1
|
Trbojevic S, Taboas JM, Almarza AJ. TGFβ-1 and Healing of Bone Defects in Large Animal and Rabbit Models: A Systematic Review. Tissue Eng Part A 2024. [PMID: 39723971 DOI: 10.1089/ten.tea.2024.0226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024] Open
Abstract
Long bone and craniofacial bone fractures amount to an overwhelming expenditure for patients and health care systems each year. Overall, 5-10% of all bone fractures result in some form of delayed or nonunion fractures. Nonunions occur from insufficient mechanical stabilization or a compromised wound environment lacking in vasculature and progenitor cells. The current standard for treating these critical-sized fractures and defects is the use of autologous bone grafts. However, advancements in tissue engineering have cultivated a shift in scientific efforts toward harnessing the body's own regenerative resources. As such, research on fracture healing has shifted as well. Transforming growth factor-beta 1 (TGFβ-1) has been studied in fracture healing for over 25 years, though many of these studies have been in vitro or in small animal models. The few studies in large animals have disagreement due to the heterogeneity within the experimental design. Because TGFβ-1 plays such a crucial role in the bone healing process, this systematic review investigates the application of TGFβ-1 in various carrier vehicles for repairing bone injuries in large animal and rabbit models. A systematic search was conducted in PubMed, Embase, and Web of Science (from database construction-October 2024). A total of 244 articles were screened, and 24 studies were included for review. Most large animal long bone studies used coated titanium implants, while most rabbit long bone studies used some form of degradable polymer constructs. TGFβ-1 doses in large animal long bone studies range from 0.005 to 750 µg, doses in large animal calvaria and mandible studies range from 1 to 5000 µg, and doses in rabbit long bone studies range from 0.05 to 120 µg. Nineteen out of 24 articles reviewed indicate successful use of TGFβ-1 for bone regeneration compared with experimental controls. It is clear that dose and controlled release of growth factor play a crucial role in defect closure, but outcome measures and success criteria were inconsistent across studies. More studies with consistent experimental designs are critical for understanding the therapeutic potential of TGFβ-1 in fracture repair, but overall, this review indicates that TGFβ-1 can be used alone or in conjunction with other growth factors to accelerate successful bone repair.
Collapse
Affiliation(s)
- Sara Trbojevic
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Juan M Taboas
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Alejandro J Almarza
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
2
|
Ueyama H, Yamamura M, Koyanagi J, Fukunaga K, Takemura S, Nakamura S. Usefulness of non-surgical treatment without weight bearing restriction versus surgical treatment for maintaining activities of daily living in patients with peri-prosthetic femoral fractures. INTERNATIONAL ORTHOPAEDICS 2024; 48:3227-3234. [PMID: 39320499 DOI: 10.1007/s00264-024-06331-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
PURPOSE Non-surgical treatment without weight-bearing restriction, even in the early post-injury phase, may have a favourable effect on the activities of daily living (ADLs) in elderly patients with peri-prosthetic femoral fractures (PFFs). This study aimed to assess the effectiveness of surgical and non-surgical treatments for PFFs in terms of ADL maintenance and clinical safety. METHODS This retrospective cohort study included 44 patients with PFFs proximal to the stem fixation site without stem loosening. Rehabilitation with weight bearing was initiated after internal fixation of the fracture site in the surgical group (n = 12) and immediately after the injury in the non-surgical group (n = 32). Clinical and radiological outcomes, including time until the first weight-bearing exercise, time until independent walking, ADL deterioration, and bone union rate, were compared between groups. Independent risk factors for ADL deterioration were also evaluated. RESULTS The time until first weight-bearing exercise was shorter and the ADL deterioration rate was smaller in the non-surgical group than in the surgical group (8.8 ± 9.2 vs. 21 ± 13 days, P = 0.004; 6.2% vs. 12.5%, P = 0.04, respectively). Bone union rates were similar between groups (91% vs. 83%, P = 0.42), and aseptic loosening of the stem was not observed. Time until first weight-bearing exercise was identified as an independent risk factor for ADL deterioration (odds ratio, 1.13; 95% confidence interval, 1.01-1.26; P = 0.03). CONCLUSION Non-surgical treatment of PFFs proximal to the stem fixation site without stem loosening, which does not restrict early weight-bearing exercise after injury, is an effective and safe treatment procedure that maintains ADL performance in elderly patients.
Collapse
Affiliation(s)
- Hideki Ueyama
- Department of Orthopedic Surgery, Osaka Rosai Hospital, 1179-3 Nagasonecho, Kitaku, Sakaishi, Osaka, 591-8025, Japan.
| | - Mitsuyoshi Yamamura
- Department of Orthopedic Surgery, Osaka Rosai Hospital, 1179-3 Nagasonecho, Kitaku, Sakaishi, Osaka, 591-8025, Japan
| | - Junichiro Koyanagi
- Department of Orthopedic Surgery, Osaka Rosai Hospital, 1179-3 Nagasonecho, Kitaku, Sakaishi, Osaka, 591-8025, Japan
| | - Kenji Fukunaga
- Department of Orthopedic Surgery, Osaka Rosai Hospital, 1179-3 Nagasonecho, Kitaku, Sakaishi, Osaka, 591-8025, Japan
| | - Susumu Takemura
- Department of Orthopedic Surgery, Osaka Rosai Hospital, 1179-3 Nagasonecho, Kitaku, Sakaishi, Osaka, 591-8025, Japan
| | - Suguru Nakamura
- Department of Orthopedic Surgery, Sano Memorial Hospital, 2-4-28 Nakamachi, Izumisanoshi, Osaka, Japan
| |
Collapse
|
3
|
Taheriazam A, Mir Ahmadi A, Abbaszadeh A, Soleimani M, Darabi R, Samberani M, Poursaleh E. Double plating and iliac crest bone graft can safely fix femoral shaft nonunion. Sci Rep 2024; 14:28988. [PMID: 39578535 PMCID: PMC11584805 DOI: 10.1038/s41598-024-79513-w] [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: 05/26/2024] [Accepted: 11/11/2024] [Indexed: 11/24/2024] Open
Abstract
Introduction Femoral shaft nonunion can be extremely vexing and result in significant morbidity. We aimed to evaluate the outcomes of patients with nonunion of the femoral shaft fracture undergoing double plating and bone grafts from the iliac crest and femoral canal. Methods This retrospective study included 44 patients with femoral shaft nonunion between March 2020 to March 2022. Patients underwent dual plating, bone grafting from the iliac crest, and the utilization of the femoral canal. Age, sex, body mass index (BMI), the two surgical interventions interval, union time after the second surgery, hip range of motion (ROM), limb length discrepancy (LLD), infections, and deep vein thrombosis (DVT) were evaluated. Results Patients comprised 21 males (47.7%) and 23 females (52.3%), with a mean age of 42.3 ± 15.2 years. The mean BMI was 26.7 ± 3.77 kg/m2. The mean duration between the two surgical interventions for the patients was 17.07 ± 6.6 months. The patients were followed for ≥ 12 months. All patients achieved successful union and fracture healing, with an average time of 5 months. The mean hip flexion and extension were 112.84 ± 7.7˚ and 14.8 ± 5.2˚ degrees, respectively. Thirteen patients showed LLD after the first surgery, with a mean LLD of 7.15 ± 5.04 millimeters. LLDs were successfully rectified in all patients following the surgical intervention. No patients experienced postoperative infections or DVT. Conclusion According to our findings, the utilization of double-plate fixation in combination with iliac crest and femoral canal bone graft has proven to be a secure, productive, and straightforward surgical alternative for the management of femoral nonunion.
Collapse
Affiliation(s)
- Afshin Taheriazam
- Department of Orthopedics, School of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - AsraSadat Mir Ahmadi
- Department of Orthopedics, School of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ahmad Abbaszadeh
- Department of Orthopedics, School of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Erfan Niayesh Hospital, No. 17, Imam Hussein Street, Bahar Intersection, Niyayesh West Highway, after Kabiri Tamam Boulevard, Tehran, Iran.
| | - Mohammad Soleimani
- Department of Epidemiology, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Rezvan Darabi
- Department of Orthopedics, School of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehdi Samberani
- Department of Orthopedics, School of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Erfan Poursaleh
- Department of Orthopedics, School of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
4
|
Zhang Z, Wang L, Zhang F, Jing S, Cen M. Functional mechanism and clinical implications of mir-1271-5p in pilon fracture healing processes. J Orthop Surg Res 2024; 19:782. [PMID: 39578827 PMCID: PMC11583746 DOI: 10.1186/s13018-024-05291-w] [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: 09/03/2024] [Accepted: 11/17/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Pilon fractures are challenging to treat and carry a risk of delayed healing. MicroRNA (miRNA) is closely associated with various diseases due to its ability to regulate gene expression. Consequently, this study aimed to examine the connection between miR-1271-5p expression levels and pilon fracture healing processes, while also exploring the underlying mechanisms. The objective of this research was to provide valuable insights for the future clinical treatment of pilon fractures. MATERIALS Venous blood samples were obtained for RNA extraction from patients with normal healing (n = 107) or delayed healing (n = 45) of pilon fractures. The expression levels of miR-1271-5p were measured using qRT-PCR. MiR-1271-5p and ZBTB7A biological functions in MC3T3-E1 cells were examined using the Cell Counting Kit-8 (CCK-8), flow cytometry, and qRT-PCR. Finally, an investigation into the underlying mechanisms was carried out using a dual luciferase reporter assay. RESULTS This study found that, compared to those who healed normally, patients who experienced delayed healing of pilon fractures had significantly higher expression of miR-1271-5p. This suggests that miR-1271-5p may be an indicator for delayed healing in pilon fractures. Moreover, the upregulation of miR-1271-5p may result in a reduction of ZBTB7A expression, which is thought to mediate the effects of miR-1271-5p on MC3T3-E1 cell activities. CONCLUSIONS MiR-1271-5p was involved in the healing processes of pilon fractures via targeting ZBTB7A. MiR-1271-5p was a possible target for the therapy of pilon fractures.
Collapse
Affiliation(s)
- Zhihan Zhang
- Department of Orthopaedic Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Li Wang
- Department of Joint and Trauma Surgery, Shenzhen University General Hospital, Shenzhen, 518055, China
| | - Fangyuan Zhang
- Department of Trauma Surgery, First Hospital of Qinhuangdao, Qinhuangdao, 066000, China
| | - Shaochun Jing
- Department of Traumatic Arthropathy, The Second Hospital of Qinhuangdao, No. 133, Chaoyang South Street, Changli County, Qinhuangdao City, 066000, Hebei Province, China.
| | - Meini Cen
- Department of Rehabilitation Medicine, The Affiliated Hospital of Youjiang Medical University for Nationalities, No.18, Zhongshan 2nd Road, Youjiang District, Baise, 533000, Guangxi, China.
| |
Collapse
|
5
|
Botega II, Guedes PMSG, Ximenez JPB, Zamarioli A, Volpon JB. Refeeding partially reverses impaired fracture callus in undernourished rats. Front Endocrinol (Lausanne) 2024; 15:1385055. [PMID: 39502565 PMCID: PMC11534802 DOI: 10.3389/fendo.2024.1385055] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 10/03/2024] [Indexed: 11/08/2024] Open
Abstract
Background Adequate nutritional intake plays a crucial role in maximizing skeletal acquisition. The specific effects of a general food restriction and refeeding on fracture healing are yet to be fully understood. The aim of this study was to assess the impact of general food restriction and refeeding on fracture repair. Methods Fifty-four male Wistar Hannover rats were randomly assigned into three groups: (1) Sham: Sham rats with femoral fracture; (2) FRes: Food-restricted rats with fracture, (3) Fres+Ref: Fres rats with refeeding. Following weaning, the FRes and Fres+Ref groups received 50% of the food amount provided to the Shams. In the sixth week of the experiment, all animals underwent a mid-right femur bone fracture, which was subsequently surgically stabilized. Following the fracture, the Fres+Ref group was refed, while the other groups maintained their pre-fracture diet. Bone calluses were analyzed on the fifth-day post-fracture by gene expression and on the sixth-week post-fracture using dual-energy X-ray absorptiometry, morphometry, histomorphometry, immunohistochemistry, computed microtomography, and torsion mechanical testing. Statistical significance was determined at a probability level of less than 0.05, and comparisons were made using the ANOVA test. Results Food restriction resulted in significant phenotypic changes in bone calluses when compared to sham rats characterized by deterioration in microstructure (i.e., BV, BV/TV, Tb.N, and Conn.D) reduced collagen deposition, bone mineral density, and mechanical strength (i.e., torque at failure, energy, and stiffness). Moreover, a higher rate of immature bone indicated a decrease in bone callus quality. Refeeding stimulated bone callus collagen formation, reduced local resorption, and effectively restored the microstructural (i.e., SMI, BCa.BV/TV, Tb.Sp, Tb.N, and Conn.D) and mechanical changes (i.e., torque at failure, energy, and angular displacement at failure) caused by food restriction. Despite these positive effects, the density of the bone callus, collagen deposition, and OPG expression remained lower when compared to the shams. Gene expression analysis didn't evidence any significant differences among the groups. Conclusions Food restriction had detrimental effects on osseous healing, which was partially improved by refeeding. Based on these findings, new research can be developed to create targeted nutritional strategies to treat and improve fracture healing.
Collapse
Affiliation(s)
- Iara I. Botega
- Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | | | - João Paulo B. Ximenez
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirao Preto, Brazil
| | - Ariane Zamarioli
- Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - José B. Volpon
- Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| |
Collapse
|
6
|
Al-Ajalein AA, Ibrahim N‘I, Fauzi MB, Mokhtar SA, Naina Mohamed I, Shuid AN, Mohamed N. Evaluating the Anti-Osteoporotic Potential of Mediterranean Medicinal Plants: A Review of Current Evidence. Pharmaceuticals (Basel) 2024; 17:1341. [PMID: 39458982 PMCID: PMC11510337 DOI: 10.3390/ph17101341] [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: 08/05/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Bones are biological reservoirs for minerals and cells, offering protection to the other organs and contributing to the structural form of the body. Osteoporosis is a prevalent bone condition that significantly impacts people's quality of life. Treatments utilizing natural products and medicinal plants have gained important attention in the management of osteoporosis and its associated implications, such as osteoporotic fractures. Even though thousands of plants grow in the Mediterranean region, the use of medicinal plants as an alternative therapy for osteoporosis is still limited. Methods: This article provides a comprehensive overview of seven Mediterranean medicinal plants that are used in osteoporosis and osteoporotic fractures in in vitro, in vivo, and clinical trials. The mechanism of action of the medicinal plants and their bioactive compounds against diseases are also briefly discussed. Results: The findings clearly indicate the ability of the seven medicinal plants (Ammi majus, Brassica oleracea, Ceratonia siliqua L., Foeniculum vulgare, Glycyrrhiza glabra, Salvia officinalis, and Silybum marianum) as anti-osteoporosis agents. Xanthotoxin, polyphenols, liquiritin, formononetin, silymarin, and silibinin/silybin were the main bioactive compounds that contributed to the action against osteoporosis and osteoporotic fractures. Conclusions: In this review, the Mediterranean medicinal plants prove their ability as an alternative agent for osteoporosis and osteoporotic fractures instead of conventional synthetic therapies. Thus, this can encourage researchers to delve deeper into this field and develop medicinal-plant-based drugs.
