1
|
Wang X, Stefanello ST, Shahin V, Qian Y. From Mechanoelectric Conversion to Tissue Regeneration: Translational Progress in Piezoelectric Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2417564. [PMID: 40434211 DOI: 10.1002/adma.202417564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 05/03/2025] [Indexed: 05/29/2025]
Abstract
Piezoelectric materials, capable of converting mechanical stimuli into electrical signals, have emerged as promising tools in regenerative medicine due to their potential to stimulate tissue repair. Despite a surge in research on piezoelectric biomaterials, systematic insights to direct their translational optimization remain limited. This review addresses the current landscape by bridging fundamental principles with clinical potential. The biomimetic basis of piezoelectricity, key molecular pathways involved in the synergy between mechanical and electrical stimulation for enhanced tissue regeneration, and critical considerations for material optimization, structural design, and biosafety is discussed. More importantly, the current status and translational quagmire of mechanisms and applications in recent years are explored. A mechanism-driven strategy is proposed for the therapeutic application of piezoelectric biomaterials for tissue repair and identify future directions for accelerated clinical applications.
Collapse
Affiliation(s)
- Xinyu Wang
- National Center for Orthopaedics, Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| | - Sílvio Terra Stefanello
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149, Münster, Germany
| | - Victor Shahin
- Institute of Physiology II, University of Münster, Robert-Koch-Str. 27b, 48149, Münster, Germany
| | - Yun Qian
- National Center for Orthopaedics, Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 200233, Shanghai, China
| |
Collapse
|
2
|
Wu Y, Chen L, Pi D, Cui J, Liang Y, Wu P, Ouyang M, Zuo Q. Saikosaponin A induces cellular senescence in triple-negative breast cancer by inhibiting the PI3K/Akt signalling pathway. Front Pharmacol 2025; 16:1532579. [PMID: 40351423 PMCID: PMC12062077 DOI: 10.3389/fphar.2025.1532579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Background Breast cancer has now become the most prevalent cancer worldwide. Existing therapeutic agents are generally accompanied by significant side effects. Here, we highlight Saikosaponin A (SSA), a promising natural metabolite characterized by low toxicity, demonstrating significant efficacy against breast cancer through the induction of cellular senescence. Methods The antitumor property of SSA was determined via MTT colorimetric assay, 5-ethynyl-2'-deoxyuridine (EdU) incorporation assay, colony formation, and propidium iodide (PI) staining in vitro, as well as xenograft in vivo model. A network approach was used to predict potential targets of SSA reevant for a potential anti-tumor effect and verified through senescence-associated β-galactosidase (SA-β-gal), flow-cytometry analysis, RT-PCR, Western blotting, and immuno-histochemistry assay. Results SSA significantly suppressed proliferation and triggered cell cycle arrest of SUM159PT and MDA-MB-231 cells. Revealed by network analysis, cellular senescence, and phosphatidylinositol-3-kinase (PI3K)/Akt signaling pathway were implemented in the anti-tumor effects of SSA. SSA-stimulated senescence was associated with increased ROS production, distinct senescence-associated secretory phenotype (SASP), and restricted PI3K/Akt signaling, as well as p21 and p53 accumulation. Furthermore, SSA displayed inhibitory effects on tumor growth with minimal toxicity in animal studies, accompanied by activated biomarkers of cellular senescence and decreased expression of p-Akt and p-PI3K. Conclusion Taken together, based on the preliminary results of network analysis and further experimental validation, this study revealed that SSA significantly induced cell cycle arrest and senescence, and the inhibition of ROS-mediated PI3K/Akt pathway may be the potential mechanism in this process.
Collapse
Affiliation(s)
- Yingchao Wu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Liushan Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China
| | - Dajin Pi
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jiaqi Cui
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yuqi Liang
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China
| | - Peng Wu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Mingzi Ouyang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Qian Zuo
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Breast, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Academy of Traditional Chinese Medicine, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Dos Santos RL, Ahmed A, Hunn BE, Addison AE, Marques DW, Bruce KA, Martin JR. Oxidation-responsive, settable bone substitute composites for regenerating critically-sized bone defects. Biomater Sci 2025; 13:1975-1992. [PMID: 40012338 PMCID: PMC11877281 DOI: 10.1039/d4bm01345j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Critically-sized bone defects that cannot spontaneously heal on their own remain a significant problem in the clinic. Synthetic polymeric implants are promising therapies for improving bone healing as they are highly tunable and avoid the potential complications associated with autologous bone grafts. However, biostable implants such as poly(methyl methacrylate) (PMMA) suffer from numerous shortcomings including negligible biodegradability and limited osseointegration with bone. Hydrolytically-degradable polymeric implants such as poly(caprolactone) (PCL) or poly(lactic-co-glycolic acid) (PLGA) have shown promise facilitating bone growth before being resorbed, but matching the degradation rate of these polyesters with the rate of bone regeneration continues to be an engineering challenge. To address these limitations with current synthetic bone implant materials, cell-degradable polymer/hydroxyapatite composites were developed as in situ-curing bone substitutes. The polymeric component was formulated from a thioketal (TK) dithiol linker and a tri-functional epoxy to facilitate rapid crosslinking upon deployment. To enable biologically-responsive implant resorption, the TK unit is specifically cleaved by cell-produced reactive oxygen species (ROS). TK bone substitutes possessed tunable curing and mechanical properties, were selectively degraded in dose-dependent concentrations of ROS, were non-cytotoxic, and demonstrated significantly greater bone regeneration capacity than PMMA in a critically-sized rat skull defect model. These combined results highlight the therapeutic potential of cell-degradable bone void fillers compared against conventional polymeric bone implants.
Collapse
Affiliation(s)
| | - Ardeena Ahmed
- Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA.
| | - Brooke E Hunn
- Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA.
| | - Adolphus E Addison
- Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA.
| | - Dylan W Marques
- Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA.
| | - Karina A Bruce
- Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA.
| | - John R Martin
- Biomedical Engineering, University of Cincinnati, Cincinnati, Ohio, USA.
| |
Collapse
|
4
|
Huang Q, Qu Y, Tang M, Lan K, Zhang Y, Chen S, Li W, Gu L. ROS-responsive hydrogel for bone regeneration: Controlled dimethyl fumarate release to reduce inflammation and enhance osteogenesis. Acta Biomater 2025; 195:183-200. [PMID: 39956305 DOI: 10.1016/j.actbio.2025.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/17/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Large bone defects, often arising from trauma or infection, pose a considerable therapeutic challenge due to their limited capacity for spontaneous healing, thus requiring bone graft materials for effective reparative procedures. The persistence of inflammation and elevated levels of reactive oxygen species (ROS) within these defect sites significantly impede bone regeneration process. Addressing this, an injectable hydrogel system with ROS-responsive functionality is developed, specifically tailored to the high ROS microenvironment characteristic of bone defects. This system incorporates hyaluronic acid functionalized with dopamine to introduce catechol moieties, and employs 4-formylphenylboronic acid as a crosslinking agent to form a dynamic hydrogel matrix (HAC) with carboxymethyl chitosan. The HAC hydrogel serves as a carrier for dimethyl fumarate (DMF), a compound with established anti-inflammatory and antioxidant effects, enabling its controlled release in response to ROS levels. Herein, we investigated the physicochemical properties of DMF loaded hydrogel (DHAC) by microstructure observation, in vitro degradation assay, self-healing test, injectability experiments, DMF drug release assay. Meanwhile, we systematically investigated its effects on inflammation, intracellular ROS, and osteogenesis. Consequently, the DHAC significantly reduced pro-inflammatory cytokines secreted by RAW264.7 cells and scavenged intracellular ROS in MC3T3 cells. This effect was accompanied by an augmentation in the osteogenic potential of MC3T3 cells and a promotion in the repair of cranial defects in rats. The DHAC, which exhibits anti-inflammatory, antioxidant, and osteogenic activity, hold great potential as an effective strategy for the management of large bone defects. STATEMENT OF SIGNIFICANCE: Here, a novel dimethyl fumarate-loaded ROS-responsive hydrogel system was developed for effective treatment of large bone defects. Our findings demonstrated that the hydrogel not only promotes bone regeneration but also controls inflammation, addressing two critical challenges in bone healing. Comprehensive evaluations show significant improvements in bone formation and reduction of pro-inflammatory cytokines in animal models. Additionally, the hydrogel exhibits excellent reactive oxygen species scavenging ability, effectively modulating oxidative stress in the bone defect microenvironment. Findings suggest the hydrogel system may serve as a promising therapeutic strategy for clinical management of critical-sized bone defects.
Collapse
Affiliation(s)
- Qiuxia Huang
- Hospital of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510080, China
| | - Yang Qu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510080, China
| | - Mengchen Tang
- Hospital of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510080, China
| | - Kaiwen Lan
- Hospital of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510080, China
| | - Yilin Zhang
- Hospital of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510080, China
| | - Sishi Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510080, China
| | - Weichang Li
- Hospital of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510080, China.
| | - Lisha Gu
- Hospital of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510055, China; Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong, Guangzhou 510080, China.
| |
Collapse
|
5
|
Jiménez-Ortega RF, López-Pérez TV, Becerra-Cervera A, Aparicio-Bautista DI, Patiño N, Salas-Martínez G, Salmerón J, Velázquez-Cruz R, Rivera-Paredez B. Impact of the dietary antioxidant index on bone mineral density gain among mexican adults: a prospective study. Arch Osteoporos 2025; 20:38. [PMID: 40067600 PMCID: PMC11897103 DOI: 10.1007/s11657-025-01518-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/22/2025] [Indexed: 03/15/2025]
Abstract
In the Mexican population, low dietary antioxidant intake (DAI) is associated with reduced bone mineral density (BMD). A decline in DAI over time further contributes to BMD loss, particularly at the total hip, femoral neck, and lumbar spine, with a more pronounced effect in women over 45 years old. PURPOSE Bone remodeling, balancing resorption and formation, is crucial for bone health. Aging shifts this balance, reducing BMD and increasing osteoporosis risk. Reactive oxygen species (ROS) contribute to bone loss through oxidative stress. Antioxidants may help mitigate this damage, but their impact on BMD in populations with inadequate nutrient intake, like Mexicans, needs to be better understood. This study explores the association between DAI changes and BMD in a Mexican population. METHODS Data were sourced from the Health Worker Cohort Study (HWCS), including 1,318 participants (aged ≥ 20) with BMD measurements and complete dietary information at two time points. The study employed a longitudinal design was used, whit data from two waves of the study (2010-2012 and 2017-2019), providing a median follow-up time of 6.4 years for men and 6.8 years for women. Dietary antioxidant intake was assessed using a validated Food Frequency Questionnaire for the Mexican diet. BMD was measured at the femoral neck, total hip, and lumbar spine using dual-energy X-ray absorptiometry (DEXA). Fixed-effects regression models were applied to analyze the association between DAI and BMD at different sites, adjusting for time-varying covariates. RESULTS Changes in DAI scores were associated with lower BMD at various sites. Each unit decrease in DAI over time was associated with a BMD loss of -0.002,-0.004 g/cm2 at the total hip, femoral neck, and lumbar spine. Notable declines were observed in women, particularly those over 45 years old, where specific antioxidant components, like zinc, magnesium, and selenium, were linked to lower BMD. CONCLUSION This study underscores the role of reduced dietary antioxidant intake in contributing lower BMD, particularly among older adults. Diets low in antioxidant may increase the risk of osteoporosis, especially in populations with insufficient nutrient intake.
Collapse
Affiliation(s)
- Rogelio F Jiménez-Ortega
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Mexico City, Mexico
- Clínica Integral Universitaria (CIU), Universidad Estatal del Valle de Ecatepec (UNEVE), Ecatepec de Morelos, 55210, Mexico, Mexico
| | - Tania V López-Pérez
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Mexico City, Mexico
- Consejo Nacional de Humanidades, Ciencias y Tecnologías, (CONAHCYT), 03940, Mexico City, Mexico
| | - Adriana Becerra-Cervera
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Mexico City, Mexico
- Consejo Nacional de Humanidades, Ciencias y Tecnologías, (CONAHCYT), 03940, Mexico City, Mexico
| | - Diana I Aparicio-Bautista
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Mexico City, Mexico
| | - Nelly Patiño
- Unidad de Citometría de Flujo (UCiF), Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Mexico City, Mexico
| | - Guadalupe Salas-Martínez
- Laboratorio de Inmunogenómica y Enfermedades Metabólicas, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Mexico City, Mexico
| | - Jorge Salmerón
- Centro de Investigación en Políticas, Población y Salud (CIPPS), Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), 04510, Mexico City, Mexico
| | - Rafael Velázquez-Cruz
- Laboratorio de Genómica del Metabolismo Óseo, Instituto Nacional de Medicina Genómica (INMEGEN), 14610, Mexico City, Mexico.
| | - Berenice Rivera-Paredez
- Centro de Investigación en Políticas, Población y Salud (CIPPS), Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), 04510, Mexico City, Mexico.
| |
Collapse
|
6
|
Hazrati P, Azadi A, Fekrazad S, Wang HL, Fekrazad R. The effect of photobiomodulation therapy on fracture healing: a systematic review and meta-analysis of animal studies. Lasers Med Sci 2025; 40:121. [PMID: 40016554 DOI: 10.1007/s10103-025-04376-0] [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: 12/06/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025]
Abstract
This systematic review and meta-analysis aimed to evaluate the impact of photobiomodulation therapy (PBMT) on fracture healing in animal models. Following PRISMA guidelines, an electronic search was conducted in PubMed/MEDLINE, Scopus, Embase, and Web of Science databases without date or language restrictions. Animal studies evaluating the effect of PBMT on the healing of complete fractures were included. SYRCLE's risk of bias assessment tool was used for quality appraisal. Meta-analysis and sensitivity analysis were performed for selected outcome measures using Stata version 16, with a significance level of 0.05. Of 1,656 studies, 27 met eligibility criteria. Rabbits and rats were used in 17 and 10 studies, respectively. The tibia was the most common site of fracture, followed by the femur, mandible, and radius. The most frequently used emitters were 780 nm LASER, followed by 808 nm and 830 nm LASER. LEDs were used in comparison to LASERs in three studies and solely in one study. The most frequent energy density and power density were 4 J/cm2 and 100 mW/cm2, respectively. Radiography, histology, mechanical testing, and spectroscopy were the most common assessment methods of fracture healing. While most studies reported PBMT's positive effect on fracture healing, meta-analysis found no significant impact on maximum fracture force or Raman peaks of hydroxyapatite, indicating no significant influence on mineralization (P > 0.05). Although PBMT shows potential for enhancing fracture healing in animal models, meta-analysis showed that it has no effect on maximum force of fracture or Raman peaks of hydroxyapatite. Registration: The protocol of this systematic review was registered on PROSPERO with the ID CRD42024514398.
Collapse
Affiliation(s)
- Parham Hazrati
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Ali Azadi
- Dentofacial Deformities Research Center, Research Institute for Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Sepehr Fekrazad
- School of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
- Radiation Sciences Research Center (RSRC), Aja University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Hom-Lay Wang
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Reza Fekrazad
- Radiation Sciences Research Center (RSRC), Aja University of Medical Sciences, Tehran, Islamic Republic of Iran.
- International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Islamic Republic of Iran.
| |
Collapse
|
7
|
Yang Y, Yao Z, Sun Y, Nie Y, Zhang Y, Li Z, Luo Z, Zhang W, Wang X, Du Y, Zhang W, Qin L, Sang H, Lai Y. 3D-printed manganese dioxide incorporated scaffold promotes osteogenic-angiogenic coupling for refractory bone defect by remodeling osteo-regenerative microenvironment. Bioact Mater 2025; 44:354-370. [PMID: 39539517 PMCID: PMC11558641 DOI: 10.1016/j.bioactmat.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/01/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
The treatment of refractory bone defects is a major clinical challenge, especially in steroid-associated osteonecrosis (SAON), which is characterized by insufficient osteogenesis and angiogenesis. Herin, a microenvironment responsiveness scaffold composed of poly-L-lactide (PLLA), and manganese dioxide (MnO2) nanoparticles is designed to enhance bone regeneration by scavenging endogenous reactive oxygen species (ROS) and modulating immune microenvironment in situ. A catalase-like catalytic reaction between MnO2 and endogenous hydrogen peroxide (H2O2) generated at the bone defect area, which typically becomes acidic and ROS-rich, triggers on-demand release of oxygen and Mn2+, significantly ameliorating inflammatory response by promoting M2-type polarization of macrophages, reprograming osteoimmune microenvironment conducive to angiogenesis and osteogenesis. Furthermore, the fundamental mechanisms were explored through transcriptome sequencing analysis, revealing that PLLA/MnO2 scaffolds (PMns) promote osteogenic differentiation by upregulating the TGF-β/Smad signaling pathway in human bone marrow mesenchymal stem cells (hBMSCs). Overall, the PMns exhibit superior immunomodulatory, excellent osteogenic-angiogenic properties and promising candidates as bone graft substitutes for therapy clinical refractory bone defects.
Collapse
Affiliation(s)
- Yipei Yang
- Department of Orthopedic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510500, China
| | - Zhenyu Yao
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yuanyi Sun
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yangyi Nie
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yuanchi Zhang
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ziyue Li
- Department of Medicine Ultrasonics, Nanfang Hospital, Southern Medical University, Guangzhou, 510500, China
| | - Zhiheng Luo
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wenjing Zhang
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiao Wang
- Department of Orthopedic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510500, China
| | - Yuhan Du
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wei Zhang
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ling Qin
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Musculoskeletal Research Laboratory, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Hong Kong SAR, 999077, China
- CAS-HK Joint Lab of Biomaterials, Shenzhen, 518055, China
| | - Hongxun Sang
- Department of Orthopedic Surgery, Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
- The Third School of Clinical Medicine, Southern Medical University, Guangzhou, 510500, China
| | - Yuxiao Lai
- Centre for Translational Medicine Research & Development, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- CAS-HK Joint Lab of Biomaterials, Shenzhen, 518055, China
| |
Collapse
|
8
|
Rasoulian B, Poormoghadam D, Hoveizi E, Rezayat SM, Tavakol S. Small but mighty: nanoemulsion particle size dictates bone regeneration potential of FTY720. NANOSCALE 2025; 17:2091-2104. [PMID: 39652087 DOI: 10.1039/d4nr02884h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The burgeoning field of nano-bone regeneration is yet to establish a definitive optimal particle size for nanocarriers. This study investigated the impacts of nanocarrier's particle size on the bone regeneration efficacy of fingolimod (FTY720)-loaded nanoemulsions. Two distinct particle sizes (60 and 190 nm, designated as NF60 and NF190, respectively) were produced using low-energy and high-energy emulsion techniques, maintaining a consistent surfactant, co-surfactant, and oil. In vitro studies using rat mesenchymal stem cells revealed that both NF60 and NF190 exhibited cell viability and reduced lactate dehydrogenase. Interestingly, NF60 demonstrated superior antioxidant properties, significantly reducing nitric oxide and intracellular reactive oxygen species (ROS) levels compared to NF190. Furthermore, NF60 significantly enhanced ALP activity and calcium deposition during osteogenic differentiation, indicating its potential to promote the early stages of bone formation. In vivo studies using a rat calvarial bone defect model demonstrated that both NF60 and NF190 significantly upregulated the expression of key osteogenic genes, including Runx2, Col, ALP, OCN, and BMP2. Notably, NF60 induced significantly higher expression of Runx2 and BMP2. X-ray and histological investigations revealed significantly improved bone regeneration in the NF60 group, highlighting the superior bone healing potential of smaller FTY720 nanoemulsions, without infiltration of inflammatory cells. The smaller particle size demonstrated superior antioxidant properties, enhanced osteogenic differentiation, and improved bone regeneration, suggesting smaller nanoparticles, with their larger surface area, accelerated drug release rate, and lower viscosity, interact more effectively with cells, leading to increased and effective drug delivery and cellular uptake. Findings highlight the importance of nanocarrier size in optimizing drug delivery for bone tissue engineering applications.
Collapse
Affiliation(s)
- Bita Rasoulian
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- School of biomedical Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Delaram Poormoghadam
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Elham Hoveizi
- Department of biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Seyed Mahdi Rezayat
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Nanomedicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shima Tavakol
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Research and Develoment Departement, Tavakol BioMimetich Technologies Co (TMBT), Tehran, Iran
| |
Collapse
|
9
|
Gerini G, Traversa A, Cece F, Cassandri M, Pontecorvi P, Camero S, Nannini G, Romano E, Marampon F, Venneri MA, Ceccarelli S, Angeloni A, Amedei A, Marchese C, Megiorni F. Deciphering the Transcriptional Metabolic Profile of Adipose-Derived Stem Cells During Osteogenic Differentiation and Epigenetic Drug Treatment. Cells 2025; 14:135. [PMID: 39851564 PMCID: PMC11763738 DOI: 10.3390/cells14020135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 01/26/2025] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) are commonly employed in clinical treatment for various diseases due to their ability to differentiate into multi-lineage and anti-inflammatory/immunomodulatory properties. Preclinical studies support their use for bone regeneration, healing, and the improvement of functional outcomes. However, a deeper understanding of the molecular mechanisms underlying ASC biology is crucial to identifying key regulatory pathways that influence differentiation and enhance regenerative potential. In this study, we employed the NanoString nCounter technology, an advanced multiplexed digital counting method of RNA molecules, to comprehensively characterize differentially expressed transcripts involved in metabolic pathways at distinct time points in osteogenically differentiating ASCs treated with or without the pan-DNMT inhibitor RG108. In silico annotation and gene ontology analysis highlighted the activation of ethanol oxidation, ROS regulation, retinoic acid metabolism, and steroid hormone metabolism, as well as in the metabolism of lipids, amino acids, and nucleotides, and pinpointed potential new osteogenic drivers like AOX1 and ADH1A. RG108-treated cells, in addition to the upregulation of the osteogenesis-related markers RUNX2 and ALPL, showed statistically significant alterations in genes implicated in transcriptional control (MYCN, MYB, TP63, and IRF1), ethanol oxidation (ADH1C, ADH4, ADH6, and ADH7), and glucose metabolism (SLC2A3). These findings highlight the complex interplay of the metabolic, structural, and signaling pathways that orchestrate osteogenic differentiation. Furthermore, this study underscores the potential of epigenetic drugs like RG108 to enhance ASC properties, paving the way for more effective and personalized cell-based therapies for bone regeneration.
Collapse
Affiliation(s)
- Giulia Gerini
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Alice Traversa
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy; (A.T.); (S.C.)
| | - Fabrizio Cece
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Matteo Cassandri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Paola Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Simona Camero
- Department of Life Sciences, Health and Health Professions, Link Campus University, 00165 Rome, Italy; (A.T.); (S.C.)
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy; (G.N.); (A.A.)
| | - Enrico Romano
- Department of Sense Organs, Sapienza University of Rome, 00161 Rome, Italy;
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Mary Anna Venneri
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Antonio Angeloni
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50121 Florence, Italy; (G.N.); (A.A.)
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| | - Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy; (G.G.); (F.C.); (M.C.); (P.P.); (M.A.V.); (S.C.); (A.A.); (C.M.)
| |
Collapse
|
10
|
Ehlen Q, Costello JP, Mirsky NA, Slavin BV, Parra M, Ptashnik A, Nayak VV, Coelho PG, Witek L. Treatment of Bone Defects and Nonunion via Novel Delivery Mechanisms, Growth Factors, and Stem Cells: A Review. ACS Biomater Sci Eng 2024; 10:7314-7336. [PMID: 39527574 PMCID: PMC11632667 DOI: 10.1021/acsbiomaterials.4c01279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Bone nonunion following a fracture represents a significant global healthcare challenge, with an overall incidence ranging between 2 and 10% of all fractures. The management of nonunion is not only financially prohibitive but often necessitates invasive surgical interventions. This comprehensive manuscript aims to provide an extensive review of the published literature involving growth factors, stem cells, and novel delivery mechanisms for the treatment of fracture nonunion. Key growth factors involved in bone healing have been extensively studied, including bone morphogenic protein (BMP), vascular endothelial growth factor (VEGF), and platelet-derived growth factor. This review includes both preclinical and clinical studies that evaluated the role of growth factors in acute and chronic nonunion. Overall, these studies revealed promising bridging and fracture union rates but also elucidated complications such as heterotopic ossification and inferior mechanical properties associated with chronic nonunion. Stem cells, particularly mesenchymal stem cells (MSCs), are an extensively studied topic in the treatment of nonunion. A literature search identified articles that demonstrated improved healing responses, osteogenic capacity, and vascularization of fractures due to the presence of MSCs. Furthermore, this review addresses novel mechanisms and materials being researched to deliver these growth factors and stem cells to nonunion sites, including natural/synthetic polymers and bioceramics. The specific mechanisms explored in this review include BMP-induced osteoblast differentiation, VEGF-mediated angiogenesis, and the role of MSCs in multilineage differentiation and paracrine signaling. While these therapeutic modalities exhibit substantial preclinical promise in treating fracture nonunion, there remains a need for further research, particularly in chronic nonunion and large animal models. This paper seeks to identify such translational hurdles which must be addressed in order to progress the aforementioned treatments from the lab to the clinical setting.
Collapse
Affiliation(s)
- Quinn
T. Ehlen
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Joseph P. Costello
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Nicholas A. Mirsky
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Blaire V. Slavin
- University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - 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
| | - Albert Ptashnik
- Biomaterials
Division, NYU Dentistry, New York, New York 10010, United States
| | - Vasudev Vivekanand Nayak
- Department
of Biochemistry and Molecular Biology, University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Paulo G. Coelho
- Department
of Biochemistry and Molecular Biology, University
of Miami Miller School of Medicine, Miami, Florida 33136, United States
- Division
of Plastic Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, United States
| | - Lukasz Witek
- Biomaterials
Division, NYU Dentistry, New York, New York 10010, United States
- Department
of Biomedical Engineering, NYU Tandon School
of Engineering, Brooklyn, New York 11201, United States
- Hansjörg
Wyss Department of Plastic Surgery, NYU
Grossman School of Medicine, New
York, New York 10016, United States
| |
Collapse
|
11
|
Li G, Wu J, Cheng X, Pei X, Wang J, Xie W. Nanoparticle-Mediated Gene Delivery for Bone Tissue Engineering. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2408350. [PMID: 39623813 DOI: 10.1002/smll.202408350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/13/2024] [Indexed: 03/17/2025]
Abstract
Critical-sized bone defects represent an urgent clinical problem, necessitating innovative treatment approaches. Gene-activated grafts for bone tissue engineering have emerged as a promising solution. However, traditional gene delivery methods are constrained by limited osteogenic efficacy and safety concerns. Recently, organic and inorganic nanoparticle (NP) vectors have attracted significant attention in bone tissue engineering for their safe, stable, and controllable gene delivery. Targeted gene delivery guided by insights into bone healing mechanisms, coupled with the multifunctional design of NPs, is crucial for enhancing therapeutic outcomes. Here, the theoretical foundations underlying NP-mediated gene therapy for enhancing bone healing across different histological stages are elucidated. Furthermore, the distinct attributes of functionalized NP vectors are discussed, and cutting-edge strategies aimed at optimizing gene delivery efficiency throughout the therapeutic process are highlighted. Additionally, the review addresses the unresolved challenges and prospects of this technology. This review may contribute to the continued development and clinical application of NP-mediated gene delivery for treating critical-sized bone defects.
Collapse
Affiliation(s)
- Guangzhao Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiaxin Wu
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xinting Cheng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, China
| | - Xibo Pei
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jian Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Wenjia Xie
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases & Department of Prosthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| |
Collapse
|
12
|
Peng B, Feng Z, Yang A, Liu J, He J, Xu L, Tian C, Sheng X, Wang Y, Chen R, Wang X, Ren X, Geng B, Xia Y. TIMP1 regulates ferroptosis in osteoblasts by inhibiting TFRC ubiquitination: an in vitro and in vivo study. Mol Med 2024; 30:226. [PMID: 39578773 PMCID: PMC11585138 DOI: 10.1186/s10020-024-01000-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/14/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND In clinical practice, alterations in the internal environment of type 2 diabetes can significantly affect bone quality. While the increased risk of fractures among diabetic patients is well-established, the precise mechanisms by which hyperglycemia influences bone quality remain largely unclear. METHODS Western blotting, immunohistochemistry (IHC), and micro-CT were used to examine ferroptosis-related protein expression and bone morphology changes in the bone tissues of type 2 diabetic mice. The CCK8 assay determined the optimal conditions for inducing ferroptosis in osteoblasts by high glucose and high fat (HGHF). Ferroptosis phenotypes in osteoblasts were analyzed using flow cytometry, Western blotting, and two-photon laser confocal microscopy. Transcriptomic sequencing of the control and HGHF groups, followed by bioinformatic analysis, identified and validated key genes. TIMP1 was knocked down in osteoblasts to assess its impact on ferroptosis, while TFRC expression was inhibited and activated to verify the role of TIMP1 in regulating ferroptosis through TFRC. The therapeutic effect of TIMP1 inhibition on osteoporosis was evaluated in a type 2 diabetic mouse model. RESULTS The expression of TIMP1 is increased in type 2 diabetic osteoporosis. In vitro, TIMP1 knockout inhibited ferroptosis in osteoblasts induced by high glucose and high fat (HGHF). However, overexpression of TFRC reversed the ferroptosis inhibition caused by TIMP1 knockout. Suppression of TIMP1 expression alleviated the progression of osteoporosis in type 2 diabetic mice. Mechanistic studies suggest that TIMP1 regulates HGHF-induced ferroptosis in osteoblasts through TFRC. CONCLUSION This study demonstrates that TIMP1 expression is increased during type 2 diabetic osteoporosis and that TIMP1 promotes ferroptosis in osteoblasts by regulating TFRC. These findings suggest that TIMP1 is a promising novel therapeutic target for type 2 diabetic osteoporosis.
Collapse
Grants
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 82060405, 82360436 The National Natural Science Foundation of China
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 2021-RC-102 Lanzhou Science and Technology Plan Program
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- 22JR5RA943, 22JR5RA956, 23JRRA1500, 22JR11RA057 Natural Science Foundation of Gansu Province
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
- CY2021-MS-A07, CY2022-MS-A19, CY2021-BJ-A13,CY2023-BJ-13 Cuiying Scientific and Technological Innovation Program of Lanzhou University Second Hospital
Collapse
Affiliation(s)
- Bo Peng
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Zhiwei Feng
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Ao Yang
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Jinmin Liu
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Jinwen He
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Lihu Xu
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Cong Tian
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Xiaoyun Sheng
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Yaobin Wang
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Rongjin Chen
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Xingwen Wang
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Xiaojun Ren
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China
| | - Bin Geng
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China.
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China.
| | - Yayi Xia
- Department of Orthopaedics, The Second Hospital of Lanzhou University, #82 Cuiyingmen, Lanzhou, Gansu, 730030, People's Republic of China.
- Intelligent Orthopedics Industry Technology Center of Gansu Province, Lanzhou, Gansu, China.
- Orthopaedic Clinical Research Center of Gansu Province, Lanzhou, Gansu, China.
| |
Collapse
|
13
|
Gomez Gramajo F, Rivoira MA, Rodríguez V, Vargas G, Vera Mesones R, Zago MP, Boccaccini AR, Gorustovich A. Lithium-containing 45S5 Bioglass-derived glass-ceramics have antioxidant activity and induce new bone formation in a rat preclinical model of type 1 diabetes mellitus. Biomed Mater 2024; 20:015006. [PMID: 39564894 DOI: 10.1088/1748-605x/ad8c8b] [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: 05/19/2024] [Accepted: 10/29/2024] [Indexed: 11/21/2024]
Abstract
Diabetes mellitus (DM) has been associated with complications that affect the skeletal system, such as alterations in bone repair, osteoporosis, and an increased risk of fractures. In this context, the use of biomaterials able to promote osteogenic differentiation and, at the same time, limit the oxidative stress induced by DM offers a novel perspective to ensure the repair of diabetic bone tissue. Since lithium (Li) has been recently identified as a biologically active ion with osteogenic and antioxidant properties, the localized and controlled release of Li ions from bioactive glass-ceramic materials represents a promising therapeutic alternative for the treatment of bone lesions in DM. Thus, the aim of this study was to evaluate the potential osteogenic and antioxidant effects of glass-ceramic microparticles derived from a 45S5-type bioactive glass (Bioglass) containing (% by weight) 45% SiO2, 24.5% Na2O, 24.5% CaO, and 6% P2O5, in which Na2O was partially substituted by 5% of Li2O (45S5.5Li), in an experimental model of type 1 DM (DM1). The results obtained demonstrate, for the first time, that both 45S5 and 45S5.5Li glass-ceramic microparticles possess antioxidant activity and stimulate bone formationin vivoboth under physiological conditions and under experimental DM1 in rats. In this sense, they would have potential application as inorganic osteogenic agents in different strategies of bone tissue regenerative medicine.
Collapse
Affiliation(s)
- Fátima Gomez Gramajo
- Cátedra de Biología del Desarrollo, Facultad de Ciencias Naturales, Universidad Nacional de Salta, Salta, Argentina
| | - María A Rivoira
- Laboratorio 'Dr Fernando Cañas', Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| | - Valeria Rodríguez
- Laboratorio 'Dr Fernando Cañas', Cátedra de Bioquímica y Biología Molecular, Facultad de Ciencias Médicas, INICSA (CONICET-Universidad Nacional de Córdoba), Córdoba, Argentina
| | - Gabriela Vargas
- Cátedra de Biología del Desarrollo, Facultad de Ciencias Naturales, Universidad Nacional de Salta, Salta, Argentina
| | - Rosa Vera Mesones
- Cátedra de Biología del Desarrollo, Facultad de Ciencias Naturales, Universidad Nacional de Salta, Salta, Argentina
| | - María P Zago
- Unidad de Conocimiento Traslacional Hospitalario, CONICET-Hospital Público Materno Infantil, Salta, Argentina
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Alejandro Gorustovich
- Laboratorio de Biomateriales, Grupo Interdisciplinario en Materiales-IESIING, Universidad Católica de Salta, grupo vinculado al INTECIN UBA-CONICET, Salta, Argentina
| |
Collapse
|
14
|
Kabir A, B M, A N, Selvaraj V, Sudhakar S. Protein Nano Coop Complexes Promote Fracture Healing and Bone Regeneration in a Zebrafish Osteoporosis Model. Biomacromolecules 2024; 25:7237-7248. [PMID: 39449233 DOI: 10.1021/acs.biomac.4c00931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Nanotherapeutic techniques are becoming increasingly important in the treatment of bone disorders owing to their targeted drug delivery. This study formulates zein nano coop composites containing chimeric antioxidants (ascorbic acid, luteolin, resveratrol, and coenzyme Q) (AZN) and evaluates its application in bone regeneration using osteoblasts and a Zebrafish osteoporosis model. In vitro experiments with human osteoblast-like MG63 cells showed enhancement of bone mineralization and regeneration. It further exhibited high biocompatibility in Zebrafish larvae, with increased calcium/phosphorus deposition and upregulation of osteogenic genes. The study has unequivocally demonstrated the potential of AZN in bone regeneration and fracture healing in both normal and osteoporosis models, underscoring the significance of this research. Further investigations using higher animal models are warranted to expand on these findings. The impact of this research seems far-reaching, with the possible development of new, effective, and safe treatment options for osteoporosis, addressing the limitations of the currently available treatments.
Collapse
Affiliation(s)
- Anisha Kabir
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Mukilarasi B
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Natarajan A
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Vimalraj Selvaraj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Swathi Sudhakar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| |
Collapse
|
15
|
Sroga GE, Vashishth D. In vivo glycation-interplay between oxidant and carbonyl stress in bone. JBMR Plus 2024; 8:ziae110. [PMID: 39386996 PMCID: PMC11458925 DOI: 10.1093/jbmrpl/ziae110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 06/18/2024] [Accepted: 07/28/2024] [Indexed: 10/12/2024] Open
Abstract
Metabolic syndromes (eg, obesity, type 2 diabetes (T2D), atherosclerosis, and neurodegenerative diseases) and aging, they all have a strong component of carbonyl and reductive-oxidative (redox) stress. Reactive carbonyl (RCS) and oxidant (ROS) stress species are commonly generated as products or byproducts of cellular metabolism or are derived from the environment. RCS and ROS can play a dual role in living organisms. Some RCS and ROS function as signaling molecules, which control cellular defenses against biological and environmental assaults. However, due to their high reactivity, RCS and ROS inadvertently interact with different cellular and extracellular components, which can lead to the formation of undesired posttranslational modifications of bone matrix proteins. These are advanced glycation (AGEs) and glycoxidation (AGOEs) end products generated in vivo by non-enzymatic amino-carbonyl reactions. In this review, metabolic processes involved in generation of AGEs and AGOEs within and on protein surfaces including extracellular bone matrix are discussed from the perspective of cellular metabolism and biochemistry of certain metabolic syndromes. The impact of AGEs and AGOEs on some characteristics of mineral is also discussed. Different therapeutic approaches with the potential to prevent the formation of RCS, ROS, and the resulting formation of AGEs and AGOEs driven by these chemicals are also briefly reviewed. These are antioxidants, scavenging agents of reactive species, and newly emerging technologies for the development of synthetic detoxifying systems. Further research in the area of in vivo glycation and glycoxidation should lead to the development of diverse new strategies for halting the progression of metabolic complications before irreversible damage to body tissues materializes.
Collapse
Affiliation(s)
- Grażyna E Sroga
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Shirley Ann Jackson PhD Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
| | - Deepak Vashishth
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY, United States
- Shirley Ann Jackson PhD Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, United States
- Center for Engineering and Precision Medicine, Rensselaer-Icahn School of Medicine at Mount Sinai, 619 West 54th Street, New York, NY 10019, United States
| |
Collapse
|
16
|
Guo Q, Zhai Q, Ji P. The Role of Mitochondrial Homeostasis in Mesenchymal Stem Cell Therapy-Potential Implications in the Treatment of Osteogenesis Imperfecta. Pharmaceuticals (Basel) 2024; 17:1297. [PMID: 39458939 PMCID: PMC11510265 DOI: 10.3390/ph17101297] [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: 06/20/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
Osteogenesis imperfecta (OI) is a hereditary disorder characterized by bones that are fragile and prone to breaking. The efficacy of existing therapies for OI is limited, and they are associated with potentially harmful side effects. OI is primarily due to a mutation of collagen type I and hence impairs bone regeneration. Mesenchymal stem cell (MSC) therapy is an attractive strategy to take advantage of the potential benefits of these multipotent stem cells to address the underlying molecular defects of OI by differentiating osteoblasts, paracrine effects, or immunomodulation. The maintenance of mitochondrial homeostasis is an essential component for improving the curative efficacy of MSCs in OI by affecting the differentiation, signaling, and immunomodulatory functions of MSCs. In this review, we highlight the MSC-based therapy pathway in OI and introduce the MSC regulation mechanism by mitochondrial homeostasis. Strategies aiming to modulate the metabolism and reduce the oxidative stress, as well as innovative strategies based on the use of compounds (resveratrol, NAD+, α-KG), antioxidants, and nanomaterials, are analyzed. These findings may enable the development of new strategies for the treatment of OI, ultimately resulting in improved patient outcomes.
Collapse
Affiliation(s)
- Qingling Guo
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China;
- Chongqing Key Laboratory of Oral Diseases, Chongqing 401147, China
| | - Qiming Zhai
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China;
- Chongqing Key Laboratory of Oral Diseases, Chongqing 401147, China
| | - Ping Ji
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China;
- Chongqing Key Laboratory of Oral Diseases, Chongqing 401147, China
| |
Collapse
|
17
|
Hammad M, Salma R, Balosso J, Rezvani M, Haghdoost S. Role of Oxidative Stress Signaling, Nrf2, on Survival and Stemness of Human Adipose-Derived Stem Cells Exposed to X-rays, Protons and Carbon Ions. Antioxidants (Basel) 2024; 13:1035. [PMID: 39334694 PMCID: PMC11429097 DOI: 10.3390/antiox13091035] [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: 06/25/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
Some cancers have a poor prognosis and often lead to local recurrence because they are resistant to available treatments, e.g., glioblastoma. Attempts have been made to increase the sensitivity of resistant tumors by targeting pathways involved in the resistance and combining it, for example, with radiotherapy (RT). We have previously reported that treating glioblastoma stem cells with an Nrf2 inhibitor increases their radiosensitivity. Unfortunately, the application of drugs can also affect normal cells. In the present study, we aim to investigate the role of the Nrf2 pathway in the survival and differentiation of normal human adipose-derived stem cells (ADSCs) exposed to radiation. We treated ADSCs with an Nrf2 inhibitor and then exposed them to X-rays, protons or carbon ions. All three radiation qualities are used to treat cancer. The survival and differentiation abilities of the surviving ADSCs were studied. We found that the enhancing effect of Nrf2 inhibition on cell survival levels was radiation-quality-dependent (X-rays > proton > carbon ions). Furthermore, our results indicate that Nrf2 inhibition reduces stem cell differentiation by 35% and 28% for adipogenesis and osteogenesis, respectively, using all applied radiation qualities. Interestingly, the results show that the cells that survive proton and carbon ion irradiations have an increased ability, compared with X-rays, to differentiate into osteogenesis and adipogenesis lineages. Therefore, we can conclude that the use of carbon ions or protons can affect the stemness of irradiated ADSCs at lower levels than X-rays and is thus more beneficial for long-time cancer survivors, such as pediatric patients.
Collapse
Affiliation(s)
- Mira Hammad
- Centre de Recherche sur les Ions, les Matériaux et la Photonique (CIMAP) UMR 6252, University of Caen Normandy, Cedex 04, F-14050 Caen, France
| | - Rima Salma
- Centre de Recherche sur les Ions, les Matériaux et la Photonique (CIMAP) UMR 6252, University of Caen Normandy, Cedex 04, F-14050 Caen, France
| | - Jacques Balosso
- Department of Radiation Oncology, Centre François Baclesse, F-14000 Caen, France
- Advanced Resource Center for HADrontherapy in Europe (ARCHADE), F-14000 Caen, France
| | - Mohi Rezvani
- Swiss Bioscience GmbH, Wagistrasse 27a, CH-8952 Schlieren, Switzerland
| | - Siamak Haghdoost
- Centre de Recherche sur les Ions, les Matériaux et la Photonique (CIMAP) UMR 6252, University of Caen Normandy, Cedex 04, F-14050 Caen, France
- Advanced Resource Center for HADrontherapy in Europe (ARCHADE), F-14000 Caen, France
- Le Laboratoire "Aliments, Bioprocédés, Toxicologie et Environnement (ABTE) UR 4651, ToxEMAC Team, University of Caen Normandy, Cedex 04, F-14050 Caen, France
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-10691 Stockholm, Sweden
| |
Collapse
|
18
|
Yang J, Zhan Z, Li X, Hu M, Zhu Y, Xiao Y, Xu X. Fullerol-reinforced antioxidantive 3D-printed bredigite scaffold for accelerating bone healing. Mater Today Bio 2024; 27:101120. [PMID: 38975240 PMCID: PMC11225861 DOI: 10.1016/j.mtbio.2024.101120] [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: 03/06/2024] [Revised: 05/14/2024] [Accepted: 06/08/2024] [Indexed: 07/09/2024] Open
Abstract
Reactive oxygen species play a vital role in tissue repair, and nonequilibrium of redox homeostasis around bone defect can compromise osteogenesis. However, insufficient antioxidant capacity and weak osteogenic performance remain major obstacles for bone scaffold materials. Herein, integrating the mussel-inspired polydopamine (PDA) coating and 3D printing technologies, we utilized the merits of both osteogenic bredigite and antioxidative fullerol to construct 3D-printed porous, biodegradable acid-buffering, reactive oxygen species (ROS) -scavenging and robust osteogenic bio-scaffold (denoted "FPBS") for in situ bone defect restoration under oxidative stress microenvironment. Initially, fullerol nanoparticles were attached to the surface of the bredigite scaffold via covalently inter-crosslinking with PDA. Upon injury, extracellular ROS capturing triggered the oxidative degradation of PDA, releasing fullerol nanoparticles to enter into cells for further intracellular ROS scavenging. In vitro, FPBS had good biocompatibility and excellent antioxidative capability. Furthermore, FPBS promoted the osteogenesis of stem cells with significant elevation of osteogenic markers. Finally, in vivo implantation of FPBS remarkably enhanced new bone formation in a rat critical calvarial defect model. Overall, with amelioration of the ROS microenvironment of injured tissue and enhancement of osteogenic differentiation of stem cells simultaneously, FPBS may hold great potential towards bone defect repair.
Collapse
Affiliation(s)
- Jielai Yang
- Department of Orthopedics, Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, PR China
| | - Zihang Zhan
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, Zhejiang Province, PR China
| | - Xingchen Li
- Department of Orthopedics, Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, PR China
| | - Mu Hu
- Department of Orthopedics, Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, PR China
| | - Yuan Zhu
- Department of Orthopedics, Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, PR China
| | - Yunchao Xiao
- College of Materials and Textile Engineering, Jiaxing University, Jiaxing, 314001, Zhejiang Province, PR China
| | - Xiangyang Xu
- Department of Orthopedics, Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, 200025, PR China
| |
Collapse
|
19
|
Dadashi Ouranj Z, Hosseini S, Alipour A, Homaeigohar S, Azari S, Ghazizadeh L, Shokrgozar M, Thomas S, Irian S, Shahsavarani H. The potent osteo-inductive capacity of bioinspired brown seaweed-derived carbohydrate nanofibrous three-dimensional scaffolds. MARINE LIFE SCIENCE & TECHNOLOGY 2024; 6:515-534. [PMID: 39219680 PMCID: PMC11358581 DOI: 10.1007/s42995-024-00241-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/11/2024] [Indexed: 09/04/2024]
Abstract
This study aimed to investigate the osteo-inductive capacity of a fucoidan polysaccharide network derived from brown algae on human adipose-derived stem cells (HA-MSCs) for bone regeneration. The physiochemical properties of the scaffold including surface morphology, surface chemistry, hydrophilicity, mechanical stiffness, and porosity were thoroughly characterized. Both in vitro and in vivo measurements implied a superior cell viability, proliferation, adhesion, and osteo-inductive performance of obtained scaffolds compared to using specific osteogenic induction medium with increased irregular growth of calcium crystallites, which mimic the structure of natural bones. That scaffold was highly biocompatible and suitable for cell cultures. Various examinations, such as quantification of mineralization, alkaline phosphatase, gene expression, and immunocytochemical staining of pre-osteocyte and bone markers confirmed that HAD-MSCs differentiate into osteoblasts, even without an osteogenic induction medium. This study provides evidence for the positive relationship and synergistic effects between the physical properties of the decellularized seaweed scaffold and the chemical composition of fucoidan in promoting the osteogenic differentiation of HA-MSCs. Altogether, the natural matrices derived from brown seaweed offers a sustainable, cost-effective, non-toxic bioinspired scaffold and holds promise for future clinical applications in orthopedics.
Collapse
Affiliation(s)
- Zahra Dadashi Ouranj
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, 15719-14911 Iran
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Saadi Hosseini
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Atefeh Alipour
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Shahin Homaeigohar
- School of Science and Engineering, University of Dundee, Dundee, DD1 4HN UK
| | - Shahram Azari
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Leila Ghazizadeh
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Mohammadali Shokrgozar
- Laboratory of Regenerative Medicine and Biomedical Innovations, National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, 13169-43551 Iran
| | - Sabu Thomas
- School of Chemical Sciences, Mahatma Gandhi University, Kottayam, Kerala India
| | - Saeed Irian
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, 15719-14911 Iran
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, 19839-69411 Iran
- Iranian Biological Resource Center, Academic Center for Education, Culture and Research (ACECR), Tehran, 1533734716 Iran
| |
Collapse
|
20
|
Bingül MB, Gul M, Dündar S, Sökmen K, Artas G, Polat ME, Tanrisever M, Ozcan EC. Effect of Different Administered Doses of Capsaicin and Titanium Implant Osseointegration. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1094. [PMID: 39064523 PMCID: PMC11279083 DOI: 10.3390/medicina60071094] [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: 05/30/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024]
Abstract
Background and Objectives: This study aimed to evaluate the histological and biochemical effects of capsaicin on implant osseointegration and oxidative stress. Materials and Methods: Male Wistar albino rats weighing between 250 and 300 g were used in this study. Twenty-four rats were randomly divided into three equal groups: implant + control (n = 8), implant + capsaicin-1 (n = 8), and implant + capsaicin-2 (n = 8). Additionally, 2.5 mm diameter and 4 mm length titanium implants were surgically integrated into the corticocancellous bone parts of the femurs. In the treatment groups, rats were injected intraperitoneally with 25 mg/kg (implant + capsaicin-1) and 50 mg/kg (implant + capsaicin-2) of capsaicin. No additional applications were made in the control group. Three rats in total died during and after the experiment as a result of the analyses performed on 21 animals. Results: The highest total antioxidant status value was found in capsaicin dose 2, according to the analysis. The control group had the highest total oxidant status and oxidative stress index values, while group 2 of capsaicin had the lowest. After analysis, we found that there was no observed positive effect on osteointegration in this study (p > 0.05), although the bone implant connection was higher in the groups treated with capsaicin. Conclusions: A positive effect on osteointegration was not observed in this study. This may be due to osteoclast activation. However, it was found that it has a positive effect on oxidative stress. Osteoclast activation may be the cause of this phenomenon. Capsaicin was found to have a positive effect on oxidative stress (p < 0.05). It was also observed to have a positive effect on oxidative stress.
Collapse
Affiliation(s)
- Muhammet Bahattin Bingül
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Harran University, Sanliurfa 63300, Turkey; (M.B.B.); (M.E.P.)
| | - Mehmet Gul
- Department of Periodontology, Faculty of Dentistry, Harran University, Sanliurfa 63300, Turkey
| | - Serkan Dündar
- Department of Periodontology, Faculty of Dentistry, Firat University, Elazig 23119, Turkey;
| | - Kevser Sökmen
- Department of Periodontology, Faculty of Dentistry, Alanya Alaaddin Keykubat University, Antalya 07070, Turkey;
| | - Gökhan Artas
- Department of Medical, Faculty of Medicine, Pathology Firat University, Elazig 23119, Turkey;
| | - Mehmet Emrah Polat
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Harran University, Sanliurfa 63300, Turkey; (M.B.B.); (M.E.P.)
| | - Murat Tanrisever
- Department of Surgery, Faculty of Veterinary Medicine, Firat University, Elazig 23119, Turkey;
| | - Erhan Cahit Ozcan
- Department of Esthetic, Faculty of Medicine, Plastic and Reconstructive Surgery, Elazig 44090, Turkey;
| |
Collapse
|
21
|
Phull S, Marx D, Akens MK, Ghert M, Towler MR. In vitroassessment of a gallium-doped glass polyalkenoate cement: chemotherapeutic potential, cytotoxicity and osteogenic effects. Biomed Mater 2024; 19:055006. [PMID: 38917820 DOI: 10.1088/1748-605x/ad5ba5] [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: 01/30/2024] [Accepted: 06/25/2024] [Indexed: 06/27/2024]
Abstract
Metastatic bone lesions are often osteolytic, which causes advanced-stage cancer sufferers to experience severe pain and an increased risk of developing a pathological fracture. Gallium (Ga) ion possesses antineoplastic and anti-bone resorption properties, suggesting the potential for its local administration to impede the growth of metastatic bone lesions. This study investigated the chemotherapeutic potential, cytotoxicity, and osteogenic effects of a Ga-doped glass polyalkenoate cement (GPC) (C-TA2) compared to its non-gallium (C-TA0) counterpart. Ion release profiles revealed a biphasic pattern characterized by an initial burst followed by a gradually declining release of ions. C-TA2 continued to release Ga steadily throughout the experimentation period (7 d) and exhibited prolonged zinc (Zn) release compared to C-TA0. Interestingly, the Zn release from both GPCs appeared to cause a chemotherapeutic effect against H1092 lung cancer cellsin vitro, with the prolonged Zn release from C-TA2 extending this effect. Unfortunately, both GPCs enhanced the viability of HCC2218 breast cancer cells, suggesting that the chemotherapeutic effects of Zn could be tied to cellular differences in preferred Zn concentrations. The utilization of SAOS-2 and MC3T3 cell lines as bone cell models yielded conflicting results, with the substantial decline in MC3T3 viability closely associated with silicon (Si) release, indicating cellular variations in Si toxicity. Despite this ambiguity, both GPCs exhibited harmful effects on the osteogenesis of primary rat osteoblasts, raising concerns about excessive burst Zn release. While Ga/Zn-doped GPCs hold promise for treating metastatic bone lesions caused by lung cancers, further optimization is required to mitigate cytotoxicity on healthy bone.
Collapse
Affiliation(s)
- Sunjeev Phull
- Department of Mechanical Engineering, Toronto Metropolitan University, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Daniella Marx
- Department of Mechanical Engineering, Toronto Metropolitan University, Toronto, ON, Canada
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON, Canada
| | - Margarete K Akens
- University Health Network, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Michelle Ghert
- Department of Surgery, McMaster University, Hamilton L8V 5C2, ON, Canada
| | - Mark R Towler
- Department of Chemical & Biochemical Engineering, Missouri S&T, Rolla, MO, United States of America
| |
Collapse
|
22
|
Abdal Dayem A, Yan E, Do M, Kim Y, Lee Y, Cho SG, Kim DH. Engineering extracellular vesicles for ROS scavenging and tissue regeneration. NANO CONVERGENCE 2024; 11:24. [PMID: 38922501 PMCID: PMC11208369 DOI: 10.1186/s40580-024-00430-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024]
Abstract
Stem cell therapy holds promise for tissue regeneration, yet significant challenges persist. Emerging as a safer and potentially more effective alternative, extracellular vesicles (EVs) derived from stem cells exhibit remarkable abilities to activate critical signaling cascades, thereby facilitating tissue repair. EVs, nano-scale membrane vesicles, mediate intercellular communication by encapsulating a diverse cargo of proteins, lipids, and nucleic acids. Their therapeutic potential lies in delivering cargos, activating signaling pathways, and efficiently mitigating oxidative stress-an essential aspect of overcoming limitations in stem cell-based tissue repair. This review focuses on engineering and applying EVs in tissue regeneration, emphasizing their role in regulating reactive oxygen species (ROS) pathways. Additionally, we explore strategies to enhance EV therapeutic activity, including functionalization and incorporation of antioxidant defense proteins. Understanding these molecular mechanisms is crucial for optimizing EV-based regenerative therapies. Insights into EV and ROS signaling modulation pave the way for targeted and efficient regenerative therapies harnessing the potential of EVs.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ellie Yan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Yoojung Kim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Yeongseo Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea.
- R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin- gu, Seoul, 05029, Republic of Korea.
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, 21205, USA.
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
23
|
Hassan N, Krieg T, Kopp A, Bach AD, Kröger N. Challenges and Pitfalls of Research Designs Involving Magnesium-Based Biomaterials: An Overview. Int J Mol Sci 2024; 25:6242. [PMID: 38892430 PMCID: PMC11172609 DOI: 10.3390/ijms25116242] [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: 04/17/2024] [Revised: 05/31/2024] [Accepted: 06/01/2024] [Indexed: 06/21/2024] Open
Abstract
Magnesium-based biomaterials hold remarkable promise for various clinical applications, offering advantages such as reduced stress-shielding and enhanced bone strengthening and vascular remodeling compared to traditional materials. However, ensuring the quality of preclinical research is crucial for the development of these implants. To achieve implant success, an understanding of the cellular responses post-implantation, proper model selection, and good study design are crucial. There are several challenges to reaching a safe and effective translation of laboratory findings into clinical practice. The utilization of Mg-based biomedical devices eliminates the need for biomaterial removal surgery post-healing and mitigates adverse effects associated with permanent biomaterial implantation. However, the high corrosion rate of Mg-based implants poses challenges such as unexpected degradation, structural failure, hydrogen evolution, alkalization, and cytotoxicity. The biocompatibility and degradability of materials based on magnesium have been studied by many researchers in vitro; however, evaluations addressing the impact of the material in vivo still need to be improved. Several animal models, including rats, rabbits, dogs, and pigs, have been explored to assess the potential of magnesium-based materials. Moreover, strategies such as alloying and coating have been identified to enhance the degradation rate of magnesium-based materials in vivo to transform these challenges into opportunities. This review aims to explore the utilization of Mg implants across various biomedical applications within cellular (in vitro) and animal (in vivo) models.
Collapse
Affiliation(s)
- Nourhan Hassan
- Department of Plastic, Reconstructive and Aesthetic Surgery, University Hospital Cologne, 50937 Cologne, Germany
- Institute for Laboratory Animal Science and Experimental Surgery, University of Aachen Medical Center, Faculty of Medicine, RWTH-Aachen University, 52074 Aachen, Germany
- Biotechnology Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Thomas Krieg
- Translational Matrix Biology, Medical Faculty, University of Cologne, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, 50937 Cologne, Germany
| | | | - Alexander D. Bach
- Department of Plastic, Aesthetic and Hand Surgery, St. Antonius Hospital Eschweiler, 52249 Eschweiler, Germany
| | - Nadja Kröger
- Institute for Laboratory Animal Science and Experimental Surgery, University of Aachen Medical Center, Faculty of Medicine, RWTH-Aachen University, 52074 Aachen, Germany
- Department of Plastic, Aesthetic and Hand Surgery, St. Antonius Hospital Eschweiler, 52249 Eschweiler, Germany
| |
Collapse
|
24
|
Cheng S, Hu X, Sun K, Huang Z, Zhao Y, Sun Y, Zeng B, Wang J, Zhao D, Lu S, Shi Q, Wang Y, Zhang W, Liu X, Shu B. Local Application of Tanshinone IIA protects mesenchymal stem cells from apoptosis and promotes fracture healing in ovariectomized mice. J Orthop Surg Res 2024; 19:309. [PMID: 38783358 PMCID: PMC11112815 DOI: 10.1186/s13018-024-04793-x] [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: 04/08/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Elderly patients suffering from osteoporotic fractures are more susceptible to delayed union or nonunion, and their bodies then are in a state of low-grade chronic inflammation with decreased antioxidant capacity. Tanshinone IIA is widely used in treating cardiovascular and cerebrovascular diseases in China and has anti-inflammatory and antioxidant effects. We aimed to observe the antioxidant effects of Tanshinone IIA on mesenchymal stem cells (MSCs), which play important roles in bone repair, and the effects of local application of Tanshinone IIA using an injectable biodegradable hydrogel on osteoporotic fracture healing. METHODS MSCs were pretreated with or without different concentrations of Tanshinone IIA followed by H2O2 treatment. Ovariectomized (OVX) C57BL/6 mice received a mid-shaft transverse osteotomy fracture on the left tibia, and Tanshinone IIA was applied to the fracture site using an injectable hydrogel. RESULTS Tanshinone IIA pretreatment promoted the expression of nuclear factor erythroid 2-related factor 2 and antioxidant enzymes, and inhibited H2O2-induced reactive oxygen species accumulation in MSCs. Furthermore, Tanshinone IIA reversed H2O2-induced apoptosis and decrease in osteogenic differentiation in MSCs. After 4 weeks of treatment with Tanshinone IIA in OVX mice, the bone mineral density of the callus was significantly increased and the biomechanical properties of the healed tibias were improved. Cell apoptosis was decreased and Nrf2 expression was increased in the early stage of callus formation. CONCLUSIONS Taken together, these results indicate that Tanshinone IIA can activate antioxidant enzymes to protect MSCs from H2O2-induced cell apoptosis and osteogenic differentiation inhibition. Local application of Tanshinone IIA accelerates fracture healing in ovariectomized mice.
Collapse
Affiliation(s)
- Shao Cheng
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
- School of Orthopedics, Henan University of Chinese Medicine, Zhengzhou, 450002, China
| | - Xiaohui Hu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Kanghui Sun
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Ziyu Huang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Yongjian Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Yueli Sun
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Bo Zeng
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Jing Wang
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Dongfeng Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Sheng Lu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Qi Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China
| | - Weian Zhang
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai, 200237, China.
| | - Xinhua Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Bing Shu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
- Spine Institute, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 200032, China.
- Key Laboratory, Ministry of Education of China, Shanghai, 200032, China.
| |
Collapse
|
25
|
Shkundin A, Halaris A. IL-8 (CXCL8) Correlations with Psychoneuroimmunological Processes and Neuropsychiatric Conditions. J Pers Med 2024; 14:488. [PMID: 38793070 PMCID: PMC11122344 DOI: 10.3390/jpm14050488] [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: 03/18/2024] [Revised: 04/26/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
Interleukin-8 (IL-8/CXCL8), an essential CXC chemokine, significantly influences psychoneuroimmunological processes and affects neurological and psychiatric health. It exerts a profound effect on immune cell activation and brain function, suggesting potential roles in both neuroprotection and neuroinflammation. IL-8 production is stimulated by several factors, including reactive oxygen species (ROS) known to promote inflammation and disease progression. Additionally, CXCL8 gene polymorphisms can alter IL-8 production, leading to potential differences in disease susceptibility, progression, and severity across populations. IL-8 levels vary among neuropsychiatric conditions, demonstrating sensitivity to psychosocial stressors and disease severity. IL-8 can be detected in blood circulation, cerebrospinal fluid (CSF), and urine, making it a promising candidate for a broad-spectrum biomarker. This review highlights the need for further research on the diverse effects of IL-8 and the associated implications for personalized medicine. A thorough understanding of its complex role could lead to the development of more effective and personalized treatment strategies for neuropsychiatric conditions.
Collapse
Affiliation(s)
| | - Angelos Halaris
- Department of Psychiatry and Behavioral Neurosciences, Loyola University Chicago Stritch School of Medicine, Loyola University Medical Center, Maywood, IL 60153, USA;
| |
Collapse
|
26
|
Zeng Y, Hu X, Cai Z, Qiu D, Ran Y, Ding Y, Shi J, Cai X, Pan Y. Photodynamic and nitric oxide therapy-based synergistic antimicrobial nanoplatform: an advanced root canal irrigation system for endodontic bacterial infections. J Nanobiotechnology 2024; 22:213. [PMID: 38689259 PMCID: PMC11059741 DOI: 10.1186/s12951-024-02483-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 04/16/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND The main issues faced during the treatment of apical periodontitis are the management of bacterial infection and the facilitation of the repair of alveolar bone defects to shorten disease duration. Conventional root canal irrigants are limited in their efficacy and are associated with several side effects. This study introduces a synergistic therapy based on nitric oxide (NO) and antimicrobial photodynamic therapy (aPDT) for the treatment of apical periodontitis. RESULTS This research developed a multifunctional nanoparticle, CGP, utilizing guanidinylated poly (ethylene glycol)-poly (ε-Caprolactone) polymer as a carrier, internally loaded with the photosensitizer chlorin e6. During root canal irrigation, the guanidino groups on the surface of CGP enabled effective biofilm penetration. These groups undergo oxidation by hydrogen peroxide in the aPDT process, triggering the release of NO without hindering the production of singlet oxygen. The generated NO significantly enhanced the antimicrobial capability and biofilm eradication efficacy of aPDT. Furthermore, CGP not only outperforms conventional aPDT in eradicating biofilms but also effectively promotes the repair of alveolar bone defects post-eradication. Importantly, our findings reveal that CGP exhibits significantly higher biosafety compared to sodium hypochlorite, alongside superior therapeutic efficacy in a rat model of apical periodontitis. CONCLUSIONS This study demonstrates that CGP, an effective root irrigation system based on aPDT and NO, has a promising application in root canal therapy.
Collapse
Affiliation(s)
- Youyun Zeng
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiangyu Hu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhibin Cai
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Dongchao Qiu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Ying Ran
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Yiqin Ding
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiayi Shi
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaojun Cai
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Yihuai Pan
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
27
|
Silva JC, Meneses J, Garrudo FFF, Fernandes SR, Alves N, Ferreira FC, Pascoal-Faria P. Direct coupled electrical stimulation towards improved osteogenic differentiation of human mesenchymal stem/stromal cells: a comparative study of different protocols. Sci Rep 2024; 14:5458. [PMID: 38443455 PMCID: PMC10915174 DOI: 10.1038/s41598-024-55234-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Electrical stimulation (ES) has been described as a promising tool for bone tissue engineering, being known to promote vital cellular processes such as cell proliferation, migration, and differentiation. Despite the high variability of applied protocol parameters, direct coupled electric fields have been successfully applied to promote osteogenic and osteoinductive processes in vitro and in vivo. Our work aims to study the viability, proliferation, and osteogenic differentiation of human bone marrow-derived mesenchymal stem/stromal cells when subjected to five different ES protocols. The protocols were specifically selected to understand the biological effects of different parts of the generated waveform for typical direct-coupled stimuli. In vitro culture studies evidenced variations in cell responses with different electric field magnitudes (numerically predicted) and exposure protocols, mainly regarding tissue mineralization (calcium contents) and osteogenic marker gene expression while maintaining high cell viability and regular morphology. Overall, our results highlight the importance of numerical guided experiments to optimize ES parameters towards improved in vitro osteogenesis protocols.
Collapse
Affiliation(s)
- João C Silva
- Department of Bioengineering and iBB-Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal.
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal.
- Centre for Rapid and Sustainable Product Development (CDRSP), Polytechnic of Leiria, Marinha Grande, 2430-028, Leiria, Portugal.
| | - João Meneses
- Centre for Rapid and Sustainable Product Development (CDRSP), Polytechnic of Leiria, Marinha Grande, 2430-028, Leiria, Portugal.
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal.
| | - Fábio F F Garrudo
- Department of Bioengineering and iBB-Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
- Instituto de Telecomunicações, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001, Lisboa, Portugal
| | - Sofia R Fernandes
- Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande, 1749-016, Lisboa, Portugal
| | - Nuno Alves
- Centre for Rapid and Sustainable Product Development (CDRSP), Polytechnic of Leiria, Marinha Grande, 2430-028, Leiria, Portugal
- Associate Laboratory for Advanced Production and Intelligent Systems (ARISE), 4050-313, Porto, Portugal
- Department of Mechanical Engineering, School of Technology and Management, Polytechnic of Leiria, Morro do Lena-Alto do Vieiro, Apartado 4163, 2411-901, Leiria, Portugal
| | - Frederico Castelo Ferreira
- Department of Bioengineering and iBB-Institute of Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal
| | - Paula Pascoal-Faria
- Centre for Rapid and Sustainable Product Development (CDRSP), Polytechnic of Leiria, Marinha Grande, 2430-028, Leiria, Portugal.
- Associate Laboratory for Advanced Production and Intelligent Systems (ARISE), 4050-313, Porto, Portugal.
- Department of Mathematics, School of Technology and Management, Polytechnic of Leiria, Morro do Lena - Alto do Vieiro, Apartado 4163, 2411-901, Leiria, Portugal.
| |
Collapse
|
28
|
Singh AK, Prasad P, Cancelas JA. Mesenchymal stromal cells, metabolism, and mitochondrial transfer in bone marrow normal and malignant hematopoiesis. Front Cell Dev Biol 2023; 11:1325291. [PMID: 38169927 PMCID: PMC10759248 DOI: 10.3389/fcell.2023.1325291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Hematopoietic stem cell (HSC) transplantation-based treatments are in different phases of clinical development, ranging from current therapies to a promise in the repair and regeneration of diseased tissues and organs. Mesenchymal stromal/stem cells (MSCs), which are fibroblast-like heterogeneous progenitors with multilineage differentiation (osteogenic, chondrogenic, and adipogenic) and self-renewal potential, and exist in the bone marrow (BM), adipose, and synovium, among other tissues, represent one of the most widely used sources of stem cells in regenerative medicine. MSCs derived from bone marrow (BM-MSCs) exhibit a variety of traits, including the potential to drive HSC fate and anti-inflammatory and immunosuppressive capabilities via paracrine activities and interactions with the innate and adaptive immune systems. The role of BM-MSC-derived adipocytes is more controversial and may act as positive or negative regulators of benign or malignant hematopoiesis based on their anatomical location and functional crosstalk with surrounding cells in the BM microenvironment. This review highlights the most recent clinical and pre-clinical findings on how BM-MSCs interact with the surrounding HSCs, progenitors, and immune cells, and address some recent insights on the mechanisms that mediate MSCs and adipocyte metabolic control through a metabolic crosstalk between BM microenvironment cells and intercellular mitochondrial transfer in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Abhishek K. Singh
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Parash Prasad
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
29
|
Bose S, Robertson SF, Vu AA. Garlic extract enhances bioceramic bone scaffolds through upregulating ALP & BGLAP expression in hMSC-monocyte co-culture. BIOMATERIALS ADVANCES 2023; 154:213622. [PMID: 37742556 DOI: 10.1016/j.bioadv.2023.213622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 08/15/2023] [Accepted: 09/07/2023] [Indexed: 09/26/2023]
Abstract
Bone homeostasis is predicated by osteoblast and osteoclast cell cycles where gene expressions are responsible for their differentiation from human mesenchymal stem cells (hMSC) and monocytes, respectively. The pro-osteogenic potential of an hMSC-monocyte co-culture can be measured through complementary DNA (mRNA synthesis) within the nucleus, known as quantitative polymerase chain reaction (qPCR). Through this technique, the effects of garlic extract (allicin) release from calcium phosphate bone scaffolds on gene expression of bone forming and bone remodeling cells was explored. Results show this complex biomaterial system enhances hMSC differentiation through the upregulation of bone-forming proteins. Osteoblastic gene markers alkaline phosphatase (ALP) and osteocalcin (BGLAP), are respectively upregulated by 3-fold and 1.6-fold by day 14. These mature osteoblasts then upregulate the receptor activator of nuclear factor-kB ligand (RANKL) which recruits osteoclast cells, as captured by a nearly 2-fold higher osteoclast expression of tartrate-resistance acid-phosphatase (ACP5). This also activates antagonist osteoprotegerin (OPG) expression in osteoblasts, decreasing osteoclast resorption potential and ACP5 expression by day 21. The pro-osteogenic environment with garlic extract release is further quantified by a 4× increase in phosphatase activity and visibly captured in immunofluorescent tagged confocal images. Also corroborated by enhanced collagen formation in a preliminary in vivo rat distal femur model, this work collectively reveals how garlic extract can enhance bioceramic scaffolds for bone tissue regenerative applications.
Collapse
Affiliation(s)
- Susmita Bose
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States.
| | - Samuel F Robertson
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States
| | - Ashley A Vu
- W. M. Keck Biomedical Materials Research Laboratory, School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, United States
| |
Collapse
|
30
|
Gaweł J, Milan J, Żebrowski J, Płoch D, Stefaniuk I, Kus-Liśkiewicz M. Biomaterial composed of chitosan, riboflavin, and hydroxyapatite for bone tissue regeneration. Sci Rep 2023; 13:17004. [PMID: 37813934 PMCID: PMC10562422 DOI: 10.1038/s41598-023-44225-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/05/2023] [Indexed: 10/11/2023] Open
Abstract
Biomaterial engineering approaches involve using a combination of miscellaneous bioactive molecules which may promote cell proliferation and, thus, form a scaffold with the environment that favors the regeneration process. Chitosan, a naturally occurring biodegradable polymer, possess some essential features, i.e., biodegradability, biocompatibility, and in the solid phase good porosity, which may contribute to promote cell adhesion. Moreover, doping of the materials with other biocompounds will create a unique and multifunctional scaffold that will be useful in regenerative medicine. This study is focused on the manufacturing and characterization of composite materials based on chitosan, hydroxyapatite, and riboflavin. The resulting films were fabricated by the casting/solvent evaporation method. Morphological and spectroscopy analyses of the films revealed a porous structure and an interconnection between chitosan and apatite. The composite material showed an inhibitory effect on Staphylococcus aureus and exhibited higher antioxidant activity compared to pure chitosan. In vitro studies on riboflavin showed increased cell proliferation and migration of fibroblasts and osteosarcoma cells, thus demonstrating their potential for bone tissue engineering applications.
Collapse
Affiliation(s)
- Justyna Gaweł
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1 St, 35‑310, Rzeszow, Poland
| | - Justyna Milan
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1 St, 35‑310, Rzeszow, Poland
- Laboratory of Innovative Toxicological Research and Analyses, Institute of Medical Studies, Medical College, Rzeszów University, Aleja Majora W. Kopisto 2a, 35-959, Rzeszow, Poland
| | - Jacek Żebrowski
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1 St, 35‑310, Rzeszow, Poland
| | - Dariusz Płoch
- Institute of Materials Engineering, College of Natural Sciences, University of Rzeszow, Pigonia 1 St, 35‑310, Rzeszow, Poland
| | - Ireneusz Stefaniuk
- Institute of Materials Engineering, College of Natural Sciences, University of Rzeszow, Pigonia 1 St, 35‑310, Rzeszow, Poland
| | - Małgorzata Kus-Liśkiewicz
- Institute of Biotechnology, College of Natural Sciences, University of Rzeszow, Pigonia 1 St, 35‑310, Rzeszow, Poland.
| |
Collapse
|
31
|
Riegger J, Schoppa A, Ruths L, Haffner-Luntzer M, Ignatius A. Oxidative stress as a key modulator of cell fate decision in osteoarthritis and osteoporosis: a narrative review. Cell Mol Biol Lett 2023; 28:76. [PMID: 37777764 PMCID: PMC10541721 DOI: 10.1186/s11658-023-00489-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/11/2023] [Indexed: 10/02/2023] Open
Abstract
During aging and after traumatic injuries, cartilage and bone cells are exposed to various pathophysiologic mediators, including reactive oxygen species (ROS), damage-associated molecular patterns, and proinflammatory cytokines. This detrimental environment triggers cellular stress and subsequent dysfunction, which not only contributes to the development of associated diseases, that is, osteoporosis and osteoarthritis, but also impairs regenerative processes. To counter ROS-mediated stress and reduce the overall tissue damage, cells possess diverse defense mechanisms. However, cellular antioxidative capacities are limited and thus ROS accumulation can lead to aberrant cell fate decisions, which have adverse effects on cartilage and bone homeostasis. In this narrative review, we address oxidative stress as a major driver of pathophysiologic processes in cartilage and bone, including senescence, misdirected differentiation, cell death, mitochondrial dysfunction, and impaired mitophagy by illustrating the consequences on tissue homeostasis and regeneration. Moreover, we elaborate cellular defense mechanisms, with a particular focus on oxidative stress response and mitophagy, and briefly discuss respective therapeutic strategies to improve cell and tissue protection.
Collapse
Affiliation(s)
- Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081, Ulm, Germany.
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Leonie Ruths
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| |
Collapse
|
32
|
Li Y, Liu X, Chu Y, Li C, Gao T, Jiang X, Zhu Z, Sheng Q, Han L. Effect of high-fructose consumption in pregnancy on the bone growth of offspring rats. Front Nutr 2023; 10:1203063. [PMID: 37662593 PMCID: PMC10469680 DOI: 10.3389/fnut.2023.1203063] [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: 04/10/2023] [Accepted: 07/24/2023] [Indexed: 09/05/2023] Open
Abstract
Growing evidence suggests that bone health is programmed in early life. Maternal diet may influence the skeletal development of offspring. We aimed to determine the possible effects of high-fructose intake during pregnancy on different aspects of long bone morphology in the offspring of rats and to initially explore the possible mechanisms. Pregnant Sprague-Dawley rats were randomly divided into four groups and intragastrically administered the same dose of distilled water (CON, n = 12), 20 g/kg/day glucose (GLU, n = 12), 10 g/kg/day fructose (LFRU, n = 12), or 20 g/kg/day fructose (HFRU, n = 12) for 21 days during gestation. Computed tomography was used to analyze the cortical and cancellous bones of the distal femur of the offspring rats, and circulating bone metabolic biomarkers were measured using enzyme immunoassay. The results showed that high-fructose intake during pregnancy could decrease body weight, impair glucose metabolism, and increase serum leptin and uric acid in offspring. The offspring in the HFRU group had higher levels of the N-terminal propeptide of type I procollagen (PINP) and the C-telopeptide of type I collagen (CTX). The bone mean density (BMD), the total cross-sectional area inside the periosteal envelope (Tt.Ar), cortical bone area (Ct.Ar), medullary (or marrow) area (Ma.Ar), and trabecular mean density of the offspring in the HFRU group were lower than those in the CON group. Tartrate-resistant acid phosphatase (Trap) staining showed that high-fructose intake during pregnancy could increase the number of osteoclasts and increase the absorption area. Our results suggested that excessive fructose intake during pregnancy could inhibit skeletal development in offspring. Thus, attention to fructose intake during pregnancy is important for bone development in offspring.
Collapse
Affiliation(s)
- Yijing Li
- Department of Nutrition, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoqian Liu
- Maternal, Child & Adolescent Health, Qingdao University, Qingdao, China
| | - Yuning Chu
- Department of Nutrition, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cai Li
- Department of Nutrition, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Tianlin Gao
- School of Public Health, Qingdao University, Qingdao, China
| | - Xiuli Jiang
- Department of Nutrition, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zihan Zhu
- Department of Nutrition, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qi Sheng
- Department of Nutrition, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Han
- Department of Nutrition, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
33
|
Zalewska A, Antonowicz B, Szulimowska J, Zieniewska-Siemieńczuk I, Leśniewska B, Borys J, Zięba S, Kostecka-Sochoń P, Żendzian-Piotrowska M, Lo Giudice R, Lo Giudice G, Żukowski P, Maciejczyk M. Mitochondrial Redox Balance of Fibroblasts Exposed to Ti-6Al-4V Microplates Subjected to Different Types of Anodizing. Int J Mol Sci 2023; 24:12896. [PMID: 37629077 PMCID: PMC10454109 DOI: 10.3390/ijms241612896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/12/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Despite the high biocompatibility of titanium and its alloys, the need to remove titanium implants is increasingly being debated due to the potential for adverse effects associated with long-term retention. Therefore, new solutions are being sought to enhance the biocompatibility of titanium implants. One of them is to increase the thickness of the passive layer of the implant made of titanium dioxide. We were the first to evaluate the effect of hard-anodized (type II) Ti-6Al-4V alloy discs on the cytotoxicity, mitochondrial function, and redox balance of fibroblasts mitochondria compared to standard-anodized (type III) and non-anodized discs. The study used fibroblasts obtained from human gingival tissue. The test discs were applied to the bottom of 12-well plates. Cells were cultured for 24 h and 7, 14, and 21 days and mitochondria were isolated. We demonstrated the occurrence of oxidative stress in the mitochondria of fibroblasts of all tested groups, regardless of the presence and type of anodization. Type II anodization prevented changes in complex II activity (vs. control). The lowest degree of citrate synthase inhibition occurred in mitochondria exposed to titanium discs with type II anodization. In the last phase of culture, the presence of type II anodization reduced the degree of cytochrome c oxidase inhibition compared to the other tests groups and the control group, and prevented apoptosis. Throughout the experiment, the release of titanium, aluminium, and vanadium ions from titanium discs with a hard-anodized passive layer was higher than from the other titanium discs, but decreased with time. The obtained results proved the existence of dysfunction and redox imbalance in the mitochondria of fibroblasts exposed to hard-anodized titanium discs, suggesting the need to search for new materials perhaps biodegradable in tissues of the human body.
Collapse
Affiliation(s)
- Anna Zalewska
- Conservative Dentistry Department, Medical University in Bialystok, 15-278 Bialystok, Poland; (J.S.); (I.Z.-S.); (P.K.-S.)
| | - Bożena Antonowicz
- Dental Surgery Department, Medical University in Bialystok, 15-278 Bialystok, Poland;
| | - Julita Szulimowska
- Conservative Dentistry Department, Medical University in Bialystok, 15-278 Bialystok, Poland; (J.S.); (I.Z.-S.); (P.K.-S.)
| | - Izabela Zieniewska-Siemieńczuk
- Conservative Dentistry Department, Medical University in Bialystok, 15-278 Bialystok, Poland; (J.S.); (I.Z.-S.); (P.K.-S.)
| | - Barbara Leśniewska
- Department of Analytical and Inorganic Chemistry, University in Bialystok, 15-328 Bialystok, Poland;
| | - Jan Borys
- Department of Maxillofacial Surgery, Medical University in Bialystok, 15-278 Bialystok, Poland;
| | - Sara Zięba
- PhD School, Medical University in Bialystok, 15-278 Bialystok, Poland
| | - Paula Kostecka-Sochoń
- Conservative Dentistry Department, Medical University in Bialystok, 15-278 Bialystok, Poland; (J.S.); (I.Z.-S.); (P.K.-S.)
| | - Małgorzata Żendzian-Piotrowska
- Department of Hygiene, Epidemiology and Ergonomics, Medical University in Bialystok, 15-278 Bialystok, Poland; (M.Ż.-P.); (M.M.)
| | - Roberto Lo Giudice
- Department of Human Pathology of the Adult and Evolutive Age G. Barresi, Messina University, 98100 Messina, Italy;
| | - Giusseppe Lo Giudice
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Messina University, 98100 Messina, Italy;
| | - Piotr Żukowski
- Restorative Dentistry Department, Croydon University, London CR9 1DX, UK;
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomics, Medical University in Bialystok, 15-278 Bialystok, Poland; (M.Ż.-P.); (M.M.)
| |
Collapse
|
34
|
Oxidative Stress and Inflammation in Osteoporosis: Molecular Mechanisms Involved and the Relationship with microRNAs. Int J Mol Sci 2023; 24:ijms24043772. [PMID: 36835184 PMCID: PMC9963528 DOI: 10.3390/ijms24043772] [Citation(s) in RCA: 128] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Osteoporosis is characterized by the alteration of bone homeostasis due to an imbalance between osteoclastic bone resorption and osteoblastic bone formation. Estrogen deficiency causes bone loss and postmenopausal osteoporosis, the pathogenesis of which also involves oxidative stress, inflammatory processes, and the dysregulation of the expression of microRNAs (miRNAs) that control gene expression at post-transcriptional levels. Oxidative stress, due to an increase in reactive oxygen species (ROS), proinflammatory mediators and altered levels of miRNAs enhance osteoclastogenesis and reduce osteoblastogenesis through mechanisms involving the activation of MAPK and transcription factors. The present review summarizes the principal molecular mechanisms involved in the role of ROS and proinflammatory cytokines on osteoporosis. Moreover, it highlights the interplay among altered miRNA levels, oxidative stress, and an inflammatory state. In fact, ROS, by activating the transcriptional factors, can affect miRNA expression, and miRNAs can regulate ROS production and inflammatory processes. Therefore, the present review should help in identifying targets for the development of new therapeutic approaches to osteoporotic treatment and improve the quality of life of patients.
Collapse
|
35
|
Tompkins YH, Liu G, Kim WK. Impact of exogenous hydrogen peroxide on osteogenic differentiation of broiler chicken compact bones derived mesenchymal stem cells. Front Physiol 2023; 14:1124355. [PMID: 36776980 PMCID: PMC9909420 DOI: 10.3389/fphys.2023.1124355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/19/2023] [Indexed: 01/27/2023] Open
Abstract
The effects of hydrogen peroxide (H2O2) on the osteogenic differentiation of primary chicken mesenchymal stem cells (MSCs) were investigated. MSCs were subjected to an osteogenic program and exposed to various concentrations of H2O2 for 14 days. Results showed that high concentrations of H2O2 (200 and 400 nM) significantly increased pro-apoptotic marker CASP8 expression and impaired osteogenic differentiation, as indicated by decreased mRNA expression levels of osteogenesis-related genes and reduced in vitro mineralization. In contrast, long-term H2O2 exposure promoted basal expression of adipogenic markers at the expense of osteogenesis in MSCs during osteogenic differentiation, and increased intracellular reactive oxygen species (ROS) production, as well as altered antioxidant enzyme gene expression. These findings suggest that long-term H2O2-induced ROS production impairs osteogenic differentiation in chicken MSCs under an osteogenic program.
Collapse
|
36
|
Yang J, Liu C, Sun H, Liu Y, Liu Z, Zhang D, Zhao G, Wang Q, Yang D. The progress in titanium alloys used as biomedical implants: From the view of reactive oxygen species. Front Bioeng Biotechnol 2022; 10:1092916. [PMID: 36601391 PMCID: PMC9806234 DOI: 10.3389/fbioe.2022.1092916] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022] Open
Abstract
Titanium and Titanium alloys are widely used as biomedical implants in oral and maxillofacial surgery, due to superior mechanical properties and biocompatibility. In specific clinical populations such as the elderly, diabetics and patients with metabolic diseases, the failure rate of medical metal implants is increased significantly, putting them at increased risk of revision surgery. Many studies show that the content of reactive oxygen species (ROS) in the microenvironment of bone tissue surrounding implant materials is increased in patients undergoing revision surgery. In addition, the size and shape of materials, the morphology, wettability, mechanical properties, and other properties play significant roles in the production of ROS. The accumulated ROS break the original balance of oxidation and anti-oxidation, resulting in host oxidative stress. It may accelerate implant degradation mainly by activating inflammatory cells. Peri-implantitis usually leads to a loss of bone mass around the implant, which tends to affect the long-term stability and longevity of implant. Therefore, a great deal of research is urgently needed to focus on developing antibacterial technologies. The addition of active elements to biomedical titanium and titanium alloys greatly reduce the risk of postoperative infection in patients. Besides, innovative technologies are developing new biomaterials surfaces conferring anti-infective properties that rely on the production of ROS. It can be considered that ROS may act as a messenger substance for the communication between the host and the implanted material, which run through the entire wound repair process and play a role that cannot be ignored. It is necessary to understand the interaction between oxidative stress and materials, the effects of oxidative stress products on osseointegration and implant life as well as ROS-induced bactericidal activity. This helps to facilitate the development of a new generation of well-biocompatible implant materials with ROS responsiveness, and ultimately prolong the lifespan of implants.
Collapse
Affiliation(s)
- Jun Yang
- School of Stomatology, Jiamusi University, Jiamusi, China,Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Chang Liu
- School of Stomatology, Jiamusi University, Jiamusi, China,Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Hui Sun
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Ying Liu
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Zhaogang Liu
- The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| | - Dan Zhang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China,*Correspondence: Donghong Yang, ; Dan Zhang,
| | - Gang Zhao
- School of Stomatology, Jiamusi University, Jiamusi, China
| | - Qiang Wang
- Liaoning Provincial Key Laboratory of Oral Diseases, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Donghong Yang
- School of Stomatology, Jiamusi University, Jiamusi, China,*Correspondence: Donghong Yang, ; Dan Zhang,
| |
Collapse
|
37
|
Chen Y, Lu C, Shang X, Wu K, Chen K. Primary cilia: The central role in the electromagnetic field induced bone healing. Front Pharmacol 2022; 13:1062119. [DOI: 10.3389/fphar.2022.1062119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Primary cilia have emerged as the cellular “antenna” that can receive and transduce extracellular chemical/physical signals, thus playing an important role in regulating cellular activities. Although the electromagnetic field (EMF) is an effective treatment for bone fractures since 1978, however, the detailed mechanisms leading to such positive effects are still unclear. Primary cilia may play a central role in receiving EMF signals, translating physical signals into biochemical information, and initiating various signalingsignaling pathways to transduce signals into the nucleus. In this review, we elucidated the process of bone healing, the structure, and function of primary cilia, as well as the application and mechanism of EMF in treating fracture healing. To comprehensively understand the process of bone healing, we used bioinformatics to analyze the molecular change and associated the results with other studies. Moreover, this review summarizedsummarized some limitations in EMFs-related research and provides an outlook for ongoing studies. In conclusion, this review illustrated the primary cilia and related molecular mechanisms in the EMF-induced bone healing process, and it may shed light on future research.
Collapse
|
38
|
Poly(Glycerol Succinate) as Coating Material for 1393 Bioactive Glass Porous Scaffolds for Tissue Engineering Applications. Polymers (Basel) 2022; 14:polym14225028. [PMID: 36433155 PMCID: PMC9697483 DOI: 10.3390/polym14225028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/06/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Aliphatic polyesters are widely used for biomedical, pharmaceutical and environmental applications due to their high biodegradability and cost-effective production. Recently, star and hyperbranched polyesters based on glycerol and ω-carboxy fatty diacids have gained considerable interest. Succinic acid and bio-based diacids similar to glycerol are regarded as safe materials according to the US Food and Drug Administration (FDA). Bioactive glass scaffolds utilized in bone tissue engineering are relatively brittle materials. However, their mechanical properties can be improved by using polymer coatings that can further control their degradation rate, tailor their biocompatibility and enhance their performance. The purpose of this study is to explore a new biopolyester poly(glycerol succinate) (PGSuc) reinforced with mesoporous bioactive nanoparticles (MSNs) as a novel coating material to produce hybrid scaffolds for bone tissue engineering. METHODS Bioactive glass scaffolds were coated with neat PGSuc, PGSuc loaded with dexamethasone sodium phosphate (DexSP) and PGSuc loaded with DexSP-laden MSNs. The physicochemical, mechanical and biological properties of the scaffolds were also evaluated. RESULTS Preliminary data are provided showing that polymer coatings with and without MSNs improved the physicochemical properties of the 1393 bioactive glass scaffolds and increased the ALP activity and alizarin red staining, suggesting osteogenic differentiation potential when cultured with adipose-derived mesenchymal stem cells. CONCLUSIONS PGSuc with incorporated MSNs coated onto 1393 bioactive glass scaffolds could be promising candidates in bone tissue engineering applications.
Collapse
|
39
|
Tompkins YH, Teng P, Pazdro R, Kim WK. Long Bone Mineral Loss, Bone Microstructural Changes and Oxidative Stress After Eimeria Challenge in Broilers. Front Physiol 2022; 13:945740. [PMID: 35923236 PMCID: PMC9340159 DOI: 10.3389/fphys.2022.945740] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/14/2022] [Indexed: 11/24/2022] Open
Abstract
The objective of this study was to evaluate the impact of coccidiosis on bone quality and antioxidant status in the liver and bone marrow of broiler chickens. A total of 360 13-day old male broilers (Cobb 500) were randomly assigned to different groups (negative control, low, medium-low, medium-high, and highest dose groups) and orally gavaged with different concentrations of Eimeria oocysts solution. Broiler tibia and tibia bone marrow were collected at 6 days post-infection (6 dpi) for bone 3-D structural analyses and the gene expression related to osteogenesis, oxidative stress, and adipogenesis using micro-computed tomography (micro-CT) and real-time qPCR analysis, respectively. Metaphyseal bone mineral density and content were reduced in response to the increase of Eimeria challenge dose, and poor trabecular bone traits were observed in the high inoculation group. However, there were no significant structural changes in metaphyseal cortical bone. Medium-high Eimeria challenge dose significantly increased level of peroxisome proliferator-activated receptor gamma (PPARG, p < 0.05) and decreased levels of bone gamma-carboxyglutamate protein coding gene (BGLAP, p < 0.05) and fatty acid synthase coding gene (FASN, p < 0.05) in bone marrow. An increased mRNA level of superoxide dismutase type 1 (SOD1, p < 0.05) and heme oxygenase 1 (HMOX1, p < 0.05), and increased enzyme activity of superoxide dismutase (SOD, p < 0.05) were found in bone marrow of Eimeria challenged groups compared with that of non-infected control. Similarly, enzyme activity of SOD and the mRNA level of SOD1, HMOX1 and aflatoxin aldehyde reductase (AKE7A2) were increased in the liver of infected broilers (p < 0.05), whereas glutathione (GSH) content was lower in the medium-high challenge group (p < 0.05) compared with non-challenged control. Moreover, the mRNA expression of catalase (CAT) and nuclear factor kappa B1 (NFKB1) showed dose-depend response in the liver, where expression of CAT and NFKB1 was upregulated in the low challenge group but decreased with the higher Eimeria challenge dosage (p < 0.05). In conclusion, high challenge dose of Eimeria infection negatively affected the long bone development. The structural changes of tibia and decreased mineral content were mainly located at the trabecular bone of metaphyseal area. The change of redox and impaired antioxidant status following the Eimeria infection were observed in the liver and bone marrow of broilers.
Collapse
Affiliation(s)
- Y. H. Tompkins
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - P. Teng
- Department of Poultry Science, University of Georgia, Athens, GA, United States
| | - R. Pazdro
- Department of Foods and Nutrition, University of Georgia, Athens, GA, United States
| | - W. K. Kim
- Department of Poultry Science, University of Georgia, Athens, GA, United States
- *Correspondence: W. K. Kim,
| |
Collapse
|