. However, the lactate accumulation induced by osteopontin regulation blocks both lipolysis and osteoclastogenesis in BMDMs by limiting the energy regeneration by ATP6V0d2 in lysosomes. Both surgical removal of eWAT and local injection of either clodronate liposomes (for depleting macrophages) or osteopontin-neutralizing antibody show comparable amelioration of HFD-induced bone loss in mice. These results provide an avenue for developing therapeutic strategies to mitigate obesity-related bone disorders.
Collapse
Affiliation(s)
- Bingyang Dai
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiankun Xu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xu Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Le Huang
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Chelsea Hopkins
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Honglian Wang
- Research Center for Integrated Medicine, Affiliated Traditional Medicine Hospital of Southwest Medical University, 646000, Luzhou, Sichuan, China
| | - Hao Yao
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jie Mi
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Lizhen Zheng
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiali Wang
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Wenxue Tong
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Dick Ho-Kiu Chow
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ye Li
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xuan He
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Peijie Hu
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Ziyi Chen
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Haiyue Zu
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yixuan Li
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yao Yao
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial & Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
36
|
Lu X, Guo H, Li J, Sun T, Xiong M. Recombinant Human Bone Morphogenic Protein-2 Immobilized Fabrication of Magnesium Functionalized Injectable Hydrogels for Controlled-Delivery and Osteogenic Differentiation of Rat Bone Marrow-Derived Mesenchymal Stem Cells in Femoral Head Necrosis Repair. Front Cell Dev Biol 2021; 9:723789. [PMID: 34900987 PMCID: PMC8656218 DOI: 10.3389/fcell.2021.723789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Femoral head necrosis (FHN) is a clinically progressive disease that leads to overwhelming complications without an effective therapeutic approach. In recent decades, transplantation of mesenchymal stem cells (MSCs) has played a promising role in the treatment of FHN in the initial stage; however, the success rate is still low because of unsuitable cell carriers and abridged osteogenic differentiation of the transplanted MSCs. Biopolymeric-derived hydrogels have been extensively applied as effective cell carriers and drug vesicles; they provide the most promising contributions in the fields of tissue engineering and regenerative medicine. However, the clinical potential of hydrogels may be limited because of inappropriate gelation, swelling, mechanical characteristics, toxicity in the cross-linking process, and self-healing ability. Naturally, gelated commercial hydrogels are not suitable for cell injection and infiltration because of their static network structure. In this study, we designed a novel thermogelling injectable hydrogel using natural silk fibroin-blended chitosan (CS) incorporated with magnesium (Mg) substitutes to improve physical cross-linking, stability, and cell osteogenic compatibility. The presented observations demonstrate that the developed injectable hydrogels can facilitate the controlled delivery of immobilized recombinant human bone morphogenic protein-2 (rhBMP-2) and rat bone marrow-derived MSCs (rBMSCs) with greater cell encapsulation efficiency, compatibility, and osteogenic differentiation. In addition, outcomes of in vivo animal studies established promising osteoinductive, bone mineral density, and bone formation rate after implantation of the injectable hydrogel scaffolds. Therefore, the developed hydrogels have great potential for clinical applications of FHN therapy.
Collapse
Affiliation(s)
- Xueliang Lu
- Department of Orthopedics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Hongyu Guo
- Clinical Medical College, Henan University of Science and Technology, Luoyang, China
| | - Jiaju Li
- Clinical Medical College, Henan University of Science and Technology, Luoyang, China
| | - Tianyu Sun
- Clinical Medical College, Henan University of Science and Technology, Luoyang, China
| | - Mingyue Xiong
- Department of Orthopedics, The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
37
|
Zheng L, Huang L, Chen Z, Cui C, Zhang R, Qin L. Magnesium supplementation alleviates corticosteroid-associated muscle atrophy in rats. Eur J Nutr 2021; 60:4379-4392. [PMID: 34052917 PMCID: PMC8164831 DOI: 10.1007/s00394-021-02598-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 05/21/2021] [Indexed: 01/22/2023]
Abstract