Collapse
Affiliation(s)
- Alhareth Abdulraheem Al-Ajalein
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.A.-A.); (N.‘I.I.); (I.N.M.)
| | - Nurul ‘Izzah Ibrahim
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.A.-A.); (N.‘I.I.); (I.N.M.)
| | - Mh Busra Fauzi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia;
- Advance Bioactive Materials-Cells (Adv-BioMaC) UKM Research Group, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia
| | - Sabarul Afian Mokhtar
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Isa Naina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.A.-A.); (N.‘I.I.); (I.N.M.)
| | - Ahmad Nazrun Shuid
- Department of Pharmacology, Faculty of Medicine, Universiti Teknologi Mara (UITM), Jalan Hospital, Sungai Buloh 47000, Malaysia;
| | - Norazlina Mohamed
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia; (A.A.A.-A.); (N.‘I.I.); (I.N.M.)
| |
Collapse
|
7
|
Wang D, Liu S, He G, Zhang Z, Li J, Zhang R, Shi J, Jia Y, Qiao H, Liu H, Wang B, Qin S, Zhang Y. Evaluating Docking Site Local Hematoma Formation and Blood Flow on its Healing Using the Accordion Technique at the End of Tibial Bone Transport. Orthop Surg 2024; 16:2264-2272. [PMID: 39556438 PMCID: PMC11572575 DOI: 10.1111/os.14234] [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: 06/09/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 11/19/2024] Open
Abstract
OBJECTIVE At present, due to the lack of early observation methods, the effect of the 'accordion' technique on the treatment of nonunion of the docking site varies greatly. In this study, color Doppler ultrasound was used to observe the docking site's local changes and investigate the relationship between local microenvironment changes and bone healing after the accordion technique. METHODS 30 patients with tibial bone transport treated at the Department of Orthopedics, Second Hospital of Shanxi Medical University, from May 2018 to June 2022, were analyzed retrospectively. Paired-sample t-tests were used for data that conformed to a normal distribution, and paired rank-sum tests were used for before-and-after comparisons that did not conform to a normal distribution. There were 26 males and 4 females, aged 47.3 ± 11.7 years. Before bone transport, the defect gap between tibial bone ends was 6.80 ± 3.61 cm. The steps of the accordion technique were as follows: compression for 7 days, ultrasonic study of the microenvironment at the docking site, distraction for 12 days, latency for 7 days, compression for 14 days, then static fixation and radiological study until complete bone healing. Ultrasound was used to detect the size of the hematoma after 7 days of pressure, and the changes in blood flow before and after the 'accordion' operation. RESULTS All patients were followed up for 11.9 ± 1.9 months. At the last follow-up, 22 patients achieved bone healing at the docking site after the treatment of the 'accordion' technique. There was a linear negative correlation between the size of the hematoma and the time of bone healing at the docking site (r = -0.639, p < 0.01). According to the Paley healing criteria, 18 of the 22 patients were excellent, and 4 patients were good. CONCLUSION Hematoma is necessary for the 'accordion' technique's success in the treatment of nonunion. The size of the hematoma is negatively related to the time of bone healing. The 'accordion' technique can increase the blood flow of tissue around the docking site. Ultrasound can be used to monitor the changes in the microenvironment at the docking site during the 'accordion' technique and guide the exact plan and prognosis of the 'accordion' technique.
Collapse
Affiliation(s)
- Dong Wang
- Shanxi Medical UniversityTaiyuanChina
- The Second Hospital of Shanxi Medical UniversityTaiyuanChina
| | | | - Guo‐Yu He
- Shanxi Medical UniversityTaiyuanChina
| | - Ze Zhang
- Shanxi Medical UniversityTaiyuanChina
| | - Juan Li
- The Second Hospital of Shanxi Medical UniversityTaiyuanChina
| | | | - Jun‐Jun Shi
- The Second Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Ying‐Wei Jia
- The Second Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Hu‐Yun Qiao
- The Second Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Hong Liu
- The Second Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Bao‐Na Wang
- The Second Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Si‐He Qin
- Rehabilitation Hospital Affiliated to National Reserach Center for Rehabilitation Technical AidsBeijingChina
| | - Yong‐Hong Zhang
- The Second Hospital of Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
8
|
Yang Q, Xu M, Fang H, Gao Y, Zhu D, Wang J, Chen Y. Targeting micromotion for mimicking natural bone healing by using NIPAM/Nb 2C hydrogel. Bioact Mater 2024; 39:41-58. [PMID: 38800718 PMCID: PMC11127186 DOI: 10.1016/j.bioactmat.2024.05.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024] Open
Abstract
Natural fracture healing is most efficient when the fine-tuned mechanical force and proper micromotion are applied. To mimick this micromotion at the fracture gap, a near-infrared-II (NIR-II)-activated hydrogel was fabricated by integrating two-dimensional (2D) monolayer Nb2C nanosheets into a thermally responsive poly(N-isopropylacrylamide) (NIPAM) hydrogel system. NIR-II-triggered deformation of the NIPAM/Nb2C hydrogel was designed to generate precise micromotion for co-culturing cells. It was validated that micromotion at 1/300 Hz, triggering a 2.37-fold change in the cell length/diameter ratio, is the most favorable condition for the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). Moreover, mRNA sequencing and verification revealed that micromotion-induced augmentation was mediated by Piezo1 activation. Suppression of Piezo1 interrupts the mechano-sensitivity and abrogates osteogenic differentiation. Calvarial and femoral shaft defect models were established to explore the biocompatibility and osteoinductivity of the Micromotion Biomaterial. A series of research methods, including radiography, micro-CT scanning, and immunohistochemical staining have been performed to evaluate biosafety and osteogenic efficacy. The in vivo results revealed that tunable micromotion strengthens the natural fracture healing process through the sequential activation of endochondral ossification, promotion of neovascularization, initiation of mineral deposition, and combinatory acceleration of full-thickness osseous regeneration. This study demonstrated that Micromotion Biomaterials with controllable mechanophysical characteristics could promote the osteogenic differentiation of BMSCs and facilitate full osseous regeneration. The design of NIPAM/Nb2C hydrogel with highly efficient photothermal conversion, specific features of precisely controlled micromotion, and bionic-mimicking bone-repair capabilities could spark a new era in the field of regenerative medicine.
Collapse
Affiliation(s)
- Qianhao Yang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Mengqiao Xu
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Haoyu Fang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Daoyu Zhu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Jing Wang
- Eye Institute and Department of Ophthalmology, Eye and ENT Hospital, Fudan University, Shanghai, 200031, China
| | - Yixuan Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
9
|
Chandran M, Akesson KE, Javaid MK, Harvey N, Blank RD, Brandi ML, Chevalley T, Cinelli P, Cooper C, Lems W, Lyritis GP, Makras P, Paccou J, Pierroz DD, Sosa M, Thomas T, Silverman S. Impact of osteoporosis and osteoporosis medications on fracture healing: a narrative review. Osteoporos Int 2024; 35:1337-1358. [PMID: 38587674 PMCID: PMC11282157 DOI: 10.1007/s00198-024-07059-8] [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: 10/10/2023] [Accepted: 03/06/2024] [Indexed: 04/09/2024]
Abstract
Antiresorptive medications do not negatively affect fracture healing in humans. Teriparatide may decrease time to fracture healing. Romosozumab has not shown a beneficial effect on human fracture healing. BACKGROUND Fracture healing is a complex process. Uncertainty exists over the influence of osteoporosis and the medications used to treat it on fracture healing. METHODS Narrative review authored by the members of the Fracture Working Group of the Committee of Scientific Advisors of the International Osteoporosis Foundation (IOF), on behalf of the IOF and the Société Internationale de Chirurgie Orthopédique et de Traumatologie (SICOT). RESULTS Fracture healing is a multistep process. Most fractures heal through a combination of intramembranous and endochondral ossification. Radiographic imaging is important for evaluating fracture healing and for detecting delayed or non-union. The presence of callus formation, bridging trabeculae, and a decrease in the size of the fracture line over time are indicative of healing. Imaging must be combined with clinical parameters and patient-reported outcomes. Animal data support a negative effect of osteoporosis on fracture healing; however, clinical data do not appear to corroborate with this. Evidence does not support a delay in the initiation of antiresorptive therapy following acute fragility fractures. There is no reason for suspension of osteoporosis medication at the time of fracture if the person is already on treatment. Teriparatide treatment may shorten fracture healing time at certain sites such as distal radius; however, it does not prevent non-union or influence union rate. The positive effect on fracture healing that romosozumab has demonstrated in animals has not been observed in humans. CONCLUSION Overall, there appears to be no deleterious effect of osteoporosis medications on fracture healing. The benefit of treating osteoporosis and the urgent necessity to mitigate imminent refracture risk after a fracture should be given prime consideration. It is imperative that new radiological and biological markers of fracture healing be identified. It is also important to synthesize clinical and basic science methodologies to assess fracture healing, so that a convergence of the two frameworks can be achieved.
Collapse
Affiliation(s)
- M Chandran
- Osteoporosis and Bone Metabolism Unit, Department of Endocrinology, Singapore General Hospital, DUKE NUS Medical School, Singapore, Singapore.
| | - K E Akesson
- Clinical and Molecular Osteoporosis Research Unit, Department of Clinical Sciences, Lund University, Department of Orthopedics, Skåne University Hospital, Malmö, Sweden
| | - M K Javaid
- NIHR Musculoskeletal Biomedical Research Unit, University of Oxford, Oxford, UK
| | - N Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - R D Blank
- Garvan Institute of Medical Research, Medical College of Wisconsin, Darlinghurst, NSW, Australia
- Medical College of Wisconsin, Milwaukee, WI, USA
| | - M L Brandi
- Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Largo Palagi 1, Florence, Italy
| | - T Chevalley
- Division of Bone Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - P Cinelli
- Department of Trauma Surgery, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - C Cooper
- MRC Lifecourse Epidemiology Centre, University of Southampton, NIHR Southampton Biomedical Research Centre, University of Southampton, University Hospitals Southampton NHS Foundation Trust, Southampton, UK
- NIHR Oxford Biomedical Research Unit, University of Oxford, Oxford, UK
| | - W Lems
- Department of Rheumatology, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - G P Lyritis
- Hellenic Osteoporosis Foundation, Athens, Greece
| | - P Makras
- Department of Medical Research, 251 Hellenic Air Force & VA General Hospital, Athens, Greece
| | - J Paccou
- Department of Rheumatology, MABlab ULR 4490, CHU Lille, Univ. Lille, 59000, Lille, France
| | - D D Pierroz
- International Osteoporosis Foundation, Nyon, Switzerland
| | - M Sosa
- University of Las Palmas de Gran Canaria, Investigation Group on Osteoporosis and Mineral Metabolism, Canary Islands, Spain
| | - T Thomas
- Department of Rheumatology, North Hospital, CHU Saint-Etienne and INSERM U1059, University of Lyon-University Jean Monnet, Saint‑Etienne, France
| | - S Silverman
- Cedars-Sinai Medical Center and Geffen School of Medicine UCLA, Los Angeles, CA, USA
| |
Collapse
|
10
|
van Brakel F, Zhao Y, van der Eerden BC. Fueling recovery: The importance of energy coupling between angiogenesis and osteogenesis during fracture healing. Bone Rep 2024; 21:101757. [PMID: 38577251 PMCID: PMC10990718 DOI: 10.1016/j.bonr.2024.101757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/20/2024] [Accepted: 03/23/2024] [Indexed: 04/06/2024] Open
Abstract
Approximately half of bone fractures that do not heal properly (non-union) can be accounted to insufficient angiogenesis. The processes of angiogenesis and osteogenesis are spatiotemporally regulated in the complex process of fracture healing that requires a substantial amount of energy. It is thought that a metabolic coupling between angiogenesis and osteogenesis is essential for successful healing. However, how this coupling is achieved remains to be largely elucidated. Here, we will discuss the most recent evidence from literature pointing towards a metabolic coupling between angiogenesis and osteogenesis. We will describe the metabolic profiles of the cell types involved during fracture healing as well as secreted products in the bone microenvironment (such as lactate and nitric oxide) as possible key players in this metabolic crosstalk.
Collapse
Affiliation(s)
- Fleur van Brakel
- Calcium and Bone Metabolism Laboratory, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Yudong Zhao
- Calcium and Bone Metabolism Laboratory, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Bram C.J. van der Eerden
- Calcium and Bone Metabolism Laboratory, Department of Internal Medicine, Erasmus MC, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
11
|
Ehlen QT, Mirsky NA, Slavin BV, Parra M, Nayak VV, Cronstein B, Witek L, Coelho PG. Translational Experimental Basis of Indirect Adenosine Receptor Agonist Stimulation for Bone Regeneration: A Review. Int J Mol Sci 2024; 25:6104. [PMID: 38892291 PMCID: PMC11172580 DOI: 10.3390/ijms25116104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Bone regeneration remains a significant clinical challenge, often necessitating surgical approaches when healing bone defects and fracture nonunions. Within this context, the modulation of adenosine signaling pathways has emerged as a promising therapeutic option, encouraging osteoblast activation and tempering osteoclast differentiation. A literature review of the PubMed database with relevant keywords was conducted. The search criteria involved in vitro or in vivo models, with clear methodological descriptions. Only studies that included the use of indirect adenosine agonists, looking at the effects of bone regeneration, were considered relevant according to the eligibility criteria. A total of 29 articles were identified which met the inclusion and exclusion criteria, and they were reviewed to highlight the preclinical translation of adenosine agonists. While preclinical studies demonstrate the therapeutic potential of adenosine signaling in bone regeneration, its clinical application remains unrealized, underscoring the need for further clinical trials. To date, only large, preclinical animal models using indirect adenosine agonists have been successful in stimulating bone regeneration. The adenosine receptors (A1, A2A, A2B, and A3) stimulate various pathways, inducing different cellular responses. Specifically, indirect adenosine agonists act to increase the extracellular concentration of adenosine, subsequently agonizing the respective adenosine receptors. The agonism of each receptor is dependent on its expression on the cell surface, the extracellular concentration of adenosine, and its affinity for adenosine. This comprehensive review analyzed the multitude of indirect agonists currently being studied preclinically for bone regeneration, discussing the mechanisms of each agonist, their cellular responses in vitro, and their effects on bone formation in vivo.
