1
|
Wang Y, Yu X, Sun F, Fu Y, Hu T, Shi Q, Man Q. METTL14 Mediates Glut3 m6A methylation to improve osteogenesis under oxidative stress condition. Redox Rep 2025; 30:2435241. [PMID: 39737912 DOI: 10.1080/13510002.2024.2435241] [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] [Indexed: 01/01/2025] Open
Abstract
OBJECTIVES Bone remodeling imbalance contributes to osteoporosis. Though current medications enhance osteoblast involvement in bone formation, the underlying pathways remain unclear. This study was aimed to explore the pathways involved in bone formation by osteoblasts, we investigate the protective role of glycolysis and N6-methyladenosine methylation (m6A) against oxidative stress-induced impairment of osteogenesis in MC3T3-E1 cells. METHODS We utilized a concentration of 200 μM hydrogen peroxide (H2O2) to establish an oxidative damage model of MC3T3-E1 cells. Subsequently, we examined the alterations in the m6A methyltransferases (METTL3, METTL14), glucose transporter proteins (GLUT1, GLUT3) and validated m6A methyltransferase overexpression in vitro and in an osteoporosis model. The osteoblast differentiation and osteogenesis-related molecules and serum bone resorption markers were measured by biochemical analysis, Alizarin Red S staining, Western blot and ELISA. RESULTS H2O2 treatment inhibited glycolysis and osteoblast differentiation in MC3T3-E1 cells. However, when METTL14 was overexpressed, these changes induced by H2O2 could be mitigated. Our findings indicate that METTL14 promotes GLUT3 expression via YTHDF1, leading to the modulation of various parameters in the H2O2-induced model. Similar positive effects of METTL14 on osteogenesis were observed in an ovariectomized mouse osteoporosis model. DISCUSSION METTL14 could serve as a potential therapeutic approach for enhancing osteoporosis treatment.
Collapse
Affiliation(s)
- Ying Wang
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xueying Yu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Fenyong Sun
- Department of Clinical Laboratory, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Yan Fu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Tingting Hu
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Qiqing Shi
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, People's Republic of China
| | - Qiuhong Man
- Department of Clinical Laboratory, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, People's Republic of China
| |
Collapse
|
2
|
Nepal S, Shi N, Hoyd R, Spakowicz DJ, Orwoll E, Shikany JM, Napoli N, Tabung FK. Role of insulinemic and inflammatory dietary patterns on gut microbial composition and circulating biomarkers of metabolic health among older American men. Gut Microbes 2025; 17:2497400. [PMID: 40296253 PMCID: PMC12045561 DOI: 10.1080/19490976.2025.2497400] [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: 01/28/2025] [Revised: 03/25/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Chronic low-grade inflammation and hyperinsulinemia are linked with metabolic dysfunction and dysbiosis. This study investigated the role of dietary inflammatory and insulinemic potential on gut microbiome and circulating health biomarkers in older men. Data from the Osteoporotic Fractures in Men (MrOS) study were analyzed. Reversed Empirical Dietary Inflammatory Pattern (rEDIP), Empirical Dietary Index for Hyperinsulinemia (rEDIH), and Healthy Eating Index (HEI)-2020 scores were computed from food frequency questionnaire data. Stool samples were profiled using 16S rRNA sequencing. Elastic net regression identified diet-associated microbial profiles and multivariable-adjusted linear regression assessed diet-biomarker associations. Higher rEDIP, rEDIH, and HEI-2020 scores were positively associated with gut microbiota alpha diversity. Specific genera, including Intestinibacter and Lachnospira, associated positively, while Dielma, Peptococcus, Feacalitalea, and Negativibaccilus associated inversely with healthier dietary patterns. When evaluating changes in dietary patterns between baseline and visit 4 ( ~ 14 years), these genera tended to define rEDIP, rEDIH more than HEI-2020. In addition, higher dietary quality was linked to better biomarker profiles, including lower creatinine, sodium, triglycerides, and insulin resistance. Beneficial effects of higher dietary quality on health may be mediated by the ability of diet to regulate gut microbial composition and metabolic biomarker profiles.
Collapse
Affiliation(s)
- Sushma Nepal
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ni Shi
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Rebecca Hoyd
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Daniel J. Spakowicz
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eric Orwoll
- Department of Medicine, Oregon Health & Sciences University, Portland, OR, USA
| | - James M. Shikany
- Division of General Internal Medicine and Population Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicola Napoli
- Unit of Endocrinology and Diabetes, Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
- Division of Bone and Mineral Diseases, Washington University, St Louis, MO, USA
| | - Fred K. Tabung
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
3
|
Chang CH, Liou HH, Wu CK. Moderate-severe aortic arch calcification and high serum alkaline phosphatase co-modify the risk of cardiovascular events and mortality among chronic hemodialysis patients. Ren Fail 2025; 47:2449572. [PMID: 39801127 PMCID: PMC11731357 DOI: 10.1080/0886022x.2024.2449572] [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: 07/03/2024] [Revised: 12/02/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Patients with end-stage kidney disease undergoing chronic hemodialysis (HD) have an unparalleled risk of vascular calcification (VC) and high alkaline phosphatase (Alk-P) levels. However, whether VC contributed to the cardiovascular risk modified by serum Alk-P levels was not addressed in the population. METHODS A retrospective cohort study was conducted on chronic HD patients, between October 1 and December 31, 2018, with aortic arch calcification (AoAC) scores and serum Alk-P levels. Patients were categorized into four groups: non-to-mild AoAC/low Alk-P, non-to-mild AoAC/high Alk-P, moderate-to-severe AoAC/low Alk-P, and moderate-to-severe AoAC/high Alk-P. The Cox proportional hazard model and Kaplan-Meier analysis were used to evaluate the risks of major adverse cardiovascular effects (MACEs) and cardiovascular and all-cause mortality after multivariate adjustment. RESULTS Among 376 chronic HD patients recruited, 125 (33%) had non-to-mild AoAC/low Alk-P, 76 (20%) had non-to-mild AoAC/high Alk-P, 89 (24%) had moderate-to-severe AoAC/low Alk-P, and 86 (23%) had moderate-to-severe AoAC/high Alk-P. After 3 years of follow-up, patients with coexisting moderate-to-severe AoAC and high Alk-P had a higher risk of MACEs (aHR 1.76; 95% CI 1.06-2.92), and cardiovascular (aHR 2.49; 95% CI 1.21-5.11) and all-cause mortality (aHR 2.67; 95% CI 1.39-5.13) compared to those with non-to-mild AoAC/low Alk-P even after adjustments for significant clinical variables. CONCLUSIONS In chronic HD patients, moderate to severe AoAC co-existed with high Alk-P levels and enhanced the risk of MACEs and cardiovascular and all-cause mortality. Interventions to attenuate these risk factors simultaneously should be emphasized in this population.
Collapse
Affiliation(s)
- Cheng-Hao Chang
- Division of Nephrology, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Hung-Hsiang Liou
- Division of Nephrology, Department of Medicine, Hsin-Jen Hospital, New Taipei County, Taiwan
| | - Chung-Kuan Wu
- Division of Nephrology, Department of Internal Medicine, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
- School of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan
| |
Collapse
|
4
|
Huang Y, Wang Z. Therapeutic potential of SOX family transcription factors in osteoarthritis. Ann Med 2025; 57:2457520. [PMID: 39887675 PMCID: PMC11789227 DOI: 10.1080/07853890.2025.2457520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND As the worldwide population ages, osteoarthritis has significantly increased. This musculoskeletal condition has become a pressing global health issue and thus, prevention and treatment of osteoarthritis have become the primary focus of domestic and international research. Scholarly investigations of the molecular mechanisms that are related to the occurrence and development of osteoarthritis have shed light on the pathological causes of this condition to a certain extent, providing a foundation for its prevention and treatment. However, further research is necessary to fully understand the critical role of the transcription factor SOX9 in chondrocyte differentiation and the development of osteoarthritis. As a result, there has been widespread interest in SOX transcription factors. While SOX9 has been utilized as a biomarker to indicate the occurrence and prognosis of osteoarthritis, investigations into other members of the SOX family and the development of targeted treatments around SOX9 are still required. PURPOSE This article considers the impact of the SOX protein on the development and inhibition of osteoarthritis and highlights the need for therapeutic approaches targeting SOX9, as supported by existing research. RESULTS SOX9 can contribute to the process of osteoarthritis through acetylation and ubiquitination modifications. The regulation of the WNT signalling pathway, Nrf2/ARE signalling pathway, NF-κB signalling pathway and SOX9 is implicated in the emergence of osteoarthritis. Non-coding RNA may play a role in the onset and progression of osteoarthritis by modulating various SOX family members, including SOX2, SOX4, SOX5, SOX6, SOX8, SOX9 and SOX11. CONCLUSION SOX9 has the capability of mitigating the onset and progression of osteoarthritis through means such as medication therapy, stem cell therapy, recombinant adeno-associated virus (rAAV) vector therapy, physical therapy and other approaches.