PURPOSE Corticosteroid (CS) therapy for infectious and rheumatological diseases showed to decrease serum magnesium (Mg++) level and induce muscle atrophy in patients. The present study investigated the effects of Mg++ supplementation on preventing CS-induced muscle atrophy in an animal model, which provided experimental data for potential clinical translation. METHODS Twelve 24-week-old male Sprague-Dawley rats were treated with lipopolysaccharide (LPS) and CS methylprednisolone (MPS) to induce muscle atrophy, with half of the rats also given daily 50 mg/kg Mg++ oral supplementation. Additional six rats without LPS + CS treatments were used as normal controls. After treatment for 6 weeks, serum was collected for Mg++ quantification, animal dual-energy X-ray absorptiometry (DXA) was performed for tissue composition, and the extensor digitorum longus (EDL) was collected for muscle functional test and histology including muscle fiber size, intramuscular fat infiltration and fiber typing. In vitro myotube atrophy model was used to study the in vitro effect associated with in vivo muscle atrophy. RESULTS LPS + CS treatments induced hypomagnesemia while the serum Mg++ level was in normal range after Mg++ supplementation. DXA showed 53.0% lower fat percent and 29.7% higher lean mass in LPS + CS + Mg group when compared to LPS + CS group. Muscle functional test showed 22.2% higher specific twitch force and 40.3% higher specific tetanic force in LPS + CS + Mg group when compared to LPS + CS group. Histological analysis showed 4.1% higher proportion of muscle fibers area to total area and 63.6% lower intramuscular fat infiltration in EDL sections in LPS + CS + Mg group when compared to LPS + CS group. LPS + CS + Mg group had 33.0% higher area proportion and 29.4% higher cross-sectional area (CSA) of type IIb muscle fiber. Myoblast culture results showed that Mg++ supplementation group had larger myotube diameter. The mRNA expressions of the muscle atrophy marker genes MuRF1 and MAFbx were lower in Mg++ supplementation group both in vitro and in vivo. CONCLUSION The current study demonstrated that Mg++ supplementation successfully alleviated CS-associated muscle atrophy in rats at both functional and morphology levels, indicating a translational potential for patients undergoing CS therapy. This study provided the evidence for the first time that Mg++ supplementation could prevent muscle atrophy-an adverse effect of CS therapy, currently also adopted for treating coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Lizhen Zheng
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Rm74026, 5/F, Clinical Science Building, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science and Innovation, Chinese Academy of Sciences Limited, Hong Kong SAR, People's Republic of China
| | - Le Huang
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Rm74026, 5/F, Clinical Science Building, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China
| | - Ziyi Chen
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Rm74026, 5/F, Clinical Science Building, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China
| | - Can Cui
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Rm74026, 5/F, Clinical Science Building, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China
| | - Ri Zhang
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Rm74026, 5/F, Clinical Science Building, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China
| | - Ling Qin
- Musculoskeletal Research Laboratory of Department of Orthopaedics and Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Rm74026, 5/F, Clinical Science Building, Prince of Wales Hospital, Shatin, Hong Kong SAR, People's Republic of China.
- Hong Kong-Shenzhen Innovation and Technology Institute (Futian), The Chinese University of Hong Kong, Hong Kong SAR, People's Republic of China.
| |
Collapse
|
38
|
Sahin E, Orhan C, Balci TA, Erten F, Sahin K. Magnesium Picolinate Improves Bone Formation by Regulation of RANK/RANKL/OPG and BMP-2/Runx2 Signaling Pathways in High-Fat Fed Rats. Nutrients 2021; 13:3353. [PMID: 34684352 PMCID: PMC8538721 DOI: 10.3390/nu13103353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/20/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Magnesium (Mg) deficiency may affect bone metabolism by increasing osteoclasts, decreasing osteoblasts, promoting inflammation/oxidative stress, and result in subsequent bone loss. The objective of the present study was to identify the molecular mechanism underlying the bone protective effect of different forms of Mg (inorganic magnesium oxide (MgO) versus organic magnesium picolinate (MgPic) compound) in rats fed with a high-fat diet (HFD). Forty-two Wistar albino male rats were divided into six group (n = 7): (i) control, (ii) MgO, (iii) MgPic, (iv) HFD, (v) HFD + MgO, and (vi) HFD + MgPic. Bone mineral density (BMD) increased in the Mg supplemented groups, especially MgPic, as compared with the HFD group (p < 0.001). As compared with the HFD + MgO group, the HFD + MgPic group had higher bone P (p < 0.05) and Mg levels (p < 0.001). In addition, as compared to MgO, MgPic improved bone formation by increasing the levels of osteogenetic proteins (COL1A1 (p < 0.001), BMP2 (p < 0.001), Runx2 (p < 0.001), OPG (p < 0.05), and OCN (p < 0.001), IGF-1 (p < 0.001)), while prevented bone resorption by reducing the levels of RANK and RANKL (p < 0.001). In conclusion, the present data showed that the MgPic could increase osteogenic protein levels in bone more effectively than MgO, prevent bone loss, and contribute to bone formation in HFD rats.