Collapse
Affiliation(s)
- Quinn T. Ehlen
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | - Blaire V. Slavin
- University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Marcelo Parra
- Center of Excellence in Morphological and Surgical Studies (CEMyQ), Faculty of Medicine, Universidad de la Frontera, Temuco 4811230, Chile
- Department of Comprehensive Adult Dentistry, Faculty of Dentistry, Universidad de la Frontera, Temuco 4811230, Chile
| | - Vasudev Vivekanand Nayak
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Bruce Cronstein
- Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Lukasz Witek
- Biomaterials Division, NYU Dentistry, New York, NY 10010, USA
- Department of Biomedical Engineering, New York University Tandon School of Engineering, Brooklyn, NY 11201, USA
- Hansjörg Wyss Department of Plastic Surgery, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Paulo G. Coelho
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- DeWitt Daughtry Family Department of Surgery, Division of Plastic Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
12
|
Kale P, Shrivastava S, Balusani P, Pundkar A. Therapeutic Potential of Platelet-Rich Plasma in Fracture Healing: A Comprehensive Review. Cureus 2024; 16:e62271. [PMID: 39006629 PMCID: PMC11245593 DOI: 10.7759/cureus.62271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 06/12/2024] [Indexed: 07/16/2024] Open
Abstract
Fracture healing is a dynamic process essential for the restoration of bone integrity and function. However, factors such as patient age, comorbidities, and the severity of the fracture can impede this process, leading to delayed healing or nonunion. Platelet-rich plasma (PRP) has emerged as a promising therapeutic option for enhancing fracture healing. PRP is an autologous blood product containing a concentrated mixture of platelets, growth factors, and cytokines known to promote tissue regeneration and repair. This comprehensive review provides an overview of the fracture healing process, emphasizing the importance of timely and efficient bone repair. We discuss the mechanisms underlying the purported efficacy of PRP in fracture healing, drawing upon both preclinical and clinical evidence. Preclinical studies in animal models have demonstrated the ability of PRP to accelerate fracture healing, stimulate osteogenesis, and enhance bone regeneration. Clinical studies have yielded mixed results, with some reporting positive outcomes in terms of accelerated healing and improved functional outcomes, while others have shown no significant benefits over standard treatments. Factors influencing the efficacy of PRP, such as timing of administration, PRP concentration, and patient-specific variables, are also examined. Furthermore, safety considerations and potential adverse effects associated with PRP therapy are discussed. Despite the promising preclinical findings, challenges remain in standardizing PRP formulations, optimizing administration protocols, and addressing unanswered questions regarding its long-term efficacy and safety. This review aims to provide insights into the therapeutic potential of PRP in fracture healing, informing future research directions and guiding clinical practice.
Collapse
Affiliation(s)
- Prathamesh Kale
- Orthopaedic Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sandeep Shrivastava
- Orthopaedic Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Prashanth Balusani
- Orthopaedic Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Aditya Pundkar
- Orthopaedic Surgery, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
13
|
Vampertzis T, Barmpagianni C, Bekiari C, Brellou GD, Zervos IA, Tsiridis E, Galanis N. The Role of Botulinum Neurotoxin A in the Conservative Treatment of Fractures: An Experimental Study on Rats. ScientificWorldJournal 2024; 2024:7446251. [PMID: 38854678 PMCID: PMC11161265 DOI: 10.1155/2024/7446251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/01/2024] [Accepted: 05/10/2024] [Indexed: 06/11/2024] Open
Abstract
This paper explores the role of botulinum neurotoxin in aiding fracture recovery through temporary muscle paralysis. Specifically, it investigates the effects of botulinum neurotoxin-induced paralysis of the sternocleidomastoid muscle on clavicle fractures in rats. The research aims to assess safety, effectiveness, and the impact on fracture healing. Healthy male Albino Wistar rats were divided into four groups: clavicle fracture, botulinum neurotoxin injection, both, and control. Surgeries were conducted under anaesthesia, and postoperatively, animals were monitored for 28 days. Euthanasia and radiological assessment followed, examining fracture healing and muscle changes, while tissues were histopathologically evaluated. The modified Lane-Sandhu scoring system was used for the radiographic evaluation of clavicle fractures, and the results varied from complete healing to nonunion. Histopathological examination at 28 days postfracture showed fibrous tissue, mesenchymal cells, and primary callus formation in all groups. Despite varied callus compositions, botulinum neurotoxin administration did not affect clavicle healing, as evidenced by similar scores to the control group. Several studies have explored botulinum neurotoxin applications in fracture recovery. Research suggests its potential to enhance functional recovery in certain types of fractures. Theoretical benefits include managing muscle spasticity, aiding reduction techniques, and preventing nonunion. However, botulinum neurotoxin's transient effect and nonuniversal applications should be considered. The present study found that botulinum toxin had no clear superiority in healing compared to controls, while histological evaluation showed potential adverse effects on muscle tissue. Further research is essential to understand its risk-benefit balance and long-term effects.
Collapse
Affiliation(s)
- Themistoklis Vampertzis
- Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Chryssa Bekiari
- Laboratory of Anatomy and Histology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia D. Brellou
- Department of Pathology, Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis A. Zervos
- Faculty of Veterinary Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleftherios Tsiridis
- Academic Orthopaedic Department, Papageorgiou General Hospital and CORE Laboratory at CIRI-AUTH, AUTH Medical School, Thessaloniki, Greece
| | - Nikiforos Galanis
- Faculty of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
14
|
Bowers KM, Anderson DE. Delayed Union and Nonunion: Current Concepts, Prevention, and Correction: A Review. Bioengineering (Basel) 2024; 11:525. [PMID: 38927761 PMCID: PMC11201148 DOI: 10.3390/bioengineering11060525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 06/28/2024] Open
Abstract
Surgical management of fractures has advanced with the incorporation of advanced technology, surgical techniques, and regenerative therapies, but delayed bone healing remains a clinical challenge and the prevalence of long bone nonunion ranges from 10 to 15% of surgically managed fractures. Delayed bone healing arises from a combination of mechanical, biological, and systemic factors acting on the site of tissue remodeling, and careful consideration of each case's injury-related, patient-dependent, surgical, and mechanical risk factors is key to successful bone union. In this review, we describe the biology and biomechanics of delayed bone healing, outline the known risk factors for nonunion development, and introduce modern preventative and corrective therapies targeting fracture nonunion.
Collapse
Affiliation(s)
| | - David E. Anderson
- Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, 2407 River Dr., Knoxville, TN 37996-4550, USA;
| |
Collapse
|
15
|
Nie R, Zhang QY, Feng ZY, Huang K, Zou CY, Fan MH, Zhang YQ, Zhang JY, Li-Ling J, Tan B, Xie HQ. Hydrogel-based immunoregulation of macrophages for tissue repair and regeneration. Int J Biol Macromol 2024; 268:131643. [PMID: 38643918 DOI: 10.1016/j.ijbiomac.2024.131643] [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: 12/13/2023] [Revised: 04/10/2024] [Accepted: 04/14/2024] [Indexed: 04/23/2024]
Abstract
The rational design of hydrogel materials to modulate the immune microenvironment has emerged as a pivotal approach in expediting tissue repair and regeneration. Within the immune microenvironment, an array of immune cells exists, with macrophages gaining prominence in the field of tissue repair and regeneration due to their roles in cytokine regulation to promote regeneration, maintain tissue homeostasis, and facilitate repair. Macrophages can be categorized into two types: classically activated M1 (pro-inflammatory) and alternatively activated M2 (anti-inflammatory and pro-repair). By regulating the physical and chemical properties of hydrogels, the phenotypic transformation and cell behavior of macrophages can be effectively controlled, thereby promoting tissue regeneration and repair. A full understanding of the interaction between hydrogels and macrophages can provide new ideas and methods for future tissue engineering and clinical treatment. Therefore, this paper reviews the effects of hydrogel components, hardness, pore size, and surface morphology on cell behaviors such as macrophage proliferation, migration, and phenotypic polarization, and explores the application of hydrogels based on macrophage immune regulation in skin, bone, cartilage, and nerve tissue repair. Finally, the challenges and future prospects of macrophage-based immunomodulatory hydrogels are discussed.
Collapse
Affiliation(s)
- Rong Nie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Qing-Yi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Zi-Yuan Feng
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Kai Huang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Chen-Yu Zou
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ming-Hui Fan
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yue-Qi Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ji-Ye Zhang
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jesse Li-Ling
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Department of Medical Genetics, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Bo Tan
- Department of Orthopedic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, PR China
| | - Hui-Qi Xie
- Department of Orthopedic Surgery and Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China; Frontier Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan 610212, PR China.
| |
Collapse
|
16
|
Anup A, Dieterich S, Oreffo ROC, Dailey HL, Lang A, Haffner-Luntzer M, Hixon KR. Embracing ethical research: Implementing the 3R principles into fracture healing research for sustainable scientific progress. J Orthop Res 2024; 42:568-577. [PMID: 38124294 DOI: 10.1002/jor.25741] [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: 08/17/2023] [Revised: 10/26/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023]
Abstract
As scientific advancements continue to reshape the world, it becomes increasingly crucial to uphold ethical standards and minimize the potentially adverse impact of research activities. In this context, the implementation of the 3R principles-Replacement, Reduction, and Refinement-has emerged as a prominent framework for promoting ethical research practices in the use of animals. This article aims to explore recent advances in integrating the 3R principles into fracture healing research, highlighting their potential to enhance animal welfare, scientific validity, and societal trust. The review focuses on in vitro, in silico, ex vivo, and refined in vivo methods, which have the potential to replace, reduce, and refine animal experiments in musculoskeletal, bone, and fracture healing research. Here, we review material that was presented at the workshop "Implementing 3R Principles into Fracture Healing Research" at the 2023 Orthopedic Research Society (ORS) Annual Meeting in Dallas, Texas.
Collapse
Affiliation(s)
- Amritha Anup
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
| | - Sandra Dieterich
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Hannah L Dailey
- Departments of Orthopaedic Surgery and Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Annemarie Lang
- Department of Mechanical Engineering and Mechanics, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Melanie Haffner-Luntzer
- Institute of Orthopaedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Katherine R Hixon
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire, USA
- Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| |
Collapse
|
17
|
Li B, Thebault P, Labat B, Ladam G, Alt V, Rupp M, Brochausen C, Jantsch J, Ip M, Zhang N, Cheung WH, Leung SYS, Wong RMY. Implants coating strategies for antibacterial treatment in fracture and defect models: A systematic review of animal studies. J Orthop Translat 2024; 45:24-35. [PMID: 38495742 PMCID: PMC10943307 DOI: 10.1016/j.jot.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 03/19/2024] Open
Abstract
Objective Fracture-related infection (FRI) remains a major concern in orthopaedic trauma. Functionalizing implants with antibacterial coatings are a promising strategy in mitigating FRI. Numerous implant coatings have been reported but the preventive and therapeutic effects vary. This systematic review aimed to provide a comprehensive overview of current implant coating strategies to prevent and treat FRI in animal fracture and bone defect models. Methods A literature search was performed in three databases: PubMed, Web of Science and Embase, with predetermined keywords and criteria up to 28 February 2023. Preclinical studies on implant coatings in animal fracture or defect models that assessed antibacterial and bone healing effects were included. Results A total of 14 studies were included in this systematic review, seven of which used fracture models and seven used defect models. Passive coatings with bacteria adhesion resistance were investigated in two studies. Active coatings with bactericidal effects were investigated in 12 studies, four of which used metal ions including Ag+ and Cu2+; five studies used antibiotics including chlorhexidine, tigecycline, vancomycin, and gentamicin sulfate; and the other three studies used natural antibacterial materials including chitosan, antimicrobial peptides, and lysostaphin. Overall, these implant coatings exhibited promising efficacy in antibacterial effects and bone formation. Conclusion Antibacterial coating strategies reduced bacterial infections in animal models and favored bone healing in vivo. Future studies of implant coatings should focus on optimal biocompatibility, antibacterial effects against multi-drug resistant bacteria and polymicrobial infections, and osseointegration and osteogenesis promotion especially in osteoporotic bone by constructing multi-functional coatings for FRI therapy. The translational potential of this paper The clinical treatment of FRI is complex and challenging. This review summarizes novel orthopaedic implant coating strategies applied to FRI in preclinical studies, and offers a perspective on the future development of orthopaedic implant coatings, which can potentially contribute to alternative strategies in clinical practice.
Collapse
Affiliation(s)
- Baoqi Li
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Pascal Thebault
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000, Rouen, France
| | - Béatrice Labat
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000, Rouen, France
| | - Guy Ladam
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, F-76000, Rouen, France
| | - Volker Alt
- Department of Trauma Surgery, University Hospital Regensburg, Germany
| | - Markus Rupp
- Department of Trauma Surgery, University Hospital Regensburg, Germany
| | | | - Jonathan Jantsch
- Institute for Medical Microbiology, Immunology, and Hygiene, and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Margaret Ip
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ning Zhang
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Wing-Hoi Cheung
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - Ronald Man Yeung Wong
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
18
|
Ryskalin L, Fulceri F, Morucci G, Dell’Agli S, Soldani P, Gesi M. Treatment of delayed union of the forearm with extracorporeal shockwave therapy: a case report and literature review. Front Endocrinol (Lausanne) 2023; 14:1286480. [PMID: 38033992 PMCID: PMC10684947 DOI: 10.3389/fendo.2023.1286480] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/25/2023] [Indexed: 12/02/2023] Open
Abstract
Compared to other long bones, forearm fractures are particularly challenging due to the high rate of complications. These include malunion, delayed/nonunion, wrist and elbow movement reduction, and pain. Surgical procedure is considered the gold standard for managing delayed union and nonunion of the long bones. However, in the last decades, extracorporeal shockwave therapy (ESWT) has emerged as an effective and less invasive approach to enhance bone regeneration and fracture healing, avoiding major complications of surgical procedures. In contrast to the broad literature reporting good clinical results of ESWT in the treatment of nonunions, there is currently limited evidence regarding the clinical application of shock waves on long bone delayed fractures, particularly those of the forearm. In the present paper, we report a case of delayed bone healing of the diaphyseal region of the ulna treated with focused ESWT. The successful case experienced bone healing at the fracture site in less than 3 months after initial ESWT treatment. Acknowledging the limitation of reporting a case report, however, the remarkable clinical results and the absence of side effects contribute valuable information in support of the use of ESWT as an effective alternative to standard surgery for forearm fractures.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Center for Rehabilitative Medicine “Sport and Anatomy”, University of Pisa, Pisa, Italy
| | | | - Gabriele Morucci
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Center for Rehabilitative Medicine “Sport and Anatomy”, University of Pisa, Pisa, Italy
| | - Stefania Dell’Agli
- Center for Rehabilitative Medicine “Sport and Anatomy”, University of Pisa, Pisa, Italy
| | - Paola Soldani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Center for Rehabilitative Medicine “Sport and Anatomy”, University of Pisa, Pisa, Italy
| | - Marco Gesi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- Center for Rehabilitative Medicine “Sport and Anatomy”, University of Pisa, Pisa, Italy
| |
Collapse
|
19
|
Ren Y, Zhang S, Weeks J, Rangel-Moreno J, He B, Xue T, Rainbolt J, Morita Y, Shu Y, Liu Y, Kates SL, Schwarz EM, Xie C. Reduced angiogenesis and delayed endochondral ossification in CD163 -/- mice highlights a role of M2 macrophages during bone fracture repair. J Orthop Res 2023; 41:2384-2393. [PMID: 36970754 PMCID: PMC10522791 DOI: 10.1002/jor.25564] [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: 10/28/2022] [Revised: 02/24/2023] [Accepted: 03/24/2023] [Indexed: 04/10/2023]
Abstract
While recent studies showed that macrophages are critical for bone fracture healing, and lack of M2 macrophages have been implicated in models of delayed union, functional roles for specific M2 receptors have yet to be defined. Moreover, the M2 scavenger receptor CD163 has been identified as a target to inhibit sepsis following implant-associated osteomyelitis, but potential adverse effects on bone healing during blockage therapy have yet to be explored. Thus, we investigated fracture healing in C57BL/6 versus CD163-/- mice using a well-established closed, stabilized, mid-diaphyseal femur fracture model. While gross fracture healing in CD163-/- mice was similar to that of C57BL/6, plain radiographs revealed persistent fracture gaps in the mutant mice on Day 14, which resolved by Day 21. Consistently, 3D vascular micro-CT demonstrated delayed union on Day 21, with reduced bone volume (74%, 61%, and 49%) and vasculature (40%, 40%, and 18%) compared to C57BL/6 on Days 10, 14, and 21 postfracture, respectively (p < 0.01). Histology confirmed large amounts of persistent cartilage in CD163-/- versus C57BL/6 fracture callus on Days 7 and 10 that resolves over time, and immunohistochemistry demonstrated deficiencies in CD206+ M2 macrophages. Torsion testing of the fractures confirmed the delayed early union in CD163-/- femurs, which display decreased yield torque on Day 21, and a decreased rigidity with a commensurate increase in rotation at yield on Day 28 (p < 0.01). Collectively, these results demonstrate that CD163 is required for normal angiogenesis, callus formation, and bone remodeling during fracture healing, and raise potential concerns about CD163 blockade therapy.
Collapse
Affiliation(s)
- Youliang Ren
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Shiyang Zhang
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Jason Weeks
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Javier Rangel-Moreno
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Bin He
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas Xue
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Joshua Rainbolt
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Yugo Morita
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Ye Shu
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Yuting Liu
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Stephen L. Kates
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA, USA
| | - Edward M. Schwarz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Chao Xie
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
- Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
20
|
Ballout N, Boullier A, Darwiche W, Ait-Mohand K, Trécherel E, Gallégo T, Gomila C, Yaker L, Gennero I, Kovensky J, Ausseil J, Toumieux S. DP2, a Carbohydrate Derivative, Enhances In Vitro Osteoblast Mineralisation. Pharmaceuticals (Basel) 2023; 16:1512. [PMID: 38004380 PMCID: PMC10674337 DOI: 10.3390/ph16111512] [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: 09/18/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 11/26/2023] Open
Abstract
Bone fracture healing is a complex biological process involving four phases coordinated over time: hematoma formation, granulation tissue formation, bony callus formation, and bone remodelling. Bone fractures represent a significant health problem, particularly among the elderly population and patients with comorbidities. Therapeutic strategies proposed to treat such fractures include the use of autografts, allografts, and tissue engineering strategies. It has been shown that bone morphogenetic protein 2 (BMP-2) has a therapeutic potential to enhance fracture healing. Despite the clinical efficacy of BMP-2 in osteoinduction and bone repair, adverse side effects and complications have been reported. Therefore, in this in vitro study, we propose the use of a disaccharide compound (DP2) to improve the mineralisation process. We first evaluated the effect of DP2 on primary human osteoblasts (HOb), and then investigated the mechanisms involved. Our findings showed that (i) DP2 improved osteoblast differentiation by inducing alkaline phosphatase activity, osteopontin, and osteocalcin expression; (ii) DP2 induced earlier in vitro mineralisation in HOb cells compared to BMP-2 mainly by earlier activation of Runx2; and (iii) DP2 is internalized in HOb cells and activates the protein kinase C signalling pathway. Consequently, DP2 is a potential therapeutical candidate molecule for bone fracture repair.
Collapse
Affiliation(s)
- Nissrine Ballout
- Société d’Accélération du Transfert de Technologie-Nord, 59800 Lille, France; (N.B.)
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, INSERM UMR1291, CNRS UMR5051, University of Toulouse III, 31024 Toulouse, France
- Service de Biochimie, Institut Fédératif de Biologie, CHU Toulouse, 31024 Toulouse, France
| | - Agnès Boullier
- Mécanismes Physiopathologiques et Conséquences des Calcifications Cardiovasculaires, UR7517, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054 Amiens, France (E.T.)
- Laboratory of Biochemistry, CHU Amiens-Picardie, 80054 Amiens, France
| | - Walaa Darwiche
- Société d’Accélération du Transfert de Technologie-Nord, 59800 Lille, France; (N.B.)
| | - Katia Ait-Mohand
- Laboratoire de Glycochimie et des Agroressources d’Amiens, UR 7378, CNRS, Université de Picardie Jules Verne, 80039 Amiens, France
| | - Eric Trécherel
- Mécanismes Physiopathologiques et Conséquences des Calcifications Cardiovasculaires, UR7517, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054 Amiens, France (E.T.)
| | - Théo Gallégo
- Société d’Accélération du Transfert de Technologie-Nord, 59800 Lille, France; (N.B.)
| | - Cathy Gomila
- Mécanismes Physiopathologiques et Conséquences des Calcifications Cardiovasculaires, UR7517, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054 Amiens, France (E.T.)
| | - Linda Yaker
- Mécanismes Physiopathologiques et Conséquences des Calcifications Cardiovasculaires, UR7517, Centre Universitaire de Recherche en Santé, CURS-UPJV, University of Picardy Jules Verne, 80054 Amiens, France (E.T.)
| | - Isabelle Gennero
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, INSERM UMR1291, CNRS UMR5051, University of Toulouse III, 31024 Toulouse, France
- Service de Biochimie, Institut Fédératif de Biologie, CHU Toulouse, 31024 Toulouse, France
| | - José Kovensky
- Laboratoire de Glycochimie et des Agroressources d’Amiens, UR 7378, CNRS, Université de Picardie Jules Verne, 80039 Amiens, France
| | - Jérôme Ausseil
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, INSERM UMR1291, CNRS UMR5051, University of Toulouse III, 31024 Toulouse, France
- Service de Biochimie, Institut Fédératif de Biologie, CHU Toulouse, 31024 Toulouse, France
| | - Sylvestre Toumieux
- Laboratoire de Glycochimie et des Agroressources d’Amiens, UR 7378, CNRS, Université de Picardie Jules Verne, 80039 Amiens, France
| |
Collapse
|
21
|
Wang Z, Mi F, Li J, Chen D, Lin M, Wang X, Wu S, Wu C, Liu C. Bone Marrow Stromal Cells Sorted by Semiconducting Polymer Nanodots for Bone Repair. ACS Biomater Sci Eng 2023; 9:5772-5781. [PMID: 37734919 DOI: 10.1021/acsbiomaterials.3c00575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/23/2023]
Abstract
The use of bone marrow stromal cells (BMSCs) for bone defect repair has shown great promise due to their differentiation potential. However, isolating the BMSCs from various cell types within the bone marrow remains challenging. To tackle this issue, we utilized semiconducting polymer dots (Pdots) as markers to select the BMSCs within a specific time frame. The therapeutic efficacy of the obtained Pdot-labeled BMSCs was assessed in a bone defect model. Initially, we evaluated the binding capacity of the Pdots with four different types of cells present in the bone marrow including BMSCs, osteoblasts, macrophages, and vascular endothelial cells, in vitro. Notably, BMSCs showed the most rapid uptake of the Pdots, being labeled within only one h of coculture, while other cells took four h to become labeled. Moreover, by colocalizing the Pdots with Prrx1, Sca-1, OSX, F480, and CD105 in the bone marrow cells of monocortical tibial defect (MTD) mice in vivo, we determined the proportions of BMSCs, macrophages, and vascular endothelial cells among all labeled cells from 1 to 8 h after the Pdots injection. It was found that BMSCs have the highest proportion (92%) among all labeled cells extracted after 1 h of Pdots injection. The therapeutic efficacy of the obtained Pdots-labeled BMSCs (1 h) was assessed in a bone defect model. Results showed that the new bone accrual was significantly increased in the treatment of Pdots-labeled BMSCs compared to the bone marrow cell-treated group. Our study revealed that BMSCs screened by the Pdots could improve bone defect repair, suggesting a promising application of the Pdots in bone healing.
Collapse
Affiliation(s)
- Ziyan Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Feixue Mi
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Jinchen Li
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Dandan Chen
- College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, Shandong 266071, China
| | - Minmin Lin
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Xinyu Wang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Siying Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Changfeng Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Chao Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
- Guangdong Provincial Key Laboratory of Advanced Biomaterials, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
22
|
Kim K, Su Y, Kucine AJ, Cheng K, Zhu D. Guided Bone Regeneration Using Barrier Membrane in Dental Applications. ACS Biomater Sci Eng 2023; 9:5457-5478. [PMID: 37650638 DOI: 10.1021/acsbiomaterials.3c00690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Guided bone regeneration (GBR) is a widely used technique in preclinical and clinical studies due to its predictability. Its main purpose is to prevent the migration of soft tissue into the osseous wound space, while allowing osseous cells to migrate to the site. GBR is classified into two main categories: resorbable and non-resorbable membranes. Resorbable membranes do not require a second surgery but tend to have a short resorption period. Conversely, non-resorbable membranes maintain their mechanical strength and prevent collapse. However, they require removal and are susceptible to membrane exposure. GBR is often used with bone substitute graft materials to fill the defect space and protect the bone graft. The membrane can also undergo various modifications, such as surface modification and biological factor loading, to improve barrier functions and bone regeneration. In addition, bone regeneration is largely related to osteoimmunology, a new field that focuses on the interactions between bone and the immune system. Understanding these interactions can help in developing new treatments for bone diseases and injuries. Overall, GBR has the potential to be a powerful tool in promoting bone regeneration. Further research in this area could lead to advancements in the field of bone healing. This review will highlight resorbable and non-resorbable membranes with cellular responses during bone regeneration, provide insights into immunological response during bone remodeling, and discuss antibacterial features.
Collapse
Affiliation(s)
- Kakyung Kim
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Yingchao Su
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| | - Allan J Kucine
- Department of Oral and Maxillofacial Surgery, Stony Brook University, Stony Brook, New York 11794, United States
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York City, New York 10027, United States
| | - Donghui Zhu
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York 11794, United States
| |
Collapse
|
23
|
Zhou H, Deng K, Wang N, Li H, Xu Z. Staphylococcal enterotoxin C as a novel strategy for treating lumbar spondylolysis in adolescents: Description of technique. Medicine (Baltimore) 2023; 102:e35224. [PMID: 37773848 PMCID: PMC10545280 DOI: 10.1097/md.0000000000035224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 08/24/2023] [Indexed: 10/01/2023] Open
Abstract
Lumbar spondylolysis is one of the most common causes of low back pain and primarily affects children and adolescents. Traditional posterior lumbar fixation and interbody fusion surgery has always been the most effective method to treat spondylolysis. However, traditional surgical management has limitations of large trauma, complex operation, high cost, postoperative biomechanical deterioration, and resulting complications. In order to avoid the trauma and complications of surgical treatment, and reduce the cost of treatment. Based on the successful clinical experience of using staphylococcal enterotoxin C (SEC) to treat nonunion after a limb fracture, we identified a minimally invasive method to effectively treat lumbar spondylolysis. A novel minimally invasive therapeutic approach is presented herein of an SEC injection guided by C-arm fluoroscopy to treat lumbar spondylolysis. We describe a novel technique applied in a patient with lumbar spondylolysis, who showed significantly improved low back pain symptoms and a computed tomography scan, including osseous fusion of the bilateral isthmus at L4 after SEC therapy. This is the first reported case description of using an SEC injection to treat lumbar spondylolysis with a successful clinical outcome.
Collapse
Affiliation(s)
- Hongdian Zhou
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Kaiwen Deng
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Nan Wang
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Hua Li
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Zujian Xu
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
24
|
Yu S, Bd YT, Bd YW, Bd MF, BMed SL, BMed GT, BMed ZY, Miron RJ, Zhang Y, Yang Z, Wang Y. Early tissue and healing responses after maxillary sinus augmentation using horizontal platelet rich fibrin bone blocks. BMC Oral Health 2023; 23:589. [PMID: 37620826 PMCID: PMC10463479 DOI: 10.1186/s12903-023-03228-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] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/14/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND The effects of horizontal platelet-rich fibrin (H-PRF) bone block on the healing and immune response during sinus augmentation have not been fully investigated histologically at early time points. METHODS Eighteenth male New Zealand white rabbits underwent bilateral sinus augmentation and were divided into two groups: deproteinized bovine bone mineral (DBBM) alone and H-PRF + DBBM (H-PRF bone block) group. Maxilla samples were collected at 3, 7 and 14 days post sinus augmentation procedures and analyzed using histological staining for the number of inflammatory cells, new blood vessels and evidence for early osteoclast bone turnover/remodeling. Furthermore, the effects of H-PRF bone blocks on the migration of osteoblasts and THP-1 macrophages were evaluated using a Transwell assay in vitro. RESULTS A higher number of immune cells were found in the H-PRF bone block group at 3 and 7 days post-surgery when compared to the DBBM alone group,most notably in the regions close to the mucosal lining and bone plates. Furthermore, a significantly greater number of new blood vessel formations and early signs of osteoclast development were found in the H-PRF bone block group at 14 days. The in vitro transwell assay further confirmed that culture medium from H-PRF bone block markedly promote the migration of osteoblasts and THP-1 macrophages. CONCLUSIONS The findings from this study have shown that H-PRF bone block is capable of increasing early immune cell infiltration leading to the acceleration of neovascularization and speeding the process of bone metabolism in vivo following maxillary sinus grafting with DBBM.
Collapse
Affiliation(s)
- Shimin Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yinping Tian Bd
- Department of Stomatology, The Central Hospital of Enshi Tujia and Miao Autonomous Perfecture, Enshi, 445000, China
| | - Yan Wei Bd
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Mengge Feng Bd
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Sensen Li BMed
- Department of Stomatology, The Central Hospital of Enshi Tujia and Miao Autonomous Perfecture, Enshi, 445000, China
| | - Guoyong Tong BMed
- Department of Stomatology, The Central Hospital of Enshi Tujia and Miao Autonomous Perfecture, Enshi, 445000, China
| | - Zhouqing Yu BMed
- Department of Stomatology, The Central Hospital of Enshi Tujia and Miao Autonomous Perfecture, Enshi, 445000, China
| | - Richard J Miron
- Department of Periodontology, School of Dental Medicine, University of Bern, Bern, Switzerland
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
- Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Zaibo Yang
- Department of Stomatology, The Central Hospital of Enshi Tujia and Miao Autonomous Perfecture, Enshi, 445000, China.
| | - Yulan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
25
|
Rundle CH, Gomez GA, Pourteymoor S, Mohan S. Sequential application of small molecule therapy enhances chondrogenesis and angiogenesis in murine segmental defect bone repair. J Orthop Res 2023; 41:1471-1481. [PMID: 36448182 PMCID: PMC10506518 DOI: 10.1002/jor.25493] [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: 04/06/2022] [Revised: 10/03/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
The increasing incidence of physiologic/pathologic conditions that impair the otherwise routine healing of endochondral bone fractures and the occurrence of severe bone injuries necessitate novel approaches to enhance clinically challenging bone fracture repair. To promote the healing of nonunion fractures, we tested an approach that used two small molecules to sequentially enhance cartilage development and conversion to the bone in the callus of a murine femoral segmental defect nonunion model of bone injury. Systemic injections of smoothened agonist 21k (SAG21k) were used to stimulate chondrogenesis through the activation of the sonic hedgehog (SHH) pathway early in bone repair, while injections of the prolyl hydroxylase domain (PHD)2 inhibitor, IOX2, were used to stimulate hypoxia signaling-mediated endochondral bone formation. The expression of SHH pathway genes and Phd2 target genes was increased in chondrocyte cell lines in response to SAG21k and IOX2 treatment, respectively. The segmental defect responded to sequential systemic administration of these small molecules with increased chondrocyte expression of PTCH1, GLI1, and SOX9 in response to SAG and increased expression of hypoxia-induced factor-1α and vascular endothelial growth factor-A in the defect tissues in response to IOX2. At 6 weeks postsurgery, the combined SAG-IOX2 therapy produced increased bone formation in the defect with the bony union over the injury. Clinical significance: This therapeutic approach was successful in promoting cartilage and bone formation within a critical-size segmental defect and established the utility of a sequential small molecule therapy for the enhancement of fracture callus development in clinically challenging bone injuries.
Collapse
Affiliation(s)
- Charles H. Rundle
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, California, USA
- Department of Medicine, Loma Linda University, Loma Linda, California, USA
| | - Gustavo A. Gomez
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, California, USA
| | - Sheila Pourteymoor
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, California, USA
| | - Subburaman Mohan
- Musculoskeletal Disease Center, VA Loma Linda Healthcare System, Loma Linda, California, USA
- Department of Medicine, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
26
|
Migliorini F, Cocconi F, Vecchio G, Schäefer L, Koettnitz J, Maffulli N. Pharmacological agents for bone fracture healing: talking points from recent clinical trials. Expert Opin Investig Drugs 2023; 32:855-865. [PMID: 37740660 DOI: 10.1080/13543784.2023.2263352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 09/22/2023] [Indexed: 09/24/2023]
Abstract
INTRODUCTION Pharmacological strategies might influence bone healing in terms of time to union or quality of mature bone. This expert opinion discussed the current level I evidence on the experimental pharmacological agents used to favor bone fracture healing. AREAS COVERED This study was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses: the 2020 PRISMA statement. In April 2023, the following databases were accessed: PubMed, Web of Science, Google Scholar, Embase. All the randomized clinical trials investigating pharmacological agents for bone fracture healing were accessed. No time constraint was set for the search. The search was restricted to RCTs. No additional filters were used in the database search. Data from 19 RCTs (4067 patients) were collected. 78% (3160 of 4067) were women. The mean length of the follow-up was 9.3 months (range, 1-26 months). The mean age of the patients was 64.4 years (range, 8-84 years). EXPERT OPINION Calcitonin could favor bone fracture healing. Bisphosphonates (alendronate, zoledronate, clodronate), monoclonal antibodies (denosumab, romosozumab), statins, vitamin D and calcium supplementation, strontium ranelate, and ibuprofen did not influence bony healing. Concerning the effect of parathormone, current level I evidence is controversial, and additional studies are required. LEVEL OF EVIDENCE Level I, systematic review of RCTs.
Collapse
Affiliation(s)
- Filippo Migliorini
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Medical Centre, Aachen, Germany
| | - Federico Cocconi
- Department of Orthopedics and Trauma Surgery, Academic Hospital of Bolzano (SABES-ASDAA), Teaching Hospital of Paracelsus Medical University, Bolzano, Italy
| | - Gianluca Vecchio
- Department of Trauma and Orthopaedic Surgery, University Hospital Sant' Andrea, University La Sapienza, Rome, Italy
| | - Luise Schäefer
- Department of Orthopaedic, Trauma, and Reconstructive Surgery, RWTH University Medical Centre, Aachen, Germany
| | - Julian Koettnitz
- Department of Orthopedics, Auguste-Viktoria Clinic, Ruhr University Bochum, Bad Oeynhausen, Germany
| | - Nicola Maffulli
- Department of Trauma and Orthopaedic Surgery, University Hospital Sant' Andrea, University La Sapienza, Rome, Italy
- School of Pharmacy and Bioengineering, Keele University Faculty of Medicine, Stoke on Trent, UK
- Centre for Sports and Exercise Medicine, Barts and the London School of Medicine and Dentistry, Mile End Hospital, Queen Mary University of London, London, UK
| |
Collapse
|
27
|
Torrecillas-Baena B, Pulido-Escribano V, Dorado G, Gálvez-Moreno MÁ, Camacho-Cardenosa M, Casado-Díaz A. Clinical Potential of Mesenchymal Stem Cell-Derived Exosomes in Bone Regeneration. J Clin Med 2023; 12:4385. [PMID: 37445420 DOI: 10.3390/jcm12134385] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 06/19/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Bone metabolism is regulated by osteoblasts, osteoclasts, osteocytes, and stem cells. Pathologies such as osteoporosis, osteoarthritis, osteonecrosis, and traumatic fractures require effective treatments that favor bone formation and regeneration. Among these, cell therapy based on mesenchymal stem cells (MSC) has been proposed. MSC are osteoprogenitors, but their regenerative activity depends in part on their paracrine properties. These are mainly mediated by extracellular vesicle (EV) secretion. EV modulates regenerative processes such as inflammation, angiogenesis, cell proliferation, migration, and differentiation. Thus, MSC-EV are currently an important tool for the development of cell-free therapies in regenerative medicine. This review describes the current knowledge of the effects of MSC-EV in the different phases of bone regeneration. MSC-EV has been used by intravenous injection, directly or in combination with different types of biomaterials, in preclinical models of bone diseases. They have shown great clinical potential in regenerative medicine applied to bone. These findings should be confirmed through standardization of protocols, a better understanding of the mechanisms of action, and appropriate clinical trials. All that will allow the translation of such cell-free therapy to human clinic applications.
Collapse
Affiliation(s)
- Bárbara Torrecillas-Baena
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Victoria Pulido-Escribano
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Gabriel Dorado
- Department Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba, 14071 Córdoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), 14004 Córdoba, Spain
| | - María Ángeles Gálvez-Moreno
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Marta Camacho-Cardenosa
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
| | - Antonio Casado-Díaz
- Unidad de Gestión Clínica de Endocrinología y Nutrición-GC17, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofía, 14004 Córdoba, Spain
- CIBER de Fragilidad y Envejecimiento Saludable (CIBERFES), 14004 Córdoba, Spain
| |
Collapse
|
28
|
Rivera KO, Cuylear DL, Duke VR, O’Hara KM, Zhong JX, Elghazali NA, Finbloom JA, Kharbikar BN, Kryger AN, Miclau T, Marcucio RS, Bahney CS, Desai TA. Encapsulation of β-NGF in injectable microrods for localized delivery accelerates endochondral fracture repair. Front Bioeng Biotechnol 2023; 11:1190371. [PMID: 37284244 PMCID: PMC10241161 DOI: 10.3389/fbioe.2023.1190371] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/02/2023] [Indexed: 06/08/2023] Open
Abstract
Introduction: Currently, there are no non-surgical FDA-approved biological approaches to accelerate fracture repair. Injectable therapies designed to stimulate bone healing represent an exciting alternative to surgically implanted biologics, however, the translation of effective osteoinductive therapies remains challenging due to the need for safe and effective drug delivery. Hydrogel-based microparticle platforms may be a clinically relevant solution to create controlled and localized drug delivery to treat bone fractures. Here, we describe poly (ethylene glycol) dimethacrylate (PEGDMA)-based microparticles, in the shape of microrods, loaded with beta nerve growth factor (β-NGF) for the purpose of promoting fracture repair. Methods: Herein, PEGDMA microrods were fabricated through photolithography. PEGDMA microrods were loaded with β-NGF and in vitro release was examined. Subsequently, bioactivity assays were evaluated in vitro using the TF-1 tyrosine receptor kinase A (Trk-A) expressing cell line. Finally, in vivo studies using our well-established murine tibia fracture model were performed and a single injection of the β-NGF loaded PEGDMA microrods, non-loaded PEGDMA microrods, or soluble β-NGF was administered to assess the extent of fracture healing using Micro-computed tomography (µCT) and histomorphometry. Results: In vitro release studies showed there is significant retention of protein within the polymer matrix over 168 hours through physiochemical interactions. Bioactivity of protein post-loading was confirmed with the TF-1 cell line. In vivo studies using our murine tibia fracture model show that PEGDMA microrods injected at the site of fracture remained adjacent to the callus for over 7 days. Importantly, a single injection of β-NGF loaded PEGDMA microrods resulted in improved fracture healing as indicated by a significant increase in the percent bone in the fracture callus, trabecular connective density, and bone mineral density relative to soluble β-NGF control indicating improved drug retention within the tissue. The concomitant decrease in cartilage fraction supports our prior work showing that β-NGF promotes endochondral conversion of cartilage to bone to accelerate healing. Discussion: We demonstrate a novel and translational method wherein β-NGF can be encapsulated within PEGDMA microrods for local delivery and that β-NGF bioactivity is maintained resulting in improved bone fracture repair.
Collapse
Affiliation(s)
- Kevin O. Rivera
- Graduate Program in Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Darnell L. Cuylear
- Graduate Program in Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Victoria R. Duke
- Center for Regenerative and Personalized Medicine, The Steadman Philippon Research Institute (SPRI), Vail, CO, United States
| | - Kelsey M. O’Hara
- Center for Regenerative and Personalized Medicine, The Steadman Philippon Research Institute (SPRI), Vail, CO, United States
| | - Justin X. Zhong
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
- UC Berkeley—UCSF Graduate Program in Bioengineering, San Francisco, CA, United States
| | - Nafisa A. Elghazali
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
- UC Berkeley—UCSF Graduate Program in Bioengineering, San Francisco, CA, United States
| | - Joel A. Finbloom
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Bhushan N. Kharbikar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Alex N. Kryger
- School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Theodore Miclau
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Ralph S. Marcucio
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Chelsea S. Bahney
- Graduate Program in Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Center for Regenerative and Personalized Medicine, The Steadman Philippon Research Institute (SPRI), Vail, CO, United States
- UC Berkeley—UCSF Graduate Program in Bioengineering, San Francisco, CA, United States
| | - Tejal A. Desai
- Graduate Program in Oral and Craniofacial Sciences, School of Dentistry, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco (UCSF), San Francisco, CA, United States
- Department of Bioengineering, University of California, Berkeley (UC Berkeley), Berkeley, CA, United States
- School of Engineering, Brown University, Providence, RI, United States
| |
Collapse
|
29
|
Exosomes Derived from Adipose Stem Cells Enhance Bone Fracture Healing via the Activation of the Wnt3a/β-Catenin Signaling Pathway in Rats with Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24054852. [PMID: 36902283 PMCID: PMC10003369 DOI: 10.3390/ijms24054852] [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: 12/28/2022] [Revised: 02/06/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Nonunion and delayed union are common complications of diabetes mellitus that pose a serious health threat to people. There are many approaches that have been used to improve bone fracture healing. Recently, exosomes have been regarded as promising medical biomaterials for improving fracture healing. However, whether exosomes derived from adipose stem cells can promote bone fracture healing in diabetes mellitus remains unclear. In this study, adipose stem cells (ASCs) and exosomes derived from adipose stem cells (ASCs-exos) are isolated and identified. Additionally, we evaluate the in vitro and in vivo effects of ASCs-exos on the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and bone repair and the regeneration in a rat model of nonunion via Western blotting, immunofluorescence assay, ALP staining, alizarin red staining, radiographic examination and histological analysis. Compared with controls, ASCs-exos promoted BMSC osteogenic differentiation. Additionally, the results of Western blotting, radiographic examination and histological analysis show that ASCs-exos improve the ability for fracture repair in the rat model of nonunion bone fracture healing. Moreover, our results further proved that ASCs-exos play a role in activating the Wnt3a/β-catenin signaling pathway, which facilitates the osteogenic differentiation of BMSCs. All these results show that ASCs-exos enhance the osteogenic potential of BMSCs by activating the Wnt/β-catenin signaling pathway, and also facilitate the ability for bone repair and regeneration in vivo, which provides a novel direction for fracture nonunion in diabetes mellitus treatment.
Collapse
|
30
|
Kurakar M, Joshi U. Comparative Study of Miniplate vs Reconstruction Plate in the Management of Bilateral Parasymphysis Mandible Fracture: FEM Analysis. J Maxillofac Oral Surg 2023; 22:9-17. [PMID: 36703660 PMCID: PMC9871087 DOI: 10.1007/s12663-021-01649-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 09/23/2021] [Indexed: 01/29/2023] Open
Abstract
Aim and Objective The present study compared the stability of fracture fragments in the management of bilateral parasymphysis mandible fracture with Miniplate fixation and Reconstruction plate fixation using finite element analysis. Material and Method 3D FE Mandible model was created using CT scanner. Two bilateral parasymphysis mandible fracture models were created. Model 1 was fixed with Miniplates, and Model 2 was fixed with Reconstruction plate. Loading forces of 120 N at molar region and 62.5 N at incisor region were applied. These two models were imported to ANSY'S Workbench Software. Result Miniplate fixation model showed comparatively reduced gap between fragments than Reconstruction plate. But the gap values of both the models were within the physiologic limit of healing under this specific loading. Analytically Miniplates were superior to Reconstruction plate in the management of bilateral parasymphysis fracture. Conclusion Analytically Miniplates are superior to Reconstruction plate in the management of bilateral parasymphysis fracture. As the masticatory forces were reduced during fracture healing period, both fixations provide satisfactory healing. So both Miniplate and Reconstruction plate can be considered as fixation method for bilateral parasymphysis mandible fracture.
Collapse
Affiliation(s)
- Manju Kurakar
- Department of Oral and Maxillofacial Surgery, Sinhgad Dental College and Hospital, Pune, India
- Affiliated to Maharashtra University of Health Science, Nashik, Maharshtra India
| | - Udupikrishna Joshi
- Department of Oral and Maxillofacial Surgery, HKE Society’s SNijalingappa Institute of Dental Science and Research, Gulbarga, India
- Affiliated to Rajiv Gandhi University of Health Sciences, Bangalore, Karnataka India
| |
Collapse
|
31
|
Frade BB, Dias RB, Gemini Piperni S, Bonfim DC. The role of macrophages in fracture healing: a narrative review of the recent updates and therapeutic perspectives. Stem Cell Investig 2023; 10:4. [PMID: 36817259 PMCID: PMC9936163 DOI: 10.21037/sci-2022-038] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 01/10/2023] [Indexed: 02/10/2023]
Abstract
Objective This review addresses the latest advances in research on the role of macrophages in fracture healing, exploring their relationship with failures in bone consolidation and the perspectives for the development of advanced and innovative therapies to promote bone regeneration. Background The bone can fully restore its form and function after a fracture. However, the regenerative process of fracture healing is complex and is influenced by several factors, including macrophage activity. These cells have been found in the fracture site at all stages of bone regeneration, and their general depletion or the knockdown of receptors that mediate their differentiation, polarization, and/or function result in impaired fracture healing. Methods The literature search was carried out in the PubMed database, using combinations of the keywords "macrophage", "fracture healing, "bone regeneration", and "bone repair". Articles published within the last years (2017-2022) reporting evidence from in vivo long bone fracture healing experiments were included. Conclusions Studies published in the last five years on the role of macrophages in fracture healing strengthened the idea that what appears to be essential when it comes to a successful consolidation is the right balance between the M1/M2 populations, which have different but complementary roles in the process. These findings opened promising new avenues for the development of several macrophage-targeted therapies, including the administration of molecules and/or biomaterials intended to regulate macrophage differentiation and polarization, the local transplantation of macrophage precursors, and the use of exosomes to deliver signaling molecules that influence macrophage activities. However, more research is still warranted to better understand the diversity of macrophage phenotypes and their specific roles in each step of fracture healing and to decipher the key molecular mechanisms involved in the in vivo crosstalk between macrophages and other microenvironmental cell types, such as endothelial and skeletal stem/progenitor cells.
Collapse
Affiliation(s)
- Bianca Braga Frade
- Laboratory of Stem Cells and Bone Regeneration, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil;,Postgraduation Program in Biological Sciences-Biophysics, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rhayra Braga Dias
- Laboratory of Stem Cells and Bone Regeneration, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil;,Postgraduation Program in Morphological Sciences, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sara Gemini Piperni
- Laboratory of Biotechnology, Bioengineering and Nanostructured Biomaterials, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Danielle Cabral Bonfim
- Laboratory of Stem Cells and Bone Regeneration, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
Gao X, Sun X, Cheng H, Ruzbarsky JJ, Mullen M, Huard M, Huard J. MRL/MpJ Mice Resist to Age-Related and Long-Term Ovariectomy-Induced Bone Loss: Implications for Bone Regeneration and Repair. Int J Mol Sci 2023; 24:ijms24032396. [PMID: 36768718 PMCID: PMC9916619 DOI: 10.3390/ijms24032396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/14/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Osteoporosis and age-related bone loss increase bone fracture risk and impair bone healing. The need for identifying new factors to prevent or treat bone loss is critical. Previously, we reported that young MRL/MpJ mice have superior bone microarchitecture and biomechanical properties as compared to wild-type (WT) mice. In this study, MRL/MpJ mice were tested for resistance to age-related and long-term ovariectomy-induced bone loss to uncover potential beneficial factors for bone regeneration and repair. Bone tissues collected from 14-month-old MRL/MpJ and C57BL/6J (WT) mice were analyzed using micro-CT, histology, and immunohistochemistry, and serum protein markers were characterized using ELISAs or multiplex assays. Furthermore, 4-month-old MRL/MpJ and WT mice were subjected to ovariectomy (OV) or sham surgery and bone loss was monitored continuously using micro-CT at 1, 2, 4, and 6 months (M) after surgery with histology and immunohistochemistry performed at 6 M post-surgery. Sera were collected for biomarker detection using ELISA and multiplex assays at 6 M after surgery. Our results indicated that MRL/MpJ mice maintained better bone microarchitecture and higher bone mass than WT mice during aging and long-term ovariectomy. This resistance of bone loss observed in MRL/MpJ mice correlated with the maintenance of higher OSX+ osteoprogenitor cell pools, higher activation of the pSMAD5 signaling pathway, more PCNA+ cells, and a lower number of osteoclasts. Systemically, lower serum RANKL and DKK1 with higher serum IGF1 and OPG in MRL/MpJ mice relative to WT mice may also contribute to the maintenance of higher bone microarchitecture during aging and less severe bone loss after long-term ovariectomy. These findings may be used to develop therapeutic approaches to maintain bone mass and improve bone regeneration and repair due to injury, disease, and aging.
Collapse
Affiliation(s)
- Xueqin Gao
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Correspondence: (X.G.); (J.H.)
| | - Xuying Sun
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Haizi Cheng
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Joseph J. Ruzbarsky
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
- The Steadman Clinic, Vail, CO 81657, USA
| | - Michael Mullen
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Matthieu Huard
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Johnny Huard
- Linda and Mitch Hart Center for Regenerative and Personalized Medicine, Steadman Philippon Research Institute, Vail, CO 81657, USA
- Department of Orthopaedic Surgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX 77054, USA
- Correspondence: (X.G.); (J.H.)
| |
Collapse
|
33
|
Novak S, Madunic J, Shum L, Vucetic M, Wang X, Tanigawa H, Ghosh M, Sanjay A, Kalajzic I. PDGF inhibits BMP2-induced bone healing. NPJ Regen Med 2023; 8:3. [PMID: 36631491 PMCID: PMC9834334 DOI: 10.1038/s41536-023-00276-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Bone regeneration depends on a pool of bone/cartilage stem/progenitor cells and signaling mechanisms regulating their differentiation. Using in vitro approach, we have shown that PDGF signaling through PDGFRβ inhibits BMP2-induced osteogenesis, and significantly attenuates expression of BMP2 target genes. We evaluated outcomes of treatment with two anabolic agents, PDGF and BMP2 using different bone healing models. Targeted deletion of PDGFRβ in αSMA osteoprogenitors, led to increased callus bone mass, resulting in improved biomechanical properties of fractures. In critical size bone defects BMP2 treatment increased proportion of osteoprogenitors, while the combined treatment of PDGF BB with BMP2 decreased progenitor number at the injury site. BMP2 treatment induced significant bone formation and increased number of osteoblasts, while in contrast combined treatment with PDGF BB decreased osteoblast numbers. This is in vivo study showing that PDGF inhibits BMP2-induced osteogenesis, but inhibiting PDGF signaling early in healing process does not improve BMP2-induced bone healing.
Collapse
Affiliation(s)
- Sanja Novak
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| | - Josip Madunic
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA ,grid.414681.e0000 0004 0452 3941Biochemistry and Organic Analytical Chemistry Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Laura Shum
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| | - Milan Vucetic
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| | - Xi Wang
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| | - Hitoshi Tanigawa
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| | - Mallika Ghosh
- grid.208078.50000000419370394Center for Vascular Biology, UConn Health, Farmington, CT USA
| | - Archana Sanjay
- grid.208078.50000000419370394Department of Orthopeadic Surgery, UConn Health, Farmington, CT USA
| | - Ivo Kalajzic
- grid.208078.50000000419370394Center for Regenerative Medicine and Skeletal Development, UConn Health, Farmington, CT USA
| |
Collapse
|
34
|
Novak S, Kalajzic I. AcanCreER lacks specificity to chondrocytes and targets periosteal progenitors in the fractured callus. Bone 2023; 166:116599. [PMID: 36309308 PMCID: PMC9832919 DOI: 10.1016/j.bone.2022.116599] [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: 09/26/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Aggrecan (Acan) is a large proteoglycan molecule constituting the extracellular matrix of cartilage, secreted by chondrocytes. To specifically target the chondrocyte lineage, researchers have widely used the AcanCreER mouse model. Evaluation of specificity and efficiency of recombination, requires Cre animals to be crossed with reporter mice. In order to accurately interpret data from Cre models, it is imperative to consider A) the amount of recombination occurring in cells/tissues that are not intended for targeting (i.e., non-specific expression), B) the efficiency of Cre recombination, which can depend on dose and duration of tamoxifen treatment, and C) the activation of CreER without tamoxifen induction, known as "Cre leakage." Using a highly sensitive reporter mouse (Ai9, tdTomato), we performed a comprehensive analysis of the AcanCreER system. Surprisingly, we observed expression in cells within the periosteum. These cells expand at a stage when chondrocytes are not yet present within the forming callus tissue (Acan/Ai9+ cells). In pulse-chase experiments, we confirmed that fibroblastic Acan/Ai9+ cells within the periosteum can directly give rise to osteoblasts. Our results show that Acan/Ai9+ is not specific for the chondrocyte lineage in the fracture callus or with the tibial holes. The expression of AcanCreER in periosteal progenitor cells complicates the interpretation of studies evaluating the transition of chondrocytes to osteoblasts (termed transdifferentiation). Awareness of these issues and the limitations of the system will lead to better data interpretation.
Collapse
Affiliation(s)
- Sanja Novak
- Department of Reconstructive Sciences, UConn Health, Farmington, CT, USA
| | - Ivo Kalajzic
- Department of Reconstructive Sciences, UConn Health, Farmington, CT, USA.
| |
Collapse
|
35
|
Yang X, Mai YX, Wei L, Peng LY, Pang FX, Wang LJ, Li ZP, Zhang JF, Jin AM. MLK3 silence suppressed osteogenic differentiation and delayed bone formation via influencing the bone metabolism and disturbing MAPK signaling. J Orthop Translat 2023; 38:98-105. [PMCID: PMC9619354 DOI: 10.1016/j.jot.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/21/2022] [Accepted: 07/05/2022] [Indexed: 11/11/2022] Open
Affiliation(s)
- Xiao Yang
- Department of Spinal Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yong-xin Mai
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lan Wei
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li-yang Peng
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Feng-xiang Pang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ling-jun Wang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhi-peng Li
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Rehabilitation, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Corresponding author. Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China. Tel: +86 13724839892.
| | - Jin-fang Zhang
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China
- Key Laboratory of Orthopaedics and Traumatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
- Corresponding author. Department of Spinal Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China. Tel: +86 13802983267.
| | - An-min Jin
- Department of Spinal Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Corresponding author. Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
36
|
Muacevic A, Adler JR, Sameer Tammar R, Essa Zidan L, Nafe Alsatty A, Fahad Bahanan S, Almutairi AF. The Relationship Between Time Until Full Weight Bearing After Hip Fractures and Vitamin D Levels in Patients Aged 50 Years and Above. Cureus 2022; 14:e32918. [PMID: 36578860 PMCID: PMC9791611 DOI: 10.7759/cureus.32918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/25/2022] [Indexed: 12/26/2022] Open
Abstract
Introduction Hip fractures are common in the elderly, especially with vitamin D deficiency. Currently, there is a paucity of case-control studies regarding the relationship between the time until full weight bearing of hip fractures and vitamin D levels in Saudi Arabia. Our aim is to determine time until full weight bearing of hip fractures in patients with vitamin D deficiency compared with normal vitamin D in the age of 50 and above. Materials and methods This was a hospital-based non-interventional retrospective case-control study conducted among patients with hip fractures aged 50 years and above between January 2017 and April 2021. It was done at King Abdulaziz university hospital, Jeddah, Saudi Arabia. A review of the medical records and operation records for the relative operation was done to check patients' lab values around the time of the operation and following the documented healing process. Data were analyzed using SPSS version 26 (IBM Inc., Armonk, New York). Correlation analysis was performed using Spearman's test, and a p-value of 0.05 was considered statistically significant. Results In our research, 36 patients were participants in the study, with about two-thirds (22) of the participants being female (61.1%). 52.8% of the participants had an age ranging from 71-80 years, with a mean age of 75.66 ± 9.53 years. A non-significant relationship between the time until full weight bearing of hip fractures and the vitamin d levels is demonstrated. By that, the research question was disproved by the given data. Conclusion The time until full weight bearing of hip fracture is not significantly related to vitamin D level. In this study, the majority of patients (77.1%) were vitamin D deficient, which raises our concern for vitamin D deficiency to be a major health problem in our society. However, this evidence should be further assessed in larger trials. Additional studies on this topic are recommended to be done.
Collapse
|
37
|
Wang W, Liang X, Zheng K, Ge G, Chen X, Xu Y, Bai J, Pan G, Geng D. Horizon of exosome-mediated bone tissue regeneration: The all-rounder role in biomaterial engineering. Mater Today Bio 2022; 16:100355. [PMID: 35875196 PMCID: PMC9304878 DOI: 10.1016/j.mtbio.2022.100355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 12/02/2022] Open
Abstract
Bone injury repair has always been a tricky problem in clinic, the recent emergence of bone tissue engineering provides a new direction for the repair of bone injury. However, some bone tissue processes fail to achieve satisfactory results mainly due to insufficient vascularization or cellular immune rejection. Exosomes with the ability of vesicle-mediated intercellular signal transmission have gained worldwide attention and can achieve cell-free therapy. Exosomes are small vesicles that are secreted by cells, which contain genetic material, lipids, proteins and other substances. It has been found to play the function of material exchange between cells. It is widely used in bone tissue engineering to achieve cell-free therapy because it not only does not produce some immune rejection like cells, but also can play a cell-like function. Exosomes from different sources can bind to scaffolds in various ways and affect osteoblast, angioblast, and macrophage polarization in vivo to promote bone regeneration. This article reviews the recent research progress of exosome-loaded tissue engineering, focusing on the mechanism of exosomes from different sources and the application of exosome-loaded scaffolds in promoting bone regeneration. Finally, the existing deficiencies and challenges, future development directions and prospects are summarized.
Collapse
Affiliation(s)
- Wentao Wang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Xiaolong Liang
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Kai Zheng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Xu Chen
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Yaozeng Xu
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| | - Guoqing Pan
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Dechun Geng
- Department of Orthopaedics, The First Affiliated Hospital of Soochow University, 188 Shizi Street, Suzhou 215006, Jiangsu, China
| |
Collapse
|
38
|
Kumari S, Katiyar S, Darshna, Anand A, Singh D, Singh BN, Mallick SP, Mishra A, Srivastava P. Design strategies for composite matrix and multifunctional polymeric scaffolds with enhanced bioactivity for bone tissue engineering. Front Chem 2022; 10:1051678. [PMID: 36518978 PMCID: PMC9742444 DOI: 10.3389/fchem.2022.1051678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/14/2022] [Indexed: 09/19/2023] Open
Abstract
Over the past few decades, various bioactive material-based scaffolds were investigated and researchers across the globe are actively involved in establishing a potential state-of-the-art for bone tissue engineering applications, wherein several disciplines like clinical medicine, materials science, and biotechnology are involved. The present review article's main aim is to focus on repairing and restoring bone tissue defects by enhancing the bioactivity of fabricated bone tissue scaffolds and providing a suitable microenvironment for the bone cells to fasten the healing process. It deals with the various surface modification strategies and smart composite materials development that are involved in the treatment of bone tissue defects. Orthopaedic researchers and clinicians constantly focus on developing strategies that can naturally imitate not only the bone tissue architecture but also its functional properties to modulate cellular behaviour to facilitate bridging, callus formation and osteogenesis at critical bone defects. This review summarizes the currently available polymeric composite matrices and the methods to improve their bioactivity for bone tissue regeneration effectively.
Collapse
Affiliation(s)
- Shikha Kumari
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Soumya Katiyar
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Darshna
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Aditya Anand
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Divakar Singh
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | - Bhisham Narayan Singh
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Sarada Prasanna Mallick
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram, Andhra Pradesh, India
| | - Abha Mishra
- School of Biochemical Engineering, IIT BHU, Varanasi, India
| | | |
Collapse
|
39
|
Zha J, Zhang G, Wang X, Li J, Di J, Guo J. Use of a Minimally Invasive Traction Repositor versus Conventional Manual Traction for the Treatment of Tibial Fractures: A Comparative Study from a Tertiary Hospital in China. Ther Clin Risk Manag 2022; 18:945-954. [PMID: 36176542 PMCID: PMC9514298 DOI: 10.2147/tcrm.s379135] [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: 06/18/2022] [Accepted: 09/15/2022] [Indexed: 11/23/2022] Open
Abstract
Background Closed reduction and intramedullary nail fixation of tibial fractures may not utilize a fracture table or reduction aids like a femoral distractor, and only manual traction will help aid the reduction process. This study aimed to describe and further investigate the effectiveness of an originally designed minimally invasive traction repositor (MITR) for the treatment of tibial fractures. Methods From January 2018 to April 2021, a total of 119 eligible patients with tibial shaft fractures were included and retrospectively assigned to two groups according to different reduction methods: MITR group vs conventional manual traction (CMT) group. The baseline characteristics between the two groups were comparable, including age, gender, BMI, residence, smoking history, drinking history, injury mechanism, fracture type, ASA, method of anesthesia, and surgical delay (all P > 0.05). The operation time, fracture reduction duration, intraoperative blood loss, fluoroscopy time, number of intraoperative fluoroscopies, VAS, HSS, fracture healing time, and complications were compared. Results All patients completed the follow-ups with an average of 18.5 months (range 12-42 months). The operation time, fracture reduction duration, intraoperative blood loss, fluoroscopy time, and number of fluoroscopies were significantly decreased in the MITR group (all P < 0.05). At one month postoperatively, the VAS score was statistically lower in the MITR group (1.8±0.8) than in the CMT group (2.6±1.5). At 6 months postoperatively, the HHS score was statistically higher in the MITR group (90.8±2.3) than in the CMT group (86.4±3.8). We observed no statistical difference in the mean fracture healing time, bone nonunion, implant failure, and infection between the two groups (all P > 0.05). Conclusion Compared with CMT, MITR facilitates the minimally invasive treatment of tibial fractures and has the advantages of operation time, fracture reduction duration, intraoperative blood loss, fluoroscopy time, number of fluoroscopies, and satisfactory VAS and HSS scores.
Collapse
Affiliation(s)
- Junpu Zha
- Department of Orthopaedics Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China.,Orthopaedic Institute of Hebei Province, Shijiazhuang, People's Republic of China
| | - Guolei Zhang
- Department of Orthopaedics Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China.,Orthopaedic Institute of Hebei Province, Shijiazhuang, People's Republic of China
| | - Xiaoqing Wang
- Department of Orthopaedics Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jie Li
- Department of Obstetrics and Gynecology, Hebei General Hospital, Shijiazhuang, People's Republic of China
| | - Jun Di
- Department of Orthopaedics Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China.,Orthopaedic Institute of Hebei Province, Shijiazhuang, People's Republic of China
| | - Junfei Guo
- Department of Orthopaedics Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China.,Orthopaedic Institute of Hebei Province, Shijiazhuang, People's Republic of China
| |
Collapse
|
40
|
Jiang Y, Wang X, Huang W, Zhu Y, Zhang K, Feng D. A novel bone graft technique combined with plating for aseptic recalcitrant long bone nonunion. BMC Musculoskelet Disord 2022; 23:873. [PMID: 36127646 PMCID: PMC9487050 DOI: 10.1186/s12891-022-05830-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Background To evaluate the outcomes and efficacy of a new technique of autogenous iliac crest bone grafting combined with locking compression plate (LCP) vertical fixation for aseptic recalcitrant long bone nonunion. Methods From July 2010 to September 2020, 36 aseptic recalcitrant long bone nonunions were treated with a bone-forming channel technique and internal LCP fixation. All the patients had received one or more failed treatments. The injury mechanism, nonunion type and duration, and prior treatments were recorded pre-operation. The routine treatment process included nonunion area exposure, previous implant removal, sclerotic bone debridement, LCP fixation, bone-forming channel creation, and iliac bone grafting, and a second LCP fixation when required. At follow-up, X-ray images were obtained to assess bone healing and implant failure. Visual analog scale (VAS), fracture site stability, limb function, activity, muscle strength, limb length, and complications were recorded. Results A total of 34 patients (24 males and 10 females) were finally enrolled, with a mean age of 49.8 ± 12.3 years. At a mean follow-up of 35.6 ± 22.0 months, 32 patients displayed bone union, with a healing rate of 94.1% and mean union time of 6.8 ± 2.4 months. The VAS score was 0.7 ± 1 at the final follow-up. The functional results showed that 19 patients were excellent, 11 patients were good, 2 patients were poor, and 2 patients did not heal. Conclusion Bone-forming channel technique combined with LCP vertical fixation is an excellent option to treat recalcitrant long bone nonunion. Level of evidence Therapeutic Level IV.
Collapse
Affiliation(s)
- Yuxuan Jiang
- Department of Orthopaedic Trauma, Hong Hui Hospital, Xi'an Jiaotong University School of Medicine, Youyi Road, Xi'an, Shaanxi Province, 710054, China
| | - Xiaolong Wang
- Department of Orthopaedic Trauma, Hong Hui Hospital, Xi'an Jiaotong University School of Medicine, Youyi Road, Xi'an, Shaanxi Province, 710054, China
| | - Wei Huang
- Department of Orthopaedic Trauma, Hong Hui Hospital, Xi'an Jiaotong University School of Medicine, Youyi Road, Xi'an, Shaanxi Province, 710054, China
| | - Yangjun Zhu
- Department of Orthopaedic Trauma, Hong Hui Hospital, Xi'an Jiaotong University School of Medicine, Youyi Road, Xi'an, Shaanxi Province, 710054, China
| | - Kun Zhang
- Department of Orthopaedic Trauma, Hong Hui Hospital, Xi'an Jiaotong University School of Medicine, Youyi Road, Xi'an, Shaanxi Province, 710054, China
| | - Dongxu Feng
- Department of Orthopaedic Trauma, Hong Hui Hospital, Xi'an Jiaotong University School of Medicine, Youyi Road, Xi'an, Shaanxi Province, 710054, China.
| |
Collapse
|
41
|
Fukase N, Duke VR, Lin MC, Stake IK, Huard M, Huard J, Marmor MT, Maharbiz MM, Ehrhart NP, Bahney CS, Herfat ST. Wireless Measurements Using Electrical Impedance Spectroscopy to Monitor Fracture Healing. SENSORS (BASEL, SWITZERLAND) 2022; 22:s22166233. [PMID: 36016004 PMCID: PMC9412277 DOI: 10.3390/s22166233] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/12/2022] [Accepted: 08/16/2022] [Indexed: 05/05/2023]
Abstract
There is an unmet need for improved, clinically relevant methods to longitudinally quantify bone healing during fracture care. Here we develop a smart bone plate to wirelessly monitor healing utilizing electrical impedance spectroscopy (EIS) to provide real-time data on tissue composition within the fracture callus. To validate our technology, we created a 1-mm rabbit tibial defect and fixed the bone with a standard veterinary plate modified with a custom-designed housing that included two impedance sensors capable of wireless transmission. Impedance magnitude and phase measurements were transmitted every 48 h for up to 10 weeks. Bone healing was assessed by X-ray, µCT, and histology. Our results indicated the sensors successfully incorporated into the fracture callus and did not impede repair. Electrical impedance, resistance, and reactance increased steadily from weeks 3 to 7-corresponding to the transition from hematoma to cartilage to bone within the fracture gap-then plateaued as the bone began to consolidate. These three electrical readings significantly correlated with traditional measurements of bone healing and successfully distinguished between union and not-healed fractures, with the strongest relationship found with impedance magnitude. These results suggest that our EIS smart bone plate can provide continuous and highly sensitive quantitative tissue measurements throughout the course of fracture healing to better guide personalized clinical care.
Collapse
Affiliation(s)
- Naomasa Fukase
- Linda and Mitch Hart Center for Regenerative & Personalized Medicine at the Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Victoria R. Duke
- Linda and Mitch Hart Center for Regenerative & Personalized Medicine at the Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Monica C. Lin
- UCSF Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA 94110, USA
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| | - Ingrid K. Stake
- Linda and Mitch Hart Center for Regenerative & Personalized Medicine at the Steadman Philippon Research Institute, Vail, CO 81657, USA
- Department of Orthopaedic Surgery, Ostfold Hospital Trust, 1714 Graalum, Norway
| | - Matthieu Huard
- Linda and Mitch Hart Center for Regenerative & Personalized Medicine at the Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Johnny Huard
- Linda and Mitch Hart Center for Regenerative & Personalized Medicine at the Steadman Philippon Research Institute, Vail, CO 81657, USA
| | - Meir T. Marmor
- UCSF Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA 94110, USA
| | - Michel M. Maharbiz
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Nicole P. Ehrhart
- Department of Clinical Sciences, Flint Animal Cancer Center, College of Veterinary Medicine, Colorado State University, Fort Collins, CO 80523, USA
| | - Chelsea S. Bahney
- Linda and Mitch Hart Center for Regenerative & Personalized Medicine at the Steadman Philippon Research Institute, Vail, CO 81657, USA
- UCSF Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA 94110, USA
- Correspondence: (C.S.B.); (S.T.H.)
| | - Safa T. Herfat
- UCSF Orthopaedic Trauma Institute, Zuckerberg San Francisco General Hospital, San Francisco, CA 94110, USA
- Correspondence: (C.S.B.); (S.T.H.)
| |
Collapse
|
42
|
Guzzini M, Arioli L, Mori F, Ferretti A. Is it really necessary to perform venous anastomosis in vascularized corticoperiosteal bone flap? A randomized prospective 4-year follow-up study. Injury 2022; 53:2859-2864. [PMID: 35753816 DOI: 10.1016/j.injury.2022.06.022] [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: 02/27/2022] [Revised: 06/12/2022] [Accepted: 06/13/2022] [Indexed: 02/02/2023]
Abstract
BACKGROUND This study aims to investigate the treatment results of atrophic nonunion of the lower and upper limb with vascularized bone flaps performed with only arterial anastomosis versus the execution of flaps performed with arterial and venous anastomosis, comparing the surgical time and the healing rate of these two techniques. METHODS 49 patients were enrolled in this study and were randomly divided into two groups: group A, the control group, consisted of 27 patients, who underwent vascularized corticoperiosteal bone flap with both arterial and venous anastomosis; group B, consisted of 22 patients, who underwent vascularized corticoperiosteal bone flap with only arterial anastomosis. The surgical time, the time to harvest the graft and the microsurgical time were evaluated. Radiological and clinical follow-ups were performed with one independent and blinded investigator to avoid bias. RESULTS A significant reduction in the duration of the intervention in group B was found: a 13.63% reduction of the total surgical time and a 41.75% reduction of the microsurgical time. A significant difference was not found between groups A and B in bone healing time. All the patients of both groups were able to return to their daily life activities. CONCLUSIONS Our investigation suggests that it isn't necessary to perform the venous anastomosis between the flap pedicle and the recipient area. Presumably, the venous blood flow reaches the systemic circulation through the vascular bone marrow network. Our procedure avoids venous trauma during the dissection and execution of the anastomosis and, therefore, can minimize complications such as venous thrombosis.
Collapse
Affiliation(s)
- Matteo Guzzini
- "Sapienza" University of Rome, Unit of Orthopedics and Traumatology, S. Andrea University Hospital, Via Grottarossa, 1035, 00186, Rome, Italy
| | - Leopoldo Arioli
- "Sapienza" University of Rome, Unit of Orthopedics and Traumatology, S. Andrea University Hospital, Via Grottarossa, 1035, 00186, Rome, Italy.
| | - Fabiana Mori
- "Università degli studi di Padova", Unit of Orthopedics and Traumatology, University Hospital of Padua, Via Nicolò Giustiniani, 1, 35128 Padua, Italy
| | - Andrea Ferretti
- "Sapienza" University of Rome, Unit of Orthopedics and Traumatology, S. Andrea University Hospital, Via Grottarossa, 1035, 00186, Rome, Italy
| |
Collapse
|
43
|
Bromer FD, Brent MB, Thomsen JS, Brüel A. Drill-Hole Bone Defects in Animal Models of Bone Healing: Protocol for a Systematic Review. JMIR Res Protoc 2022; 11:e34887. [PMID: 35849443 PMCID: PMC9345022 DOI: 10.2196/34887] [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: 11/11/2021] [Revised: 04/29/2022] [Accepted: 05/24/2022] [Indexed: 11/28/2022] Open
Abstract
Background Bone fractures are common conditions of the musculoskeletal system. Several animal models of bone fractures have been established to help elucidate the complex process of bone healing. In the last decades, drill-hole bone defects have emerged as a method to study bone healing. Animal models of drill-hole defects are easy to standardize and do not require external fixation of the bone. However, current studies of drill-hole bone defects lack detailed descriptions of techniques and interstudy standardization. Objective This systematic review aims to present a detailed description of the different methods used to induce drill-hole bone defects in long bones of laboratory animals and to provide a comprehensive overview of their methodology and potential for investigation of bone healing. Methods A systematic search of PubMed and Embase will be performed of abstracts containing variations of the following four keywords: “long bone,” “drill-hole,” “regeneration,” and “animal model.” Abstract screening and full-text screening will be performed independently by 2 reviewers, and data will be extracted to a predesigned extraction protocol. The primary outcome of the included studies is the technique used to create the drill-hole bone defect, and secondary outcomes are any measurements or analyses of bone defect and regeneration. A narrative synthesis will be used to present the primary outcome, while information on secondary outcomes will be displayed graphically. The study protocol follows the PRISMA-P (Preferred Reporting Items for Systematic Review and Meta-analysis Protocols) guidelines. Results Abstract and full-text screening is ongoing and is expected to be completed by October 2022. Data extraction will commence immediately after, and the manuscript is expected to be completed by December 2023. The systematic review will follow the PRISMA statement. Conclusions The strength of this systematic review is that it provides a comprehensive methodological overview of the different drill-hole methods and their advantages and disadvantages. This will assist researchers in choosing which model to use when studying different aspects of bone healing. Trial Registration International Prospective Register of Systematic Reviews CRD42020213076; https://tinyurl.com/bp56wdwe International Registered Report Identifier (IRRID) PRR1-10.2196/34887
Collapse
Affiliation(s)
| | - Mikkel Bo Brent
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | | | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
44
|
Lowen GB, Garrett KA, Moore-Lotridge SN, Uppuganti S, Guelcher SA, Schoenecker JG, Nyman JS. Effect of Intramedullary Nailing Patterns on Interfragmentary Strain in a Mouse Femur Fracture: A Parametric Finite Element Analysis. J Biomech Eng 2022; 144:051007. [PMID: 34802060 PMCID: PMC8822464 DOI: 10.1115/1.4053085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 11/17/2021] [Indexed: 11/08/2022]
Abstract
Delayed long bone fracture healing and nonunion continue to be a significant socioeconomic burden. While mechanical stimulation is known to be an important determinant of the bone repair process, understanding how the magnitude, mode, and commencement of interfragmentary strain (IFS) affect fracture healing can guide new therapeutic strategies to prevent delayed healing or nonunion. Mouse models provide a means to investigate the molecular and cellular aspects of fracture repair, yet there is only one commercially available, clinically-relevant, locking intramedullary nail (IMN) currently available for studying long bone fractures in rodents. Having access to alternative IMNs would allow a variety of mechanical environments at the fracture site to be evaluated, and the purpose of this proof-of-concept finite element analysis study is to identify which IMN design parameters have the largest impact on IFS in a murine transverse femoral osteotomy model. Using the dimensions of the clinically relevant IMN as a guide, the nail material, distance between interlocking screws, and clearance between the nail and endosteal surface were varied between simulations. Of these parameters, changing the nail material from stainless steel (SS) to polyetheretherketone (PEEK) had the largest impact on IFS. Reducing the distance between the proximal and distal interlocking screws substantially affected IFS only when nail modulus was low. Therefore, IMNs with low modulus (e.g., PEEK) can be used alongside commercially available SS nails to investigate the effect of initial IFS or stability on fracture healing with respect to different biological conditions of repair in rodents.
Collapse
Affiliation(s)
- Gregory B. Lowen
- Vanderbilt University, Department of Chemical and Biomolecular Engineering, 2201 West End Ave, Nashville, TN 37235
| | - Katherine A. Garrett
- Vanderbilt University Medical Center, Department of Orthopaedic Surgery, 1215 21 Ave. S., Suite 4200, Nashville, TN 37232
| | - Stephanie N. Moore-Lotridge
- Vanderbilt University Medical Center, Department of Orthopaedic Surgery, 1215 21 Ave. S., Suite 4200, Nashville, TN 37232;Vanderbilt University Medical Center, Vanderbilt Center for Bone Biology, 1211 Medical Center Dr., Nashville, TN 37212
| | - Sasidhar Uppuganti
- Vanderbilt University Medical Center, Department of Orthopaedic Surgery, 1215 21 Ave. S., Suite 4200, Nashville, TN 37232;Vanderbilt University Medical Center, Vanderbilt Center for Bone Biology, 1211 Medical Center Dr., Nashville, TN 37212
| | - Scott A. Guelcher
- Vanderbilt University, Department of Chemical and Biomolecular Engineering, 2201 West End Ave, Nashville, TN 37235; Vanderbilt University, Department of Biomedical Engineering, 5824 Stevenson Center, Nashville, TN 37232; Vanderbilt University Medical Center, Vanderbilt Center for Bone Biology, 1211 Medical Center Dr., Nashville, TN 37212; Vanderbilt University Medical Center, Division of Clinical Pharmacology, 1211 Medical Center Dr, Nashville, TN 37217
| | - Jonathan G. Schoenecker
- Vanderbilt University, Department of Pharmacology, 465 21 Ave South, 7124 Medical Research Building III, Nashville, TN 37232; Vanderbilt University Medical Center, Vanderbilt Center for Bone Biology, 1211 Medical Center Dr., Nashville, TN 37212; Vanderbilt University Medical Center, Department of Pathology, Microbiology, and Immunology, 1161 21 Ave S C-3322 Medical Center North, Nashville, TN 37232; Vanderbilt University Medical Center, Department of Pediatrics, 2200 Children's Way, Suite 2404, Nashville, TN 37232
| | - Jeffry S. Nyman
- Vanderbilt University, Department of Biomedical Engineering, 5824 Stevenson Center, Nashville, TN 37232; Vanderbilt University Medical Center, Department of Orthopaedic Surgery, 1215 21 Ave. S., Suite 4200, Nashville, TN 37232; Vanderbilt University Medical Center, Vanderbilt Center for Bone Biology, 1211 Medical Center Dr., Nashville, TN 37212; Tennessee Valley Healthcare System, Department of Veterans Affairs, 1310 24 Ave. S, Nashville, TN 37212
| |
Collapse
|
45
|
Hu Y, He Y, Fang J, Liu Y, Cao Y, Tong W, Chen W, Shao Z, Liu Y, Tian H. Wnt10b-overexpressing umbilical cord mesenchymal stem cells promote fracture healing via accelerated cartilage callus to bone remodeling. Bioengineered 2022; 13:10313-10323. [PMID: 35436412 PMCID: PMC9161882 DOI: 10.1080/21655979.2022.2062954] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
The aim of this study was to investigate whether HUCMSCsWnt10b could promote long bone fracture healing. Commercially-available HUCMSCsEmp (human umbilical cord mesenchymal stem cells transfected with empty vector) in hydrogel, HUCMSCsWnt10b in hydrogel and HUCMSCsWnt10b with the Wnt signaling pathway inhibitor IWR-1 were transplanted into the fracture site in a rat model of femoral fracture. We found that transplantation of HUCMSCsWnt10b significantly accelerated bone healing in a rat model of femoral fracture. Meanwhile, three-point bending test proved that the mechanical properties of the bone at the fracture site in the HUCMSCWnt10b treatment group were significantly better than those of the other treatment groups. To understand the cellular mechanism, we explored the viability of periosteal stem cells (PSCs), as they contribute the greatest number of osteoblast lineage cells to the callus. In line with in vivo data, we found that conditioned medium from HUCMSCsWnt10b enhanced the migration and osteogenic differentiation of PSCs. Furthermore, conditioned medium from HUCMSCsWnt10b also induced endothelial cells to form capillary-like structures in a tube formation assay, which was blocked by SU5416, an angiogenesis inhibitor, suggesting that enhanced vessel formation and growth also contribute to accelerated hard callus formation. In summary, our study demonstrates that HUCMSCsWnt10b promote fracture healing via accelerated hard callus formation, possibly due to enhanced osteogenic differentiation of PSCs and vessel growth. Therefore, HUCMSCsWnt10b may be a promising treatment for long bone fractures.
Collapse
Affiliation(s)
- Yuxiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Yu He
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Jiarui Fang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Yunlu Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Yulin Cao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Wei Tong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Wei Chen
- Department of Orthopedics, The Third Hospital of Hebei Medical University, Shi Jiazhuang, Hebei, China.,Nhc Key Laboratory of Intelligent Orthopedic Equipment (The Third Hospital of Hebei Medical University), Shi Jiazhuang, Hebei, China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Yong Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| | - Hongtao Tian
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan Hubei, China
| |
Collapse
|
46
|
Yang Q, Liu J, Tan L, Jiang Y, Zhu D. Polyostotic Fibrous Dysplasia Complicated by Pathological Fracture of Right Femoral Shaft with Nonunion: A Case Report. Front Surg 2022; 9:879550. [PMID: 35495762 PMCID: PMC9039455 DOI: 10.3389/fsurg.2022.879550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 03/28/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Fibrous dysplasia is a benign fibrous bone tumor that accounts for 5% to 10% of benign bone tumors. It can manifest as simple fibrous dysplasia (70%–80%), polyostotic fibrous dysplasia (20%–30%), with approximately the same incidence in men and women. We report a patient with a rare case of multiple fibrous dysplasia combined with proximal femoral shepherd deformity with pathological fracture of the femoral shaft complicated by nonunion. It is necessary to understand the disease in more detail to avoid overtreatment of benign lesions or misdiagnosis of malignant tumors and other diseases. Case presentation A 58-year-old man with polyostotic fibrous dysplasia, bilateral proximal femur deformity, Shepherd’s angle deformity, right femoral shaft pathological fracture complicated by nonunion, we under fluoroscopy, in the obvious proximal fracture, take osteotomy, and process the shape of the cut bone fragment to adapt it to the corrected force line, and then restore it back to its original position, using intramedullary nailing technology complete the operation. Three months after the operation, he came to the hospital for re-examination, and an X-ray of the right femur was taken. It was found that the fractured end had a tendency to heal. The patient was instructed to gradually bear weight. After six months of re-examination, the patient could walk with a walker. One year after the operation, the patient could walk without a walker and take care of himself at home. However, there was still stretch-like pain in the right lower back, but it was tolerable. Conclusions For patients with polyostotic fibrous dysplasia combined with proximal femoral shepherd deformity and pathological fracture of the femoral shaft with nonunion, osteotomy combined with intramedullary nailing is a simple and convenient way to correct the deformity and obtain correct fracture alignment.
Collapse
Affiliation(s)
- Qifan Yang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Jing Liu
- The First Clinical Medical College of Bin Zhou Medical College, Binzhou, China
| | - Lei Tan
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Ye Jiang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Dong Zhu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
- Correspondence: Dong Zhu zhu
| |
Collapse
|
47
|
Zhu L, Liu Y, Wang A, Zhu Z, Li Y, Zhu C, Che Z, Liu T, Liu H, Huang L. Application of BMP in Bone Tissue Engineering. Front Bioeng Biotechnol 2022; 10:810880. [PMID: 35433652 PMCID: PMC9008764 DOI: 10.3389/fbioe.2022.810880] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 03/01/2022] [Indexed: 01/15/2023] Open
Abstract
At present, bone nonunion and delayed union are still difficult problems in orthopaedics. Since the discovery of bone morphogenetic protein (BMP), it has been widely used in various studies due to its powerful role in promoting osteogenesis and chondrogenesis. Current results show that BMPs can promote healing of bone defects and reduce the occurrence of complications. However, the mechanism of BMP in vivo still needs to be explored, and application of BMP alone to a bone defect site cannot achieve good therapeutic effects. It is particularly important to modify implants to carry BMP to achieve slow and sustained release effects by taking advantage of the nature of the implant. This review aims to explain the mechanism of BMP action in vivo, its biological function, and how BMP can be applied to orthopaedic implants to effectively stimulate bone healing in the long term. Notably, implantation of a system that allows sustained release of BMP can provide an effective method to treat bone nonunion and delayed bone healing in the clinic.
Collapse
Affiliation(s)
- Liwei Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
| | - Yuzhe Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Ao Wang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Zhengqing Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Youbin Li
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Chenyi Zhu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Zhenjia Che
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - Tengyue Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
| | - He Liu
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- Orthopaedic Research Institute of Jilin Province, Changchun, China
- *Correspondence: He Liu, ; Lanfeng Huang,
| | - Lanfeng Huang
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: He Liu, ; Lanfeng Huang,
| |
Collapse
|
48
|
Bin-Bin Z, Da-Wa ZX, Chao L, Lan-Tao Z, Tao W, Chuan L, Chao-Zheng L, De-Chun L, Chang F, Shu-Qing W, Zu-Nan D, Xian-Wei P, Zhang ZX, Ke-Wen L. M2 macrophagy-derived exosomal miRNA-26a-5p induces osteogenic differentiation of bone mesenchymal stem cells. J Orthop Surg Res 2022; 17:137. [PMID: 35246197 PMCID: PMC8895825 DOI: 10.1186/s13018-022-03029-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/18/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cells have always been a heated research topic in bone tissue regeneration and repair because of their self-renewal and multi-differentiation potential. A large number of studies have been focused on finding the inducing factors that will promote the osteogenic differentiation of bone marrow mesenchymal stem cells. Previous studies have shown that macrophage exosomes or miRNA-26a-5p can make it work, but the function of this kind of substance on cell osteogenic differentiation has not been public. METHODS M2 macrophages are obtained from IL-4 polarized bone marrow-derived macrophages. Exosomes were isolated from the supernatant of M2 macrophages and identified via transmission electron microscopy (TEM), western blotting, and DLS. Chondrogenic differentiation potential was detected by Alcian blue staining. Oil red O staining was used to detect the potential for lipogenic differentiation. And MTT would detect the proliferative capacity of cells. Western blot was performed to detect differential expression of osteogenic differentiation-related proteins. RESULTS The results showed that M2 macrophage exosomes will promote bone differentiation and at the same time inhibit lipid differentiation. In addition, M2 macrophage-derived exosomes have the function of promoting the expression of SOX and Aggrecan suppressing the level of MMP13. The exosome inhibitor GW4689 suppresses miRNA-26a-5p in M2 macrophage exosomes, and the treated exosomes do not play an important role in promoting bone differentiation. Moreover, miRNA-26a-5p can enable to promote bone differentiation and inhibit lipid differentiation. miRNA-26a-5p can promote the expression of ALP (alkaline phosphatase), RUNX-2 (Runt-related transcription factor 2), OPN(osteopontin), and Col-2(collagen type II). Therefore, it is speculated that exosomal miRNA-26a-5p is indispensable in osteogenic differentiation. CONCLUSIONS The present study indicated that M2 macrophage exosomes carrying miRNA-26a-5p can induce osteogenic differentiation of bone marrow-derived stem cells to inhibit lipogenic differentiation, and miRNA-26a-5p will also promote the expression of osteogenic differentiation-related proteins ALP, RUNX-2, OPN, and Col-2.
Collapse
Affiliation(s)
- Zhang Bin-Bin
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Zha Xi Da-Wa
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Li Chao
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Zhang Lan-Tao
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Wu Tao
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Lu Chuan
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Liu Chao-Zheng
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Li De-Chun
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Feng Chang
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Wei Shu-Qing
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Dong Zu-Nan
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Pei Xian-Wei
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China
| | - Zhi-Xia Zhang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Out-Patient, School of Stomatology, Fourth Military Medical University, Xi'an, 710032, China.
| | - Li Ke-Wen
- Department of Joint Surgery, Qinghai University Affiliated Hospital, Xining, 810000, Qinghai Province, China.
| |
Collapse
|
49
|
Rondanelli AM, Gómez-Sierra MA, Ossa AA, Hernández RD, Torres M. Damage control in orthopaedical and traumatology. Colomb Med (Cali) 2022; 52:e4184802. [PMID: 35027781 PMCID: PMC8754164 DOI: 10.25100/cm.v52i2.4802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/30/2021] [Accepted: 06/27/2021] [Indexed: 01/08/2023] Open
Abstract
In Orthopedics, damage control is indicated in patients with pelvic and/or long bone fractures associated with hemodynamic instability. It is inappropriate to perform a complex definitive reduction and fixation surgery for severely injured trauma patients with hemodynamic instability. In these cases, it is recommended to perform minimally invasive procedures that temporarily stabilize the fractures and bleeding control. Closed or open fractures of the long bones such as femur, tibia, humerus, and pelvis can lead to hemodynamic instability and shock. Thus, orthopedic damage control becomes a priority. However, if the patient is hemodynamically stable, it is recommended to stabilize all fractures with an early permanent internal fixation. These patients will have a shorter hospital length of stay and a reduction in mechanical ventilation, blood components transfusions and complications. Therefore, the concept of orthopedic damage control should be individualized according to the hemodynamic status and the severity of the injuries. Open fractures, dislocations, and vascular injuries could lead to permanent sequelae and complications if a correct management and approach are not performed.
Collapse
Affiliation(s)
- Alfredo Martínez Rondanelli
- Fundación Valle del Lili, Departamento de Cirugía Ortopédica, Cali, Colombia.,Universidad ICESI, Cali, Colombia
| | - María Antonia Gómez-Sierra
- Fundación Valle del Lili, Departamento de Cirugía Ortopédica, Cali, Colombia.,Universidad ICESI, Cali, Colombia
| | - Arley Alberto Ossa
- Fundación Valle del Lili, Departamento de Cirugía Ortopédica, Cali, Colombia.,Universidad ICESI, Cali, Colombia.,Hospital Universitario del Valle, Departamento de Cirugía Ortopédica, Cali, Colombia
| | - Rubén Darío Hernández
- Hospital Universitario del Valle, Departamento de Cirugía Ortopédica, Cali, Colombia
| | - Mauricio Torres
- Fundación Valle del Lili, Departamento de Cirugía Ortopédica, Cali, Colombia.,Universidad ICESI, Cali, Colombia
| |
Collapse
|
50
|
Xu H, Liu Y, Li Y, Luo W, Liu Z, Jian Y. Therapeutic Mechanism of Chinese Medicine on the Healing of Early and Middle Fractures in Rabbits Under the Expression Level of Bone Morphogenetic Protein-2 (BMP-2) in Bone Tissue. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.2857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In order to explore the therapeutic mechanism of Chinese medicine on the healing of rabbits early and middle fractures, a rabbit fracture model was established in this study. The study was divided into several groups, i.e., treatment group (TG) (fed with Chinese medicine Capsule) and
control group (CG) (fed with normal saline (NS)). The materials were collected at 1, 3, and 5 weeks after the start of the experiment for analysis. The experiment content included: callus Hematoxylin-Eosin staining (HE staining); Bone Morphogenetic protein-2 (BMP-2) protein level detection;
Type I and type II bone collagen (BC) detection; and serum biochemical factors detection. The experimental results showed that the formation of callus in the TG was better than in the CG; the BMP-2 protein expression level in the TG was higher than in the CG, and there were statistically significant
differences (SSDs); the type I and type II BC levels in the TG were higher than the CG, there were SSDs; the levels of serum calcium (SC), phosphorus ion (PI), and alkaline phosphatase (ALP) in the TG were also higher than in the CG, and there were SSDs.
Collapse
Affiliation(s)
- Hegui Xu
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, 550002, Guiyang, Guizhou, China
| | - Yang Liu
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, 550002, Guiyang, Guizhou, China
| | - Yuxiong Li
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, 550002, Guiyang, Guizhou, China
| | - Wenbing Luo
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, 550002, Guiyang, Guizhou, China
| | - Zhenyang Liu
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, 550002, Guiyang, Guizhou, China
| | - Yuekui Jian
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, 550002, Guiyang, Guizhou, China
| |
Collapse
|