Collapse
Affiliation(s)
- Yue Huang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Zhuo Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
5
|
Rivera Antonio AM, Padilla Martínez II, Torres-Ramos MA, Rosales-Hernández MC. Myeloperoxidase as a therapeutic target for oxidative damage in Alzheimer's disease. J Enzyme Inhib Med Chem 2025; 40:2456282. [PMID: 39950933 DOI: 10.1080/14756366.2025.2456282] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/14/2024] [Accepted: 01/14/2025] [Indexed: 05/09/2025] Open
Abstract
Alzheimer's disease (AD) is a major neurodegenerative disorder more common in older adults. One of the leading AD hypotheses involves the amyloid beta (A) production, it is associated to oxidative stress, neuroinflammation, and neurovascular damage. The interaction of A with the blood vessel wall contributes to the disruption of the blood-brain barrier (BBB), allowing neutrophil infiltration containing the myeloperoxidase enzyme (MPO), which produces hypochlorous acid (HOCl) a potent oxidant. Also, MPO could be released from the microglia cells and interact with the amyloid beta plaques. This review aims to study the role of MPO in the progression of AD, in particular its contribution to oxidative stress and neuroinflammation. Furthermore, to explore the MPO-potential as AD-biomarker to evaluate the therapeutic potential of its inhibitors to mitigate the neurotoxicity. Finally, revise MPO inhibitors that could act as dual inhibitors acting on MPO and acetylcholinesterase and or another target involved in AD.
Collapse
Affiliation(s)
- Astrid Mayleth Rivera Antonio
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón s/n, MéxicoCiudad de México, México
| | - Itzia Irene Padilla Martínez
- Laboratorio de Química Supramolecular y Nanociencias, Unidad Profesional Interdisciplinaria de Biotecnología, Instituto Politécnico Nacional, MéxicoCiudad de México, México
| | - Mónica A Torres-Ramos
- Dirección de investigación del Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez. Av, Ciudad de México. C.P, México
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatálisis, Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Salvador Díaz Mirón s/n, MéxicoCiudad de México, México
| |
Collapse
|
6
|
Liang Y, Li Y, Chen Y, Meng K, Zhou F, Pei Y, Liu Y, Qiu J. The impact of low energy availability risk on pre-competition physiological function in Chinese female combat athletes. J Int Soc Sports Nutr 2025; 22:2490170. [PMID: 40254934 PMCID: PMC12013142 DOI: 10.1080/15502783.2025.2490170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 04/02/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Low energy availability (LEA) can negatively impact athletes' physiological function and performance. This study aims to examine the prevalence of LEA in Chinese female combat athletes and monitor changes in physiological function and performance during the pre-competition period. METHOD We assessed the incidence of low energy availability (LEA) and eating disorder (ED) risks in 84 female combat athletes (judo, freestyle wrestling, and sanda) from Beijing using the Low Energy Availability in Females Questionnaire (LEAF-Q) and the Eating Disorder Examination Questionnaire (EDE-Q). From this group, 11 judo athletes who were preparing for competition were selected and divided into a low energy availability (LEA) group and a non-LEA group based on their energy availability levels. Dietary intake, training energy expenditure, body composition, resting metabolic rate, blood markers, and special judo fitness tests were monitored at 4 weeks, 2 weeks, and 0 weeks before the competition. RESULTS Among the 84 athletes, 45.2% of athletes (n = 38) were at increased risk of LEA, and 21.4% of athletes (n = 18) were classified as high in eating disorder risk. There were no significant differences in LEA and ED risk between elite and recreational athletes. Among the 11 athletes preparing for competition, 6 athletes (45.5%) were in a state of LEA at the initial stage (4 weeks before the competition), and by the competition week, all 11 athletes exhibited LEA. Additionally, athletes in the LEA group experienced significant reductions in VO2 and resting metabolic rate at 0 week of the competition compared to 4 weeks prior (p < 0.05). Thyroid function indicators and IGF-1 levels of LEA group also significantly decreased (p < 0.05). After completing the four-week pre-competition weight loss, heart rate recovery during the special judo fitness test improved significantly in both the LEA and non-LEA groups (p < 0.05). CONCLUSION The current study identified a risk of LEA among Chinese female combat sport athletes, with no significant difference in the prevalence of LEA between elite and recreational athletes. It is essential for Chinese coaches and sports medicine staff to implement LEA-related nutritional education across all performance levels. Moreover, preventive measures during training are recommended to mitigate the impact of LEA on physiological function during the pre-competition weight loss phase.
Collapse
Affiliation(s)
- Yiheng Liang
- Beijing Sport University, Department of Exercise Biochemistry, Exercise Science School, China
| | - Yuxuan Li
- Beijing Sport University, Department of Exercise Biochemistry, Exercise Science School, China
| | - Yan Chen
- Beijing Sport University, Department of Exercise Biochemistry, Exercise Science School, China
| | - Kun Meng
- Beijing Sport University, Department of Exercise Biochemistry, Exercise Science School, China
| | - Fanyang Zhou
- Beijing Research Institute of Sports Science, Beijing, China
| | - Yiran Pei
- Beijing Research Institute of Sports Science, Beijing, China
| | - Yong Liu
- Beijing Research Institute of Sports Science, Beijing, China
| | - Junqiang Qiu
- Beijing Sport University, Department of Exercise Biochemistry, Exercise Science School, China
- Beijing Sports Nutrition Engineering Research Center, Beijing, China
| |
Collapse
|
7
|
Sun T, Wang J, Liu X, Huang H, Wang J, Suo M, Zhang J, Li Z. Finite element models of intervertebral disc: recent advances and prospects. Ann Med 2025; 57:2453089. [PMID: 39840609 PMCID: PMC11755745 DOI: 10.1080/07853890.2025.2453089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 11/27/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025] Open
Abstract
OBJECTIVES The incidence rate of intervertebral disc degeneration (IVDD) is increasing year by year, which brings great harm to our health. The change of biomechanical factors is an important reason for IVDD. Therefore, more and more studies use finite element (FE) models to analyze the biomechanics of spine. METHODS In this review, literatures which reported the FE model of intervertebral disc (IVD) were reviewed. We summarized the types and constructional methods of the FE models and analyzed the applications of some representative FE models. RESULTS The most widely used model was the nonlinear model which considers the behavior of porous elastic materials. As more advanced methods, More and more models which involve penetration parameters were used to simulate the biological behavior and biomechanical properties of IVD. CONCLUSIONS Personalized modeling should be carried out in order to better provide accurate basis for the diagnosis and treatment of the disease. In addition, microstructure, cell behavior and complex load should be considered in the process of model construction to build a more realistic model.