Collapse
Affiliation(s)
- Emre Sahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig 23119, Turkey; (E.S.); (C.O.)
| | - Cemal Orhan
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig 23119, Turkey; (E.S.); (C.O.)
| | - Tansel Ansal Balci
- Department of Nuclear Medicine, School of Medicine, Firat University, Elazig 23119, Turkey;
| | - Fusun Erten
- Department of Veterinary Medicine, Pertek Sakine Genc Vocational School, Munzur University, Tunceli 62500, Turkey;
| | - Kazim Sahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig 23119, Turkey; (E.S.); (C.O.)
| |
Collapse
|
39
|
Xu J, Hu P, Zhang X, Chen J, Wang J, Zhang J, Chen Z, Yu MK, Chung YW, Wang Y, Zhang X, Zhang Y, Zheng N, Yao H, Yue J, Chan HC, Qin L, Ruan YC. Magnesium implantation or supplementation ameliorates bone disorder in CFTR-mutant mice through an ATF4-dependent Wnt/β-catenin signaling. Bioact Mater 2021; 8:95-108. [PMID: 34541389 PMCID: PMC8424424 DOI: 10.1016/j.bioactmat.2021.06.034] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/26/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023] Open
Abstract
Magnesium metal and its alloys are being developed as effective orthopedic implants; however, the mechanisms underlying the actions of magnesium on bones remain unclear. Cystic fibrosis, the most common genetic disease in Caucasians caused by the mutation of CFTR, has shown bone disorder as a key clinical manifestation, which currently lacks effective therapeutic options. Here we report that implantation of magnesium-containing implant stimulates bone formation and improves bone fracture healing in CFTR-mutant mice. Wnt/β-catenin signaling in the bone is enhanced by the magnesium implant, and inhibition of Wnt/β-catenin by iCRT14 blocks the magnesium implant to improve fracture healing in CFTR-mutant mice. We further demonstrate that magnesium ion enters osteocytes, increases intracellular cAMP level and activates ATF4, a key transcription factor known to regulate Wnt/β-catenin signaling. In vivo knockdown of ATF4 abolishes the magnesium implant-activated β-catenin in bones and reverses the improved-fracture healing in CFTR-mutant mice. In addition, oral supplementation of magnesium activates ATF4 and β-catenin as well as enhances bone volume and density in CFTR-mutant mice. Together, these results show that magnesium implantation or supplementation may serve as a potential anabolic therapy for cystic fibrosis-related bone disease. Activation of ATF4-dependent Wnt/β-catenin signaling in osteocytes is identified as a previously undefined mechanism underlying the beneficial effect of magnesium on bone formation. Magnesium implant ameliorates bone defects and improves the impaired bone fracture healing in CFTR-deficient mice. Oral magnesium supplementation improves bone quality in CFTR-deficient mice. Extracellular Mg2+ enters bone cells through Mg2+ channels and transporters. Mg2+ elevates cAMP level to activate ATF4-dependent Wnt/β-catenin signalingin bone cells.
Collapse
Affiliation(s)
- Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Peijie Hu
- Deparment of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Xiaotian Zhang
- Deparment of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Junjiang Chen
- Deparment of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China.,Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiali Wang
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, China
| | - Jieting Zhang
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ziyi Chen
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China.,Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Mei Kuen Yu
- Deparment of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China.,Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yiu Wa Chung
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yan Wang
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaohu Zhang
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yifeng Zhang
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Nianye Zheng
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Hao Yao
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiang Yue
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Hsiao Chang Chan
- Epithelial Cell Biology Research Centre, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopedics & Traumatology, The Chinese University of Hong Kong, Hong Kong, China
| | - Ye Chun Ruan
- Deparment of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
40
|
Wu Q, Xu S, Wang F, He B, Wang X, Sun Y, Ning C, Dai K. Double-edged effects caused by magnesium ions and alkaline environment regulate bioactivities of magnesium-incorporated silicocarnotite in vitro. Regen Biomater 2021; 8:rbab016. [PMID: 34484805 PMCID: PMC8411036 DOI: 10.1093/rb/rbab016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 12/17/2022] Open
Abstract
Magnesium (Mg) is an important element for its enhanced osteogenic and angiogenic properties in vitro and in vivo, however, the inherent alkalinity is the adverse factor that needs further attention. In order to study the role of alkalinity in regulating osteogenesis and angiogenesis in vitro, magnesium-silicocarnotite [Mg-Ca5(PO4)2SiO4, Mg-CPS] was designed and fabricated. In this study, Mg-CPS showed better osteogenic and angiogenic properties than CPS within 10 wt.% magnesium oxide (MgO), since the adversity of alkaline condition was covered by the benefits of improved Mg ion concentrations through activating Smad2/3-Runx2 signaling pathway in MC3T3-E1 cells and PI3K-AKT signaling pathway in human umbilical vein endothelial cells in vitro. Besides, provided that MgO was incorporated with 15 wt.% in CPS, the bioactivities had declined due to the environment consisting of higher-concentrated Mg ions, stronger alkalinity and lower Ca/P/Si ions caused. According to the results, it indicated that bioactivities of Mg-CPS in vitro were regulated by the double-edged effects, which were the consequence of Mg ions and alkaline environment combined. Therefore, if MgO is properly incorporated in CPS, the improved bioactivities could cover alkaline adversity, making Mg-CPS bioceramics promising in orthopedic clinical application for its enhancement of osteogenesis and angiogenesis in vitro.
Collapse
Affiliation(s)
- Qiang Wu
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, Huangpu District 200011, China
| | - Shunxiang Xu
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, No. 100, Guilin Road, Shanghai, Xuhui District 200234, China
| | - Fei Wang
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, No. 100, Guilin Road, Shanghai, Xuhui District 200234, China
| | - Bo He
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, No. 100, Guilin Road, Shanghai, Xuhui District 200234, China
| | - Xin Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, No.169, East Lake Road, Wuchang District, Wuhan 430071, China
| | - Ye Sun
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, No.300, Guangzhou Road, Drum-tower District, Nanjing, 210029, China
| | - Congqin Ning
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, No. 100, Guilin Road, Shanghai, Xuhui District 200234, China.,State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, No.1295, Dingxi Road, Changning District, Shanghai 200050, China
| | - Kerong Dai
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai Key Laboratory of Orthopedic Implant, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai, Huangpu District 200011, China
| |
Collapse
|
41
|
Zhang X, Liu W, Liu J, Hu Y, Dai H. Poly-ε-caprolactone/Whitlockite Electrospun Bionic Membrane with an Osteogenic-Angiogenic Coupling Effect for Periosteal Regeneration. ACS Biomater Sci Eng 2021; 7:3321-3331. [PMID: 34148343 DOI: 10.1021/acsbiomaterials.1c00426] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The periosteum is rich in vascular networks, osteoprogenitor cells, and stem cells and plays an important role in bone defect repair. However, existing artificial periosteum materials still have difficulty in meeting clinical requirements, such as good mechanical properties and bionic structure construction, osteogenic differentiation, and vascularization capabilities. Here, a poly-ε-caprolactone (PCL)/whitlockite (WH, 5, 10, 15 wt %) artificial periosteum with different doping amounts was prepared by electrospinning technology. According to the results of in vitro mineralization experiments, the rapid ion release from WH promotes the deposition of mineralized hydroxyapatite. Inductively coupled plasma-optical emission spectroscopy, in vitro angiogenesis, and cell migration experiments showed that the bionic periosteum of the 15% WH group had the best release rate of Mg2+ and the best ability to promote the human umbilical vein endothelial cell angiogenesis and migration. In addition, this group promoted collagen formation and calcium deposition. Finally, the subcutaneous implantation model was used to verify the biocompatibility and angiogenesis ability of the proposed membrane in vivo. Overall, this biomimetic PCL/WH nanofiber membrane combines the positive osteogenic differentiation ability and angiogenic ability of calcium phosphate materials and thus has good application prospects in the field of periosteal repair in the future.
Collapse
Affiliation(s)
- Xiangke Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Wenbin Liu
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China.,Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, Hunan 410008, China
| | - Jiawei Liu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Yihe Hu
- Department of Orthopedics, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China.,Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Changsha, Hunan 410008, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P. R. China.,Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan 528200, China
| |
Collapse
|
42
|
Long J, Zhang W, Chen Y, Teng B, Liu B, Li H, Yao Z, Wang D, Li L, Yu XF, Qin L, Lai Y. Multifunctional magnesium incorporated scaffolds by 3D-Printing for comprehensive postsurgical management of osteosarcoma. Biomaterials 2021; 275:120950. [PMID: 34119886 DOI: 10.1016/j.biomaterials.2021.120950] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 05/25/2021] [Accepted: 05/29/2021] [Indexed: 12/12/2022]
Abstract
Clinical treatment of Osteosarcoma (OS) encounters great challenges of postsurgical tumor recurrence and extensive bone defect. To address these issues, innovative multifunctional PLGA/Mg porous scaffolds were designed for comprehensive postsurgical management of OS. The PLGA/Mg composite scaffolds exhibited several unique features: (1) The multiple functions of Mg particles were explored for the first time to fulfill the requirement for postsurgical management of OS. The intact Mg particles exhibits excellent photothermal effect for tumor eradication, and the released Mg ions could subsequently promote bone regeneration, thus endowing the PLGA/Mg scaffolds dual functions of suppressing OS recurrence and repairing bone defect in a sequential way; (2) A low temperature rapid prototyping (LT-RP) 3D-printing technology was used to fabricate the scaffolds with biomimetic hierarchical porous structures, which could structurally promote bone regeneration; (3) The PLGA/Mg scaffolds have excellent biodegradability and biocompatibility, exhibiting great promise for clinical translation. Finally, the PLGA/Mg scaffolds achieved complete suppression of tumor recurrence in the presence of near-infrared laser irradiation, as well as efficient bone defect repair in vivo. Activation of the AKT and β-catenin pathways of osteoblast cells by PLGA/Mg scaffolds was identified, which might be the modulators to accelerate the ossification. The innovative PLGA/Mg scaffolds demonstrated excellent capabilities in postsurgical OS recurrence suppression and bone regeneration, providing a promising clinical strategy for comprehensive postsurgical management of OS.
Collapse
Affiliation(s)
- Jing Long
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Wei Zhang
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Yingqi Chen
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bin Teng
- Research Center for Reproduction and Health Development, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ben Liu
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Huilin Li
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Zhenyu Yao
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dou Wang
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Long Li
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Xue-Feng Yu
- Materials and Interfaces Center, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ling Qin
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, SAR, Hong Kong, China; CAS-HK Joint Lab of Biomaterials, Shenzhen, China
| | - Yuxiao Lai
- Centre for Translational Medicine Research & Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China; Key Laboratory of Health Informatics, Chinese Academy of Sciences, Shenzhen, China; CAS-HK Joint Lab of Biomaterials, Shenzhen, China.
| |
Collapse
|
43
|
Pan P, Yue Q, Li J, Gao M, Yang X, Ren Y, Cheng X, Cui P, Deng Y. Smart Cargo Delivery System based on Mesoporous Nanoparticles for Bone Disease Diagnosis and Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2004586. [PMID: 34165902 PMCID: PMC8224433 DOI: 10.1002/advs.202004586] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/11/2021] [Indexed: 05/05/2023]
Abstract
Bone diseases constitute a major issue for modern societies as a consequence of progressive aging. Advantages such as open mesoporous channel, high specific surface area, ease of surface modification, and multifunctional integration are the driving forces for the application of mesoporous nanoparticles (MNs) in bone disease diagnosis and treatment. To achieve better therapeutic effects, it is necessary to understand the properties of MNs and cargo delivery mechanisms, which are the foundation and key in the design of MNs. The main types and characteristics of MNs for bone regeneration, such as mesoporous silica (mSiO2 ), mesoporous hydroxyapatite (mHAP), mesoporous calcium phosphates (mCaPs) are introduced. Additionally, the relationship between the cargo release mechanisms and bone regeneration of MNs-based nanocarriers is elucidated in detail. Particularly, MNs-based smart cargo transport strategies such as sustained cargo release, stimuli-responsive (e.g., pH, photo, ultrasound, and multi-stimuli) controllable delivery, and specific bone-targeted therapy for bone disease diagnosis and treatment are analyzed and discussed in depth. Lastly, the conclusions and outlook about the design and development of MNs-based cargo delivery systems in diagnosis and treatment for bone tissue engineering are provided to inspire new ideas and attract researchers' attention from multidisciplinary areas spanning chemistry, materials science, and biomedicine.
Collapse
Affiliation(s)
- Panpan Pan
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Qin Yue
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610051, China
| | - Juan Li
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Meiqi Gao
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Xuanyu Yang
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Yuan Ren
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Xiaowei Cheng
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
| | - Penglei Cui
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yonghui Deng
- Department of Chemistry, Department of Gastroenterology, Zhongshan Hospital of Fudan University, State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200433, China
- State Key Lab of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| |
Collapse
|
44
|
Liu Y, Wang Q, Zhang Z, Fu R, Zhou T, Long C, He T, Yang D, Li Z, Peng S. Magnesium supplementation enhances mTOR signalling to facilitate myogenic differentiation and improve aged muscle performance. Bone 2021; 146:115886. [PMID: 33592327 DOI: 10.1016/j.bone.2021.115886] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/08/2021] [Accepted: 02/09/2021] [Indexed: 12/25/2022]
Abstract
Magnesium (Mg2+), as an essential mineral, supports and sustains the health and activity of the organs of the human body. Despite some clinical evidence on the association of Mg2+ deficiency with muscle regeneration dysfunction and sarcopenia in older-aged individuals, there is no consensus on the action mode and molecular mechanism by which Mg2+ influences aged muscle size and function. Here, we identified the appropriate Mg2+ environment that promotes the myogenic differentiation and myotube hypertrophy in both C2C12 myoblast and primary aged muscle stem cell (MuSC). Through animal experiments, we demonstrated that Mg2+ supplementation in aged mice significantly promotes muscle regeneration and conserves muscle mass and strength. Mechanistically, Mg2+ stimulation activated the mammalian target of rapamycin (mTOR) signalling, inducing the myogenic differentiation and protein synthesis, which consequently offers protections against the age-related decline in muscle regenerative potential and muscle mass. These findings collectively provide a promising therapeutic strategy for MuSC dysfunction and sarcopenia through Mg2+ supplementation in the elderly.
Collapse
Affiliation(s)
- Yuantong Liu
- Department of Spine Surgery, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China; The First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Qinghe Wang
- Department of Spine Surgery, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Zengfu Zhang
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Runhan Fu
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tianjian Zhou
- Shenzhen Key Laboratory of Tissue and Functional Reconstruction of Sports System, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China
| | - Canling Long
- Department of Spine Surgery, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Tongzhong He
- Department of Spine Surgery, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China
| | - Dazhi Yang
- Department of Spine Surgery, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China.
| | - Zhizhong Li
- The First Affiliated Hospital, Jinan University, Guangzhou 510630, China.
| | - Songlin Peng
- Department of Spine Surgery, Shenzhen People's Hospital, The Second Clinical Medical College, Jinan University, Shenzhen 518020, China; Shenzhen Key Laboratory of Tissue and Functional Reconstruction of Sports System, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen 518020, China.
| |
Collapse
|
45
|
Liu Y, Wu J, Zhang H, Wu Y, Tang C. Covalent immobilization of the phytic acid-magnesium layer on titanium improves the osteogenic and antibacterial properties. Colloids Surf B Biointerfaces 2021; 203:111768. [PMID: 33872830 DOI: 10.1016/j.colsurfb.2021.111768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 03/03/2021] [Accepted: 04/12/2021] [Indexed: 11/15/2022]
Abstract
In order to improve early osseointegration and long-term survival rate of implants, a multifunctional titanium surface that promotes osteogenesis and antibacterial properties is expected. Incorporation of bioactive trace elements such as magnesium ions was proved a promising method to improve osseointegration of titanium. Phytic acid has strong chelating ability with multivalent cations, which has been used in surface modification. Moreover, phytic acid was proved antibacterial potential. Herein, to improve the osteogenic and antibacterial properties, a phytic acid-magnesium (PA-Mg) layer was introduced on titanium using phytic acid as a cross-linker molecule. No obvious changes of the surface characterization were observed by scanning electron microscopy and atomic force microscopy. X-ray photoelectron spectroscopy confirmed that the PA-Mg layer covalently bond to the Ti surface, and the thickness of the PA-Mg layer was about 150 nm. Besides, improved hydrophilic and more protein adsorption were observed on Ti-PA-Mg. Notably, a relatively controlled magnesium release was also observed on Ti-PA-Mg. Human bone mesenchymal stem cells showed better adhesion, proliferation, and osteogenic differentiation on Ti-PA-Mg samples, indicating improved biocompatibility and osteoinductivity. Moreover, Ti-PA-Mg had better antibacterial properties against porphyromonas gingivalis than Ti. Overall, the PA-Mg layer on Ti surface improved the osteogenic and antibacterial properties, which may have promise for use in dental implantation.
Collapse
Affiliation(s)
- Yajing Liu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China; Department of Dental Implantology, Affiliated Stomatological Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Jin Wu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China; Department of Dental Implantology, Affiliated Stomatological Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Hao Zhang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China; Department of Dental Implantology, Affiliated Stomatological Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China; Department of Stomatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yizhen Wu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China; Department of Dental Implantology, Affiliated Stomatological Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China
| | - Chunbo Tang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, 210029, China; Department of Dental Implantology, Affiliated Stomatological Hospital of Stomatology, Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
46
|
Oakes B, Bolia IK, Weber AE, Petrigliano FA. Vitamin C in orthopedic practices: Current concepts, novel ideas, and future perspectives. J Orthop Res 2021; 39:698-706. [PMID: 33300201 DOI: 10.1002/jor.24947] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 02/04/2023]
Abstract
Vitamin C (ascorbic acid), is an important antioxidant that has been applied broadly in the field of orthopaedics. Current research on vitamin C examines the molecule's role in bone and tendon physiology, as well as joint replacement and Postoperative pain. Most laboratory and human studies associate the use of vitamin C with improved bone health and tendon healing. Recent literature moderately supports the use of vitamin C to improve functional outcomes, decreased postoperative pain, and prevent complex regional pain syndrome following orthopaedic procedures. The perioperative use of vitamin C in patients undergoing joint replacement surgery and anterior cruciate ligament reconstruction is still under investigation. Overall, there is need for high-quality human trials to confirm whether vitamin C can potentiate the outcomes of orthopaedic procedures and to determine optimal dosage and means of administration to maximize its proposed benefits. The purpose of this review was to summarize the application of vitamin C in orthopaedic practices and to identify potential areas for future study.
Collapse
Affiliation(s)
- Bennett Oakes
- Department of Orthopaedic Surgery, University of Southern California Los Angeles, Los Angeles, California, USA
| | - Ioanna K Bolia
- Department of Orthopaedic Surgery, University of Southern California Los Angeles, Los Angeles, California, USA
| | - Alexander E Weber
- Department of Orthopaedic Surgery, University of Southern California Los Angeles, Los Angeles, California, USA
| | - Frank A Petrigliano
- Department of Orthopaedic Surgery, University of Southern California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
47
|
Zhao J, Yue T, Lu S, Meng H, Lin Q, Ma H, Liu G, Li H, Lu Q, Wang A, Xu W, Feng J, Wan Y, Liao S, Zhou X, Peng J. Local administration of zoledronic acid prevents traumatic osteonecrosis of the femoral head in rat model. J Orthop Translat 2021; 27:132-138. [PMID: 33786320 PMCID: PMC7972932 DOI: 10.1016/j.jot.2020.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 12/05/2022] Open
Abstract
Background Osteonecrosis of the femoral head (ONFH) is a refractory disease due to its unclear pathomechanism. Neither conservative treatment nor surgical treatment during the early stage of ONFH achieves satisfactory results. Therefore, this study aims to explore the available evidence on the effect of zoledronic acid on early-stage ONFH. Methods For groups were established:the Normal group, model group, Normal saline group(NS group) and zoledronic acid-treated group. The blood supply to the femoral head of animals in the model group and zoledronic acid-treated group was interrupted via a surgical procedure, and zoledronic acid was then locally administered to the femoral head. Four weeks after surgery, all the hips were harvested and evaluated by micro-CT and histopathology(H&E staining, TRAP staining, Toluidine blue staining and masson staining). Results The values of BMD, BS/BV and Tb.Th in the Normal group and zoledronic acid-treated group were significantly higher than those in the model group and NS group (p < 0.05). The outcome of H&E staining, Toluidine blue staining and masson staining were consistent with that of micro-CT. Conclusion The local administration of zoledronic acid in the femoral head had positive effects on the bone structure of the femoral head in a modified rat model of traumatic ONFH and offered a promising therapeutic strategy during the early stage of ONFH. The Translational potential of this article This article could provide a choice for treating patients who have osteonecrosis of femora head and can be the basic research for advanced development over this disease
Collapse
Affiliation(s)
- Jun Zhao
- Medical School of Chinese PLA, Beijing, 100853, China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Tian Yue
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Shibi Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Haoye Meng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Qiuxia Lin
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Haiyang Ma
- Medical School of Chinese PLA, Beijing, 100853, China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Guangbo Liu
- Medical School of Chinese PLA, Beijing, 100853, China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Huo Li
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Qiang Lu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Aiyuan Wang
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Wenjing Xu
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Jing Feng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Yiqun Wan
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Sida Liao
- Medical School of Chinese PLA, Beijing, 100853, China.,Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| | - Xuefeng Zhou
- Strategic Support Force Medical Center of chinese PLA, AnxiangBeili, Beijing, 100101, China
| | - Jiang Peng
- Institute of Orthopedics, Chinese PLA General Hospital, Beijing Key Laboratory (No BZ0128), Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, Beijing, 100853, China
| |
Collapse
|
48
|
Li Y, Pan Q, Xu J, He X, Li HA, Oldridge DA, Li G, Qin L. Overview of methods for enhancing bone regeneration in distraction osteogenesis: Potential roles of biometals. J Orthop Translat 2021; 27:110-118. [PMID: 33575164 PMCID: PMC7859169 DOI: 10.1016/j.jot.2020.11.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/10/2020] [Accepted: 11/19/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Distraction osteogenesis (DO) is a functional tissue engineering approach that applies gradual mechanical traction on the bone tissues after osteotomy to stimulate bone regeneration. However, DO still has disadvantages that limit its clinical use, including long treatment duration. METHODS Review the current methods of promoting bone formation and consolidation in DO with particular interest on biometal. RESULTS Numerous approaches, including physical therapy, gene therapy, growth factor-based therapy, stem-cell-based therapy, and improved distraction devices, have been explored to reduce the DO treatment duration with some success. Nevertheless, no approach to date is widely accepted in clinical practice due to various reasons, such as high expense, short biologic half-life, and lack of effective delivery methods. Biometals, including calcium (Ca), magnesium (Mg), zinc (Zn), copper (Cu), manganese (Mn), and cobalt (Co) have attracted attention in bone regeneration attributed to their biodegradability and bioactive components released during in vivo degradation. CONCLUSION This review summarizes the current therapies accelerating bone formation in DO and the beneficial role of biometals in bone regeneration, particularly focusing on the use of biometal Mg and its alloy in promoting bone formation in DO. Translational potential: The potential clinical applications using Mg-based devices to accelerate DO are promising. Mg stimulates expression of multiple intrinsic biological factors and the development of Mg as an implantable component in DO may be used to argument bone formation and consolidation in DO.
Collapse
Affiliation(s)
- Ye Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Qi Pan
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Xuan He
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Helen A. Li
- School of Medicine, University of East Anglia, Norwich, England, UK
| | - Derek A. Oldridge
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, USA
| | - Gang Li
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| | - Ling Qin
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
49
|
Synergistic effects of magnesium ions and simvastatin on attenuation of high-fat diet-induced bone loss. Bioact Mater 2021; 6:2511-2522. [PMID: 33665494 PMCID: PMC7889436 DOI: 10.1016/j.bioactmat.2021.01.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 12/17/2022] Open
Abstract
Introduction Magnesium (Mg) has a prophylactic potential against the onset of hyperlipidemia. Similar to statin, Mg is recommended as lipid-lowering medication for hypercholesterolemia and concomitantly exhibits an association with increased bone mass. The combination of statin with Mg ions (Mg2+) may be able to alleviate the high-fat diet (HFD)-induced bone loss and reduce the side-effects of statin. This study aimed to explore the feasibility of combined Mg2+ with simvastatin (SIM) for treating HFD-induced bone loss in mice and the involving mechanisms. Materials and methods C57BL/6 male mice were fed with a HFD or a normal-fat diet (NFD). Mice were intraperitoneally injected SIM and/or orally received water with additional Mg2+ until sacrificed. Enzyme-linked immunosorbent assay was performed to measure cytokines and cholesterol in serum and liver lysates. Bone mineral density (BMD) and microarchitecture were assessed by micro-computed tomography (μCT) in different groups. The adipogenesis in palmitate pre-treated HepG2 cells was performed under various treatments. Results μCT analysis showed that the trabecular bone mass was significantly lower in the HFD-fed group than that in NFD-fed group since week 8. The cortical thickness in HFD-fed group had a significant decrease at week 24, as compared with NFD-fed group. The combination of Mg2+ and SIM significantly attenuated the trabecular bone loss in HFD-fed mice via arresting the osteoclast formation and bone resorption. Besides, such combination also reduced the hepatocytic synthesis of cholesterol and inhibited matrix metallopeptidase 13 (Mmp13) mRNA expression in pre-osteoclasts. Conclusions The combination of Mg2+ and SIM shows a synergistic effect on attenuating the HFD-induced bone loss. Our current formulation may be a cost-effective alternative treatment to be indicated for obesity-related bone loss. High-fat diet-fed mouse has a susceptibility to lower trabecular bone mass as compared with that of normal-fat diet-fed mouse. The combination of Mg2+ and simvastatin attenuates the trabecular bone loss in high-fat diet-fed mice. The combination of Mg2+ and simvastatin reduces the hepatocytic synthesis of cholesterol.
Collapse
|
50
|
Bedair TM, Heo Y, Ryu J, Bedair HM, Park W, Han DK. Biocompatible and functional inorganic magnesium ceramic particles for biomedical applications. Biomater Sci 2021; 9:1903-1923. [PMID: 33506843 DOI: 10.1039/d0bm01934h] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Magnesium ceramics hold promise for numerous biological applications. This review covers the synthesis of magnesium ceramic particles with specific morphologies and potential modification techniques. Magnesium ceramic particles possess multiple characteristics directly applicable to human biology; they are anti-inflammatory, antibacterial, antiviral, and offer anti-cancer effects. Based on these advantages, magnesium hydroxide nanoparticles have been extensively utilized across biomedical fields. In a vascular stent, the incorporation of magnesium ceramic nanoparticles enhances re-endothelialization. Additionally, tissue regeneration for bone, cartilage, and kidney can be promoted by magnesium ceramics. This review enables researchers to identify the optimum synthetic conditions to prepare magnesium ceramics with specific morphologies and sizes and select the appropriate modification protocols. It is also intended to elucidate the desirable physicochemical properties and biological benefits of magnesium ceramics.
Collapse
Affiliation(s)
- Tarek M Bedair
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi 13488, Korea.
| | | | | | | | | | | |
Collapse
|