Collapse
Affiliation(s)
- Tianze Sun
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, The People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, Dalian, The People’s Republic of China
| | - Junlin Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, The People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, Dalian, The People’s Republic of China
| | - Xin Liu
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, The People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, Dalian, The People’s Republic of China
| | - Huagui Huang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, The People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, Dalian, The People’s Republic of China
| | - Jinzuo Wang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, The People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, Dalian, The People’s Republic of China
| | - Moran Suo
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, The People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, Dalian, The People’s Republic of China
| | - Jing Zhang
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, The People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, Dalian, The People’s Republic of China
| | - Zhonghai Li
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, The People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopedic Diseases, Liaoning Province, Dalian, The People’s Republic of China
| |
Collapse
|
8
|
Zheng XQ, Wang DB, Jiang YR, Song CL. Gut microbiota and microbial metabolites for osteoporosis. Gut Microbes 2025; 17:2437247. [PMID: 39690861 DOI: 10.1080/19490976.2024.2437247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/13/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024] Open
Abstract
Osteoporosis is an age-related bone metabolic disease. As an essential endocrine organ, the skeletal system is intricately connected with extraosseous organs. The crosstalk between bones and other organs supports this view. In recent years, the link between the gut microecology and bone metabolism has become an important research topic, both in preclinical studies and in clinical trials. Many studies have shown that skeletal changes are accompanied by changes in the composition and structure of the gut microbiota (GM). At the same time, natural or artificial interventions targeting the GM can subsequently affect bone metabolism. Moreover, microbiome-related metabolites may have important effects on bone metabolism. We aim to review the relationships among the GM, microbial metabolites, and bone metabolism and to summarize the potential mechanisms involved and the theory of the gut‒bone axis. We also describe existing bottlenecks in laboratory studies, as well as existing challenges in clinical settings, and propose possible future research directions.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Ding-Ben Wang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Yi-Rong Jiang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| |
Collapse
|
9
|
Zhang L, Fang L, Zou J, Zhou D, Xie H, Chen A, Wu Q. Causal associations of metabolic dysfunction-associated steatotic liver disease with gestational hypertension and preeclampsia: a two-sample Mendelian randomization study. Hypertens Pregnancy 2025; 44:2441862. [PMID: 39704480 DOI: 10.1080/10641955.2024.2441862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/08/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Hypertensive disorders of pregnancy (HDPs), which include gestational hypertension (GH) and preeclampsia (PE), are the primary causes of maternal morbidity and mortality worldwide. Recent studies have found a correlation between metabolic dysfunction-associated steatotic liver disease (MASLD) and HDPs, but the causality of this association remains to be identified. Therefore, this study aims to evaluate the causal relationship between MASLD and HDPs through Mendelian randomization (MR) analysis. METHODS The summary statistics from genome-wide association studies were employed to conduct a two-sample MR analysis. Five complementary MR methods, including inverse variance weighting (IVW), MR-Egger, weighted median, simple mode and weighted mode were performed to assess the causality of MASLD on GH and PE. Furthermore, we conducted various sensitivity analyses to ensure the stability and reliability of the results. RESULTS Genetically predicted MASLD significantly increased the risk of GH (IVW: OR = 1.138, 95% CI: 1.062-1.220, p < 0.001), while there was little evidence of a causal relationship between MASLD and PE (IVW: OR = 0.980, 95% CI: 0.910-1.056, p = 0.594). The sensitivity analyses indicated no presence of heterogeneity and horizontal pleiotropy. CONCLUSION This MR study provided evidence supporting the causal effect of MASLD on GH. Our findings underscore the significance of providing more intensive prenatal care and early intervention for pregnant women with MASLD to prevent potential adverse obstetric outcomes.
Collapse
Affiliation(s)
- Lu Zhang
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Liang Fang
- Department of Gastroenterology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Jiahua Zou
- Department of Oncology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Dong Zhou
- Department of Oncology, Huanggang Central Hospital of Yangtze University, Huanggang, China
| | - Haonan Xie
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Aihua Chen
- Department of Gastroenterology, China Resources & WISCO General Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Qingming Wu
- School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Liu M, Gong S, Sheng X, Zhang Z, Wang X. Bioinformatic identification of important roles of COL1A1 and TNFRSF12A in cartilage injury and osteoporosis. J Int Soc Sports Nutr 2025; 22:2454641. [PMID: 39847474 PMCID: PMC11758804 DOI: 10.1080/15502783.2025.2454641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/12/2025] [Indexed: 01/25/2025] Open
Abstract
OBJECTIVE The aim of this study was to identify the key regulatory mechanisms of cartilage injury and osteoporosis through bioinformatics methods, and to provide a new theoretical basis and molecular targets for the diagnosis and treatment of the disease. METHODS Microarray data for cartilage injury (GSE129147) and osteoporosis (GSE230665) were first downloaded from the GEO database. Differential expression analysis was applied to identify genes that were significantly up-or down-regulated in the cartilage injury and osteoporosis samples. These genes were subjected to GO enrichment analysis and KEGG pathway analysis. In addition, we employed SVA and RRA methods to merge the two sets of data, eliminating batch effects and enhancing the statistical power of the analysis. Through WGCNA, we identified gene modules that were closely associated with disease phenotypes and then screened for key genes that intersected with differentially expressed genes. The diagnostic value of these genes as potential biomarkers was evaluated by ROC analysis. Moreover, we performed an immune infiltration analysis to explore the correlation between these core genes and immune cell infiltration. RESULTS We performed GO enrichment analysis and KEGG pathway analysis of genes significantly up-or down-regulated in cartilage injury and osteoporosis samples. Important biological processes, cellular components and molecular functions, and key metabolic or signaling pathways associated with osteoporosis and cartilage injury were identified. Through WGCNA, we identified gene modules that were closely associated with the disease phenotype, from which we then screened for key genes that intersected with differentially expressed genes. Ultimately, we focused on two identified core genes, COL1A1 and TNFRSF12A, and assessed the diagnostic value of these genes as potential biomarkers by ROC analysis. Meanwhile, GSVA provided an in-depth view of the role of these genes in disease-specific biological pathways. Immune infiltration analysis further revealed the possible key role of COL1A1 and TNFRSF12A in regulating immune cell infiltration in osteoporosis and cartilage injury. CONCLUSION COL1A1 and TNFRSF12A as key regulatory molecules in osteoporosis and cartilage injury.
Collapse
Affiliation(s)
- Muzi Liu
- Jiujiang No.1 People’s Hospital, Department of Orthopedics, Jiujiang City Key Laboratory of Cell Therapy, Jiujiang, China
| | - Shiguo Gong
- Jiujiang No.1 People’s Hospital, Department of Orthopedics, Jiujiang City Key Laboratory of Cell Therapy, Jiujiang, China
| | - Xin Sheng
- Jiujiang No.1 People’s Hospital, Department of Orthopedics, Jiujiang City Key Laboratory of Cell Therapy, Jiujiang, China
- The First Affiliated Hospital of Nanchang University, Department of Orthopedics, Nanchang, China
| | - Zihong Zhang
- Jiujiang No.1 People’s Hospital, Department of Orthopedics, Jiujiang City Key Laboratory of Cell Therapy, Jiujiang, China
| | - Xichun Wang
- Jiujiang No.1 People’s Hospital, Department of Orthopedics, Jiujiang City Key Laboratory of Cell Therapy, Jiujiang, China
| |
Collapse
|
11
|
Huang J, Wang L, Zhou J, Dai T, Zhu W, Wang T, Wang H, Zhang Y. Unveiling the ageing-related genes in diagnosing osteoarthritis with metabolic syndrome by integrated bioinformatics analysis and machine learning. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2025; 53:57-68. [PMID: 40022676 DOI: 10.1080/21691401.2025.2471762] [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: 08/12/2024] [Revised: 12/16/2024] [Accepted: 02/16/2025] [Indexed: 03/03/2025]
Abstract
Ageing significantly contributes to osteoarthritis (OA) and metabolic syndrome (MetS) pathogenesis, yet the underlying mechanisms remain unknown. This study aimed to identify ageing-related biomarkers in OA patients with MetS. OA and MetS datasets and ageing-related genes (ARGs) were retrieved from public databases. The limma package was used to identify differentially expressed genes (DEGs), and weighted gene coexpression network analysis (WGCNA) screened gene modules, and machine learning algorithms, such as random forest (RF), support vector machine (SVM), generalised linear model (GLM), and extreme gradient boosting (XGB), were employed. The nomogram and receiver operating characteristic (ROC) curve assess the diagnostic value, and CIBERSORT analysed immune cell infiltration. We identified 20 intersecting genes among DEGs of OA, key module genes of MetS, and ARGs. By comparing the accuracy of the four machine learning models for disease prediction, the SVM model, which includes CEBPB, PTEN, ARPC1B, PIK3R1, and CDC42, was selected. These hub ARGs not only demonstrated strong diagnostic values based on nomogram data but also exhibited a significant correlation with immune cell infiltration. Building on these findings, we have identified five hub ARGs that are associated with immune cell infiltration and have constructed a nomogram aimed at early diagnosing OA patients with MetS.
Collapse
Affiliation(s)
- Jian Huang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Wang
- Department of Neurology, The Central Hospital of Xiaogan, Xiaogan, China
| | - Jiangfei Zhou
- Department of Orthopedics, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Tianming Dai
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Weicong Zhu
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Tianrui Wang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hongde Wang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, China
- Beijing Key Laboratory of Sports Injuries, Beijing, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, China
| | - Yingze Zhang
- Department of Orthopedics, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
12
|
Jin X, Lu Y, Fan Z. Exploring NamiRNA networks and time-series gene expression in osteogenic differentiation of adipose-derived stem cells. Ann Med 2025; 57:2478323. [PMID: 40100054 PMCID: PMC11921168 DOI: 10.1080/07853890.2025.2478323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 02/12/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Adipose-derived stem cells (ADSCs) are a type of stem cell found in adipose tissue with the capacity to differentiate into multiple lineages, including osteoblasts. The differentiation of ADSCs into osteoblasts underlies osteogenic and pathological cellular basis in osteoporosis, bone damage and repair. METHODS Focused on ADSCs osteogenic differentiation, we conducted mRNA, microRNA expression and bioinformatics analysis, including gene differential expression, time series-based trend analysis, functional enrichment, and generates potential nuclear activating miRNAs (NamiRNA) regulatory network. The screened mRNAs in NamiRNA regulatory network were validated with correlation analysis. RESULTS The NamiRNA Regulatory Network reveals 4 mRNAs (C12orf61, MIR31HG, NFE2L1, and PCYOX1L) significantly downregulated in differentiated group and may be associated with ADSCs stemness. Furthermore, the significantly upregulated 10 genes (ACTA2, TAGLN, LY6E, IFITM3, NGFRAP1, TCEAL4, ATP5C1, CAV1, RPSA, and KDELR3) were significantly enriched in osteogenic-related pathways, and negatively correlated with ADSCs cell stemness in vitro. CONCLUSION These findings uncover potential genes related to ADSCs osteogenic differentiation, and provide theoretical basis for underlying ADSCs osteogenic differentiation and related diseases.
Collapse
Affiliation(s)
- Xin Jin
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yi Lu
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhihong Fan
- Department of Plastic Surgery, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Parolin M, Caruso A, Sartorato P, Lisotti A, Benvenuti S, De Menis E, Fusaroli P. Ectopic mediastinal parathyroid diagnosed by contrast-enhanced endoscopic ultrasound-guided fine-needle aspiration and eluate parathormone level. Endoscopy 2025; 57:E126-E127. [PMID: 39914481 PMCID: PMC11802267 DOI: 10.1055/a-2516-2740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Affiliation(s)
- Matteo Parolin
- Medicina Generale 2, Ospedale Santa Maria di Ca Foncello, Treviso, Italy
| | - Antonino Caruso
- Gastroenterology, Ospedale Santa Maria di Ca Foncello, Treviso, Italy
| | - Paola Sartorato
- Medicina Generale 2, Ospedale Santa Maria di Ca Foncello, Treviso, Italy
| | - Andrea Lisotti
- Gastroenterology and Digestive Endoscopy Unit, Hospital of Imola, Università di Bologna, Imola, Italy
| | - Stefano Benvenuti
- Gastroenterology, Ospedale Santa Maria di Ca Foncello, Treviso, Italy
| | - Ernesto De Menis
- Medicina Generale 2, Ospedale Santa Maria di Ca Foncello, Treviso, Italy
| | - Pietro Fusaroli
- Gastroenterology and Digestive Endoscopy Unit, Hospital of Imola, Università di Bologna, Imola, Italy
| |
Collapse
|
14
|
Zhao WQ, Yu KQ, Xie RZ, Liang YF, Huang JF. Risk factors for periprosthetic femoral fractures following hip arthroplasty: a systematic review and meta-analysis. Ann Med 2025; 57:2494679. [PMID: 40265296 DOI: 10.1080/07853890.2025.2494679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 02/25/2025] [Accepted: 04/04/2025] [Indexed: 04/23/2025] Open
Abstract
INTRODUCTION Periprosthetic femoral fracture (PPFF) is a serious complication following hip arthroplasty. The objective of this study was to determine the risk factors for PPFF following hip arthroplasty from existing studies. METHODS A comprehensive systematic search was performed in 4 databases: Pubmed, Embase, Web of Science, and Cochrane Library. The last search was carried out on 26th July 2024. We focused on identifying risk factors for PPFF following hip arthroplasty. Study eligibility required PPFF as an outcome and reporting of associated risk factors. Quality assessment was performed using the Newcastle-Ottawa Scale (NOS), with evidence certainty evaluated via Grading of Recommendations, Assessment, Development, and Evaluations (GRADE). Meta-analyses employed both fixed-effect and random-effects models to pool odds ratios for identified risk factors. RESULTS Out of 1553 articles, 36 studies published between 2006 and 2024 were included. Risk factors associated with increased incidence of PPFF ranges from very Low to High. High-quality evidence supported the use of uncemented stems (Odds Ratio [OR]: 3.36, 95% Confidence Interval [95% CI]: 3.02-3.74), major teaching hospital (OR: 2.04, 95% CI: 1.37-3.05). Moderate-quality evidence: female gender (OR: 1.60, 95% CI: 1.43-1.78), morbid obesity (OR: 1.44, 95% CI: 1.01-2.16), higher Deyo-Charlson index (OR: 1.44, 95% CI: 1.18-1.77), rheumatoid arthritis (OR: 1.89, 95% CI: 1.16-3.06), femoral Dorr type C (OR: 4.23, 95% CI: 2.82-6.33). Low evidence: age > 70 years (OR: 1.67, 95% CI: 1.19-2.34), revision hip arthroplasty (OR: 2.60, 95% CI: 1.59-4.27). BMI > 30 and history of hip surgery are not the risk (very low). Diagonized as osteoarthritis before surgery is a protective factor (OR:0.51, 95%CI: 0.40-0.65, quality = High). CONCLUSION This meta-analysis provided some low-to-high evidence about the risk of PPFF following hip arthroplasty. It's recommended that clinicians consider these risk factors when evaluating patients for hip arthroplasty and take steps to mitigate their impact, like optimizing patients health preoperatively, using cemented stems, and monitoring high-risk patients closely.
Collapse
Affiliation(s)
- Wei-Qiang Zhao
- Department of Orthopaedics & Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ke-Qin Yu
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Rong-Zhen Xie
- Department of Orthopaedics & Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu-Feng Liang
- Department of Orthopaedics & Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie-Feng Huang
- Department of Orthopaedics & Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
15
|
Liu Y, Zuo L, Xin Y, Liu Y, Tian Z, Shang X. Radical Resection of Differentiated Thyroid Cancer in Elderly Patients: Evaluation of the Efficacy of the Immunocolloidal Gold Strip Method Combined with Nanocarbon Negative Imaging Tracing Technology for Parathyroid Gland Imaging. J INVEST SURG 2025; 38:2447850. [PMID: 39807041 DOI: 10.1080/08941939.2024.2447850] [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: 07/16/2024] [Accepted: 12/22/2024] [Indexed: 01/16/2025]
Abstract
OBJECTIVE Extant imaging methods used for the proper identification of the parathyroid glands to prevent post-operative hypothyroidism associated with the resection of differentiated thyroid cancer (DTC) are limited by factors such as low specificity, high cost, and technical complexity. This study, therefore, sought to investigate the efficacy of the immunocolloidal gold strip method combined with nanocarbon negative imaging tracing technology for parathyroid gland imaging during radical resection of DTC in elderly patients. METHODS A total of 100 elderly patients with DTC were enrolled and randomly divided into two groups: the control group and the observation group. The control group underwent conventional radical thyroidectomy with bilateral cervical lymph node dissection, while the observation group received the immunocolloidal gold strip method combined with nanocarbon negative imaging tracing technology for parathyroid gland imaging during the surgery. The baseline characteristics, intraoperative findings, postoperative parathyroid hormone (PTH), and serum calcium levels, as well as postoperative complications, were compared between the two groups. RESULTS There were no significant differences in age, gender, body mass index, comorbidities, or smoking history between the two groups. The observation group had a significantly higher number of parathyroid glands identified during surgery compared with the control group. The postoperative PTH and serum calcium levels at postoperative days 1 and 3 and at 6 months were significantly higher in the observation group than those in the control group. The incidence of postoperative hypoparathyroidism was significantly lower in the observation group. CONCLUSION The immunocolloidal gold strip method combined with nanocarbon negative imaging tracing technology is effective in identifying and preserving parathyroid glands during radical resection of DTC in elderly patients.
Collapse
Affiliation(s)
- YanBin Liu
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - LiJuan Zuo
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - YunChao Xin
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - YaChao Liu
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - ZeDong Tian
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - XiaoLing Shang
- The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| |
Collapse
|
16
|
Yan J, Gu Q, Li J, Zhou Z, Jiang W, Guan W, Chen B, Chen Y, Yang M. MS-275 facilitates osseointegration in osteoporotic rats by mitigating oxidative stress via activation of the miR-200a/Keap1/Nrf2 signaling pathway. Redox Rep 2025; 30:2466142. [PMID: 39973077 PMCID: PMC11843653 DOI: 10.1080/13510002.2025.2466142] [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] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVES Osteoporosis, a prevalent metabolic bone disease affecting millions worldwide. Although MS-275 has been reported to inhibit oxidative stress, its ability to protect osteoblasts from oxidative stress damage has yet to be clarified. This study investigated whether MS-275 can inhibit oxidative stress and promote osteogenesis by activating the miRNA-200a/Keap1/Nrf2 signaling pathway. METHODS In vitro, MC3T3-E1 cells underwent induction with carbonyl cyanide 3-chlorophenylhydrazone, leading to the establishment of an oxidative stress model, investigating the underlying mechanism. In vivo, using a rat model of ovariectomized osteoporosis, evaluating the effects of MS-275. RESULTS In vitro, MS-275 treatment of oxidation-induced MC3T3-E1 cells resulted in up-regulation of osteoblast protein, increased expression of miRNA-200a, increased binding of miRNA-200a to Keap1 mRNA, decreased expression of Keap1 protein, and dissociation of Nrf2 from Keap1. The expressions of total Nrf2, nuclear Nrf2 and HO-1 were increased, mitochondrial function was enhanced, and oxidative damage was reduced. However, these effects were reversed after interference with miRNA-200a. In vivo,MS-275 effectively enhanced the microstructural features of distal femoral trabecular bone, increased the mineralization capacity of osteoblasts, and promoted bone formation. DISCUSSION MS-275 can reverse oxidative stress-induced cell damage, promote bone healing, and improve osteoporosis by activating the miRNA-200a/Keap1/Nrf2 pathway.
Collapse
Affiliation(s)
- Junjie Yan
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, People’s Republic of China
| | - Qinsong Gu
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, People’s Republic of China
| | - Jianqiao Li
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, People’s Republic of China
| | - Zhi Zhou
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, People’s Republic of China
| | - Wenkai Jiang
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, People’s Republic of China
| | - Wengang Guan
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, People’s Republic of China
| | - Bin Chen
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, People’s Republic of China
| | - Yuhu Chen
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, People’s Republic of China
| | - Min Yang
- Department of Trauma Orthopedics, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital, Wuhu, People’s Republic of China
| |
Collapse
|
17
|
Liu B, Wang S, Meng F, Wu B, Zhang Y, Cao J. Study on immortalization of Mongolian sheep fibroblast cells. Anim Biotechnol 2025; 36:2459915. [PMID: 39918276 DOI: 10.1080/10495398.2025.2459915] [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: 07/12/2024] [Accepted: 01/22/2025] [Indexed: 05/08/2025]
Abstract
This study aims to establish an immortalized fibroblast cell line from Mongolian sheep. Primary Mongolian sheep fibroblasts (SSF) were isolated using tissue explant and enzymatic digestion methods, followed by microscopic observation, growth curve plotting, and karyotype analysis. The results confirmed the successful isolation of SSF. Human (hTERT) and sheep (sTERT) telomerase reverse transcriptase vectors were separately introduced into SSF, with cells passaged up to 36 generations following G418 selection. Microscopic examination and qRT-PCR results demonstrated that TERT transfection did not alter the morphology of SSF and led to stable, high levels of TERT expression (P < 0.01). Cell counting and flow cytometry revealed that TERT-transfected cells had higher viability and lower apoptosis rates compared to SSF (P < 0.05). Karyotype and soft agar colony formation assays indicated that hTERT and sTERT-transfected cells maintained normal characteristics without malignant transformation. β-galactosidase staining indicated that TERT transfection significantly reduced cellular senescence (P < 0.001). Additionally, sTERT-transfected cells exhibited higher TERT expression, enhanced viability, proliferation, and anti-senescence effects compared to hTERT-transfected cells (P < 0.05). In summary, the introduction of hTERT and sTERT effectively extends the lifespan of SSF, with sTERT demonstrating a more pronounced effect. This study provides critical evidence for preserving Mongolian sheep genetic resources and developing immortalized cell lines.
Collapse
Affiliation(s)
- Bin Liu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, People's Republic of China
| | - Shichao Wang
- Department of Medical Laboratory, Hohhot First Hospital, Hohhot, Inner Mongolia, People's Republic of China
| | - Fanhua Meng
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, People's Republic of China
| | - Bei Wu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, People's Republic of China
| | - Yanru Zhang
- College of Medicine, Hainan Vocational University of Science and Technology, Haikou, Hainan, People's Republic of China
| | - Junwei Cao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, People's Republic of China
| |
Collapse
|
18
|
Yendluri A, Yu J, Stocchi C, Kurapatti M, Namiri NK, Song J, Corvi JJ, Cordero JK, Yacovelli S, Hayden BL, Forsh DA. Sociodemographic variables are rarely included in femoral neck fracture randomized controlled trials: A systematic review. J Orthop 2025; 69:79-85. [PMID: 40183034 PMCID: PMC11964601 DOI: 10.1016/j.jor.2025.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/14/2025] [Indexed: 04/05/2025] Open
Abstract
Introduction Sociodemographic factors may affect outcomes after surgery for patients with femoral neck fractures. The purpose of this study was to assess the inclusion of sociodemographic variables in high-impact randomized controlled trials (RCTs) related to femoral neck fracture operative management. Methods PubMed, Embase, and Medline were queried from January 1, 2017 to March 31, 2024 for RCTs pertaining to operative treatment of femoral neck fracture patients in high impact journals were included. The journal of publication, year of publication, and interventions assessed by the RCTs were extracted. Each RCT was assessed for inclusion of the following sociodemographic variables: age, sex/gender, body mass index (BMI)/weight, race/ethnicity, education level, insurance, smoking/tobacco use, socioeconomic status, marital status, alcohol use, English proficiency, geographic measures (i.e. proximity to hospital), employment status, and prefracture residence status. Temporal reporting trends were analyzed using Chi-square test. Results Of 1038 RCTs identified, 37 were included for analysis. All 37 studies reported age and sex/gender. BMI/weight was reported in 22 studies (59.5 %). Patients' prefracture residence status was reported in 11 studies (29.7 %). Smoking/tobacco use was reported in 9 studies (24.3 %). Race/ethnicity was reported in only 5 studies (13.5 %). Socioeconomic status, English proficiency, geographic measures, marital status, education level, insurance, and employment variables were all reported in less than 10 % of the analyzed RCTs. Furthermore, there was no significant difference in the proportion of studies reporting at least one sociodemographic variable (excluding age, sex/gender, and BMI/weight) in 2017-2020 (10/22) versus 2021-2024 (8/15; p = 0.743). Conclusion Our analysis of high-impact RCTs revealed a large gap in the reporting of sociodemographic variables. RCTs relating to femoral neck fracture management should consistently report key sociodemographic variables to ensure generalizability of study findings. Level of evidence 1.
Collapse
Affiliation(s)
- Avanish Yendluri
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jennifer Yu
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolina Stocchi
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark Kurapatti
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nikan K. Namiri
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Junho Song
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John J. Corvi
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John K. Cordero
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven Yacovelli
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brett L. Hayden
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David A. Forsh
- Department of Orthopaedic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
19
|
Yang Y, Zeng Q, Zhao C, Shi J, Wang W, Liang Y, Li C, Guan Q, Chen B, Li W. Metal-free antioxidant nanozyme incorporating bioactive hydrogel as an antioxidant scaffold for accelerating bone reconstruction. Biomaterials 2025; 320:123285. [PMID: 40127506 DOI: 10.1016/j.biomaterials.2025.123285] [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: 12/17/2024] [Revised: 01/30/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
Oxidative stress at bone defect sites mediates inflammation and even osteoblast apoptosis, severely hindering the repair process. While current antioxidant bone tissue engineering (BTE) scaffolds lack broad-spectrum reactive oxygen species (ROS) scavenging capability and structure-activity elucidation. Herein, we report a three-dimensional nitrogen-doped carbon antioxidant nanozyme (ZIFC) derived from metal-organic frameworks, which exhibits cascading superoxide dismutase- and catalase-like activities, along with the ability to scavenge other harmful free radicals. Through the experimental studies and theoretical calculations, we reveal that the catalase-like activity arises from the synergistic catalytic interaction between graphitized pyridinic nitrogen and its adjacent carbon atom. Moreover, hybrid double network hydrogel integrated with ZIFC is utilized to construct composite scaffold (Gel/ZIFC) by 3D printing. In vivo transcriptome analysis confirms that Gel/ZIFC can rapidly activate antioxidant defense system and suppress local inflammation under oxidative stress microenvironment, thereby protecting cells from oxidative damage. Subsequently, owing to the unique osteoinductive property of carbon nanomaterials and the osteoconductive property of 3D-printed scaffold, Gel/ZIFC composite scaffold exhibits desirable bone repair efficacy. The elucidation of structure-activity relationship and therapeutic mechanism provides new insights and guidance for devising antioxidant BTE scaffolds, and demonstrates their feasibility for clinical application.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, PR China
| | - Qianrui Zeng
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, PR China
| | - Chaoyue Zhao
- Ningbo Key Laboratory of Agricultural Germplasm Resources Mining and Environmental Regulation, College of Science and Technology, Ningbo University, Ningbo, 315000, PR China
| | - Jie Shi
- The School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Wanmeng Wang
- Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Yunkai Liang
- Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Changyi Li
- Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Qingxin Guan
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, PR China.
| | - Bo Chen
- Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China.
| | - Wei Li
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
20
|
Zhao Y, Liu J, Hu L, Yao X, Tu R, Goto T, Zhang L, Wu X, Liu G, Dai H. Novel "hot spring"-mimetic scaffolds for sequential neurovascular network reconstruction and osteoporosis reversion. Biomaterials 2025; 320:123191. [PMID: 40056610 DOI: 10.1016/j.biomaterials.2025.123191] [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: 12/03/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 03/10/2025]
Abstract
Neurovascular network damage and excessive hydrogen peroxide (H2O2) accumulation are the main obstacles for osteoporotic bone defect repair. It is extremely essential to endow the implants with sequential neuroangiogenesis promotion and osteoporosis pathological microenvironment improvement. Hot springs exhibits excellent facilitation on angiogenesis and bone regeneration due to abundant minerals, trace elements and modest thermal stimulation. Inspired by the hot spring effect, we propose a novel porous photothermal calcium magnesium phosphate bone cement (MCPC) compounded with manganese-substituted Fe3O4 (MnxFe3-xO4), which is perfused by temperature-responsive PLGA hydrogel loaded with vascular endothelial growth factor (VEGF) and nerve growth factor (NGF). At the initial stage of implantation, MnxFe3-xO4 scavenges excessive H2O2 under the heat stimulation triggered by near-infrared (NIR) light, and the factors are released from the hydrogel that stimulate the impaired neurovascular network reconstruction; at the later stage, the continuous hot spring effect maintains mild thermal stimulation and sustained release of bioactive ions (Ca2+, Mn2+, Mg2+ and PO43-), which inhibits osteoclast function and activity, and promotes osteogenic differentiation and mineralization. The osteoporotic bone defect model in ovariectomized (OVX) rats further verifies that a synergy effect of photothermal therapy and bioactive factors/ions significantly promotes neurovascular bone regeneration. It demonstrates that the hot spring mimetic effect possesses huge potential for the sequential treatment of osteoporosis bone defect.
Collapse
Affiliation(s)
- Yanan Zhao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Jiawei Liu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Liangcong Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaokang Yao
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Rong Tu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Takashi Goto
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Lianmeng Zhang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Xiaopei Wu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China; National energy key laboratory for new hydrogen-ammonia energy technologies,Foshan Xianhu Laboratory, Foshan 528200, China.
| |
Collapse
|
21
|
Yang P, Chen X, Qin Y, Yu L, Ge G, Yin W, Zhang W, Li W, Li W, Xia W, Wu Z, Ding F, Bai J, Meng F, Geng D. Regulation of osteoimmune microenvironment via functional dynamic hydrogel for diabetic bone regeneration. Biomaterials 2025; 320:123273. [PMID: 40121832 DOI: 10.1016/j.biomaterials.2025.123273] [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: 11/23/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Bone regeneration and repair face formidable challenges under diabetic conditions, primarily due to the disruption of macrophage polarization induced by diabetes and the inflammatory imbalance within the bone microenvironment. We have developed a novel dynamic hydrogel system (AG-CD@LINA), constructed through the coordination crosslinking of thiolated gelatin (SH-Gelatin) and gold ions (Au3+), followed by grafting with cyclodextrin to load the ligand linagliptin. This hydrogel effectively inhibits the formation of M1 macrophages and the expression of pro-inflammatory cytokines by gradually releasing linagliptin. Simultaneously, it promotes the formation of M2 macrophages and the expression of anti-inflammatory cytokines, thus improving the inflammatory microenvironment of diabetic bone defects. Consequently, it facilitates the migration of mesenchymal stem cells and angiogenic cells, augments osteogenic activity, and promotes vascularization, collectively accelerating the regeneration of diabetic bone tissue. Mechanistically, polarization occurs through the TLR3-NF-κB signaling pathway. In vivo experiments demonstrate that the in-situ injection of the hydrogel enhances the regeneration of bone tissue and the restoration of bone structure in diabetic bone defects, effectively modulating local inflammation and promoting vascular formation. This study suggests that functionalized dynamic hydrogels can improve the inflammatory microenvironment by regulating in situ macrophage polarization, thereby facilitating the reconstruction of bone microstructure. This approach represents a promising novel therapeutic strategy for diabetic bone defects.
Collapse
Affiliation(s)
- Peng Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China; Department of Orthopedics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, 215006, Jiangsu, China; Suzhou Key Laboratory of Orthopedic Medical Engineering, Suzhou, 215006, Jiangsu, China
| | - Xu Chen
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Yi Qin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Lei Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Gaoran Ge
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Weiling Yin
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Wei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wenming Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wenhao Li
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Wenyu Xia
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Zebin Wu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Fan Ding
- Institute for Advanced Materials, School of Materials Science and Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Jiaxiang Bai
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China.
| | - Fanwen Meng
- Department of Implant Dentistry, Suzhou Stomatological Hospital, Suzhou, 215005, China.
| | - Dechun Geng
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
22
|
Tian M, Du W, Yang S, Liao Q, Guo F, Li S. Application and progress of hyperbaric oxygen therapy in cardiovascular diseases. Med Gas Res 2025; 15:427-434. [PMID: 40251023 PMCID: PMC12054664 DOI: 10.4103/mgr.medgasres-d-24-00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/14/2024] [Accepted: 12/12/2024] [Indexed: 04/20/2025] Open
Abstract
Cardiovascular diseases remain the leading cause of death worldwide, underscoring the urgent need for additional therapeutic strategies to reduce their mortality rates. This review systematically outlines the historical development and recent advances of hyperbaric oxygen therapy in cardiovascular diseases, with a focus on its therapeutic mechanisms and clinical outcomes. Hyperbaric oxygen therapy enhances oxygen delivery to ischemic and reperfused tissues, promotes angiogenesis, and significantly suppresses oxidative stress, inflammatory cascades, and cardiomyocyte apoptosis, demonstrating multifaceted therapeutic potential in cardiovascular conditions. Specifically, hyperbaric oxygen therapy combined with reperfusion strategies has been shown to markedly improve left ventricular ejection fraction in acute myocardial infarction. In heart failure, it facilitates myocardial repair and enhances cardiac function. For arrhythmias, hyperbaric oxygen therapy effectively reduces the frequency and duration of premature ventricular contractions and paroxysmal tachycardia, while mitigating the risk of neurological complications following atrial fibrillation ablation. Furthermore, hyperbaric oxygen therapy preconditioning in cardiac surgery has demonstrated improvements in left ventricular stroke work, reductions in postoperative myocardial injury, and a decrease in related complications. Despite its promising applications, the widespread adoption of hyperbaric oxygen therapy remains hindered by the lack of standardized treatment protocols and high-quality evidence from rigorous clinical trials. In conclusion, this review underscores the potential value of hyperbaric oxygen therapy in the cardiovascular domain while highlighting the need for further optimization of therapeutic parameters and exploration of its synergistic effects with conventional therapies to provide clearer guidance for clinical implementation.
Collapse
Affiliation(s)
- Menglin Tian
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Clinical Medicine Center for Cardiovascular Disease of Yunnan Province, Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Wenyin Du
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Clinical Medicine Center for Cardiovascular Disease of Yunnan Province, Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Sen Yang
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Clinical Medicine Center for Cardiovascular Disease of Yunnan Province, Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Qiwei Liao
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Clinical Medicine Center for Cardiovascular Disease of Yunnan Province, Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Fuding Guo
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Clinical Medicine Center for Cardiovascular Disease of Yunnan Province, Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| | - Shaolong Li
- Key Laboratory of Cardiovascular Disease of Yunnan Province, Clinical Medicine Center for Cardiovascular Disease of Yunnan Province, Department of Cardiology, Yan’an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China
| |
Collapse
|
23
|
Quan H, Ren C, Xie H, He Z, Ding H, Li J, Li T, Wang F, Dong S, Jiang H. An injectable hydrogel loaded with miRNA nanocarriers promotes vessel-associated osteoclast (VAO)-mediated angiogenesis and bone regeneration in osteonecrosis of the rat femoral head. Biomaterials 2025; 320:123252. [PMID: 40081225 DOI: 10.1016/j.biomaterials.2025.123252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 02/21/2025] [Accepted: 03/09/2025] [Indexed: 03/15/2025]
Abstract
Osteonecrosis of the femoral head (ONFH) remains a significant clinical challenge. Despite various strategies aimed at promoting bone repair and halting disease progression, an effective cure remains elusive. Recent studies have identified a non-bone-resorbing osteoclast subtype, vessel-associated osteoclasts (VAOs), distinct from classical bone-associated osteoclasts (BAOs), offering new therapeutic opportunities for ONFH. Notably, we observed alterations in the populations and distributions of VAOs and BAOs in the femoral head of ONFH patients, suggesting that the imbalance between these two osteoclast subtypes contributes to ONFH pathology. Here, we developed an injectable alginate/hydroxyapatite hydrogel (AHH) loaded with graphene oxide-based miR-7b nanocarriers (GPC@miR) for ONFH treatment. The controlled release of GPC@miR from AHH/GPC@miR inhibited BAO formation by suppressing dendritic cell-specific transmembrane protein (DC-STAMP), thereby reducing bone resorption. Meanwhile, mono-/bi-nucleated VAOs were preserved and increased in number, promoting angiogenesis of type H vessels and osteogenesis via platelet-derived growth factor-BB (PDGF-BB) and vascular endothelial growth factor-A (VEGF-A) secretion. Intraosseous administration of AHH/GPC@miR rebalanced VAOs and BAOs, restored the femoral head microenvironment, and enhanced vascularization and bone regeneration in ONFH rat models. This study introduces a novel biomaterial-based strategy for ONFH repair by regulating osteoclast subtypes, providing insights into VAO-mediated angiogenesis and osteogenesis for bone regeneration.
Collapse
Affiliation(s)
- Hongyu Quan
- College of Bioengineering, Chongqing University, Chongqing, 400044, China; Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Chencan Ren
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Hongkun Xie
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Zibo He
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Haibin Ding
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Jinbao Li
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Taiyang Li
- Department of Anesthesiology, The 958th Hospital of Chinese People's Liberation Army, Chongqing, 400020, China
| | - Fuyou Wang
- Center for Joint Surgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Hong Jiang
- Department of Biomedical Materials Science, College of Biomedical Engineering, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
24
|
Bi X, Mao Z, Zhang Y, Ren Z, Yang K, Yu C, Chen L, Zheng R, Guan J, Liu Z, Yu B, Huang Y, Shu X, Zheng Y. Endogenous dual-responsive and self-adaptive silk fibroin-based scaffold with enhancement of immunomodulation for skull regeneration. Biomaterials 2025; 320:123261. [PMID: 40132357 DOI: 10.1016/j.biomaterials.2025.123261] [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: 12/15/2024] [Revised: 02/19/2025] [Accepted: 03/16/2025] [Indexed: 03/27/2025]
Abstract
Despite the current biomaterials (e.g. titanium mesh and polyether ether ketone) have been applied to clinical skull repair, the limitations on mechanical match, shape adaptability, bioactivity and osteointegration have greatly limited their clinical application. In this work, we constructed a water and inflammatory microenvironment dual-responsive self-adaptive silk fibroin-magnesium oxide-based scaffold with the matrix metalloproteinase-2-responsive gelatin-methacryloyl-interleukin-4 (IL-4) coating, which presented good mechanical compliance, quickly shape matching and intraoperative reprocessability. With the capability of responding to an acute inflammation microenvironment followed by a triggered on-demand release of the IL-4, the combination of immunoactive IL-4 and Mg2+ co-ordinately facilitated metabolic reprogramming by suppressing glycolysis, promoting mitochondrial oxidative phosphorylation and modulating adenosine 5'-monophosphate-activated protein kinase (AMPK) signalling pathways in macrophages, resulting in significantly facilitating M2 macrophage activation. During the stage of tissue remodelling, the sustained release of Mg2+ further promoted macrophage M2 polarization and the expression of anti-inflammatory cytokines, significantly reduced immune response and improved ectopic osteogenesis ability. Meanwhile, the cranial defect models of male rats demonstrated that this scaffold could significantly enhance biomineralized deposition and vascularisation, and achieve good bone regeneration of cranial defects. Overall, the bioactive scaffold provides a promising biomaterial and alternative repair strategy for critical-size skull defect repair.
Collapse
Affiliation(s)
- Xuewei Bi
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China; School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Zhinan Mao
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China; School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| | - Yilin Zhang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China
| | - Zeqi Ren
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China
| | - Kang Yang
- School of Materials Science and Engineering, Anhui University of Technology, Maanshan 243002, China
| | - Chunhao Yu
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China; School of Life, Beijing Institute of Technology, No.5, Zhongguancun South Street, Haidian District, Beijing, China
| | - Lei Chen
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Rui Zheng
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Juan Guan
- International Research Center for Advanced Structural and Biomaterials, School of Materials Science & Engineering, Beihang University, Beijing 100191, China
| | - Zhenhai Liu
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Binsheng Yu
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China
| | - Yongcan Huang
- Shenzhen Engineering Laboratory of Orthopaedic Regenerative Technologies, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, China.
| | - Xiong Shu
- Beijing Research Institute of Orthopedics and Traumatology, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
| | - Yufeng Zheng
- School of Materials Science and Engineering, Peking University, Beijing, 100871, China.
| |
Collapse
|
25
|
Ma L, Li Y, Wang CS, Chen ZH, Zhao SY, Cheng B, Li CL. Bioactive Zn ingredients endow Ti-Zn composites with exceptional mechanical and osteogenic properties as biomedical implants. BIOMATERIALS ADVANCES 2025; 174:214308. [PMID: 40215942 DOI: 10.1016/j.bioadv.2025.214308] [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: 01/20/2025] [Revised: 03/15/2025] [Accepted: 04/04/2025] [Indexed: 05/07/2025]
Abstract
Titanium-based (Ti-) alloys are promising materials as bioimplants with superior mechanical properties and excellent biocompatibility. However, their bioinertia and high elastic moduli are not comparable to natural bone tissue; thus, novel Ti alloys with good biomechanical adaptation and high bioactivity are desired. Zinc (Zn) is recognized for ideal biodegradability and its biological effects can be considered to endow pure Ti with rewarding bio functions. Herein, this study has employed a designed spark plasma sintering (SPS) procedure to effectively diffuse varying amounts of Zn into pure Ti as bioactive ingredients and generate novel TiZn composites as bone defect implants. The as-sintered TiZn samples feature a gradient core-shell structure, achieving a match of high strength and low elastic moduli to satisfy the load-bearing requirements while avoiding the stress-shielding effect. A moderate degradation of the Zn component allows TiZn materials to maintain stable mechanical support and exhibit satisfactory cytocompatibility. Ti20Zn, Ti30Zn, and Ti90Zn are confirmed to exert antibacterial and osteogenic abilities by in vitro experiments. Further analyses of in vivo implantation in the rat femur show they exhibit qualified biosafety and are superior to pure Ti in treating bone defects through bio-friendly Zn ions release. This study achieves a reasonable combination of the mechanical properties of pure Ti and the biological functions of pure Zn, providing a better choice for bone injury and fracture treatment.
Collapse
Affiliation(s)
- Li Ma
- School of Power and Mechanical Engineering, Wuhan University, Wuhan 430072, China.
| | - Yue Li
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China.
| | - Chang-Shun Wang
- School of Power and Mechanical Engineering, Wuhan University, Wuhan 430072, China.
| | - Zi-Hao Chen
- School of Power and Mechanical Engineering, Wuhan University, Wuhan 430072, China.
| | - Si-Yu Zhao
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China.
| | - Bo Cheng
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China.
| | - Cheng-Lin Li
- School of Power and Mechanical Engineering, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
26
|
Jia P, Yang Y, Tang X. Global trends in proximal femoral trabecular research: A bibliometric and visualized analysis. J Orthop 2025; 66:84-91. [PMID: 39896859 PMCID: PMC11779657 DOI: 10.1016/j.jor.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/25/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Introduction Hip disease is a global public health issue, associated with high morbidity, mortality, and healthcare costs. Although research on proximal femoral trabeculae has been conducted for over a century, no bibliometric analysis has been carried out. The purpose of this study is to evaluate the existing research landscape, identify emerging trends, and offer insights for future studies. Method The scientific output related to the trabeculae within the human proximal femur from 2004 to 2023 was sourced from the Web of Science Core Collection. Moreover, both the annual publications and cumulative totals over this period were summarized in Excel. The VOS viewer was utilized to analyze co-authorship and co-citation relationship between authors, institutions, countries, references and journals. CiteSpace was used to cluster the keywords and research frontiers in this field. Results A total of 365 publications were extracted, with the USA emerging as the primary contributor to this field, accounting for 133 publications with 5807 total citations, averaging 43.7 citations per publication. The Journal of Bone and Mineral Research has been identified as the most co-cited journal with a total of 1742 citations. The journals can be categorized into 5 distinct clusters, including medical imaging, orthopedic clinical research, research on endocrine and metabolic related diseases, human evolution and anatomy related research, biomechanics and modeling. The keyword with the highest co-occurrence frequency is "bone mineral density". The keywords were stratified into six clusters, including DXA, bone remodeling, diagnosis, titanium alloy bionic cannulated screws, individual trabecula segmentation, and QCT. More recently, the focus has expanded to three-dimensional modeling, falls, microarchitecture, and avascular necrosis. Conclusions Evaluation of proximal femoral strength can be improved by combining structural parameters with bone mineral density by DXA or QCT. Three-dimensional analysis, microarchitecture, and bionic implants are emerging as significant areas of focus and trends for future research.
Collapse
Affiliation(s)
- Peng Jia
- Department of Orthopedics, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
- Department of Orthopedics, Shandong Second Provincial General Hospital, Jinan, Shandong, 250023, China
| | - Yi Yang
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
| | - Xin Tang
- Department of Orthopedics, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, China
| |
Collapse
|
27
|
Tandon B, Aguilar Cosme JR, Xue R, Srirussamee K, Aguilar-Tadeo J, Ballestrem C, Blaker JJ, Cartmell SH. Co-stimulation with piezoelectric PVDF films and low intensity pulsed ultrasound enhances osteogenic differentiation. BIOMATERIALS ADVANCES 2025; 173:214283. [PMID: 40086006 DOI: 10.1016/j.bioadv.2025.214283] [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: 01/03/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
Bone tissue engineering has emerged as a promising approach to address the challenges of bone fracture repair and regeneration. The application of external stimuli (mechanical and electrical) can drive specific cellular responses and osteogenic differentiation, leading to the development of more effective treatments. Piezoelectric materials modulate cellular proliferation and osteogenic differentiation under both static (without mechanical stimulation) and dynamic (with mechanical stimulation) conditions, activating distinct gene expression pathways. In this work, we investigate the combinatorial effect of poly (vinylidene fluoride) (PVDF) poled and non-poled films, and low-intensity pulsed ultrasound (LIPUS) on early-stage osteogenic differentiation of mouse pre-osteoblasts. Static culture with PVDF poled films enhanced Runx2 and Col1α1 expression without impacting alkaline phosphatase (ALP) activity. Inhibition of ERK phosphorylation using U0126 in PVDF poled films resulted in a ~ 6-8-fold increase in ALP activity, suggesting the involvement of an alternative pathway in osteogenic differentiation. Dynamic culture with LIPUS generated an electric potential of approximately 500 mV across PVDF films and an electrical field of 0-10 mV mm-1. Co-stimulation led to a ~3-fold increase of ALP activity on stimulated PVDF compared to unstimulated films. This study underscores the potential of piezoelectric materials as non-invasive electrical stimulators to enhance the efficacy of ultrasound-based therapies for bone fracture repair.
Collapse
Affiliation(s)
- Biranche Tandon
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Jose R Aguilar Cosme
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Ruikang Xue
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Department of Mechanical Engineering, Faculty of Engineering Science, University College London, London WC1E 7JE, UK
| | - Kasama Srirussamee
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Department of Biomedical Engineering, School of Engineering, KMITL, Bangkok 10520, Thailand
| | - Julio Aguilar-Tadeo
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK
| | - Christoph Ballestrem
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK
| | - Jonny J Blaker
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Henry Royce Institute, The University of Manchester, Manchester, UK
| | - Sarah H Cartmell
- Department of Materials Science, School of Natural Sciences, Faculty of Science and Engineering, The University of Manchester, Manchester, UK; Henry Royce Institute, The University of Manchester, Manchester, UK.
| |
Collapse
|
28
|
Wen Z, Li S, Liu Y, Liu X, Qiu H, Che Y, Bian L, Zhou M. An engineered M2 macrophage-derived exosomes-loaded electrospun biomimetic periosteum promotes cell recruitment, immunoregulation, and angiogenesis in bone regeneration. Bioact Mater 2025; 50:95-115. [PMID: 40242509 PMCID: PMC12002949 DOI: 10.1016/j.bioactmat.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/04/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
The periosteum, a fibrous tissue membrane covering bone surfaces, is critical to osteogenesis and angiogenesis in bone reconstruction. Artificial periostea have been widely developed for bone defect repair, but most of these are lacking of periosteal bioactivity. Herein, a biomimetic periosteum (termed PEC-Apt-NP-Exo) is prepared based on an electrospun membrane combined with engineered exosomes (Exos). The electrospun membrane is fabricated using poly(ε-caprolactone) (core)-periosteal decellularized extracellular matrix (shell) fibers via coaxial electrospinning, to mimic the fibrous structure, mechanical property, and tissue microenvironment of natural periosteum. The engineered Exos derived from M2 macrophages are functionalized by surface modification of bone marrow mesenchymal stem cell (BMSC)-specific aptamers to further enhance cell recruitment, immunoregulation, and angiogenesis in bone healing. The engineered Exos are covalently bonded to the electrospun membrane, to achieve rich loading and long-term effects of Exos. In vitro experiments demonstrate that the biomimetic periosteum promotes BMSC migration and osteogenic differentiation via Rap1/PI3K/AKT signaling pathway, and enhances vascular endothelial growth factor secretion from BMSCs to facilitate angiogenesis. In vivo studies reveal that the biomimetic periosteum promotes new bone formation in large bone defect repair by inducing M2 macrophage polarization, endogenous BMSC recruitment, osteogenic differentiation, and vascularization. This research provides valuable insights into the development of a multifunctional biomimetic periosteum for bone regeneration.
Collapse
Affiliation(s)
- Zhuohao Wen
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Shuyi Li
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yi Liu
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Xueyan Liu
- Affiliated Stomatology Hospital of Guangzhou Medical University, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, 510182, China
| | - Huiguo Qiu
- Zhuhai Stomatological Hospital, Zhuhai, 519000, China
| | - Yuejuan Che
- Department of Biomedical Engineering, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510641, China
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, China
| | - Miao Zhou
- Department of Stomatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| |
Collapse
|
29
|
Ma T, Liu Q, Zhang Z, Nan J, Liu G, Yang Y, Hu Y, Xie J. Fused exosomal targeted therapy in periprosthetic osteolysis through regulation of bone metabolic homeostasis. Bioact Mater 2025; 50:171-188. [PMID: 40248188 PMCID: PMC12005309 DOI: 10.1016/j.bioactmat.2025.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/18/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
The onset of periprosthetic osteolysis is mediated by wear particles following artificial arthroplasty. This manifests as a disturbed bone metabolism microenvironment, characterized by insufficient osteogenesis and angiogenesis, and enhanced osteoclastic activity. To target and remodel the homeostatic environment of bone metabolism in the sterile region around the prosthesis, we successfully pioneered the proposal and construction of a fused exosome (f-exo) system with M2 macrophage-derived exosomes (M2-exo) and urine-derived stem cell exosomes (USC-exo). The results demonstrate that f-exo effectively combines the osteolysis region-targeting capabilities of M2-exo with the bone metabolic homeostasis modulation effects of two exosomes (M2-exo and USC-exo), thereby achieving a significantly enhanced bone metabolic homeostasis targeting effect in the periprosthetic osteolysis region. The proteomic analysis of M2-exo, USC-exo, and f-exo revealed the potential mechanism of f-exo in targeting-regulation of bone metabolic homeostasis. Our study employs an innovative approach utilizing the fused exosome system for exosome targeted delivery, which offers a novel intervention strategy for the clinical management of periprosthetic osteolysis. Furthermore, it provides a novel conceptual framework for the development of exosome-based drug-targeting delivery systems.
Collapse
Affiliation(s)
| | | | - Zheyu Zhang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, Zhejiang, 310003, China
| | - Jiangyu Nan
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, Zhejiang, 310003, China
| | - Guanzhi Liu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, Zhejiang, 310003, China
| | - Yute Yang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, Zhejiang, 310003, China
| | - Yihe Hu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, Zhejiang, 310003, China
| | - Jie Xie
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, No.79 Qingchun Road, Hangzhou, Zhejiang, 310003, China
| |
Collapse
|
30
|
Gill AS, Sharma P, Nassar M, Marte E. Hypophosphatasia: A case report. World J Clin Cases 2025; 13:103642. [DOI: 10.12998/wjcc.v13.i21.103642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/28/2025] [Accepted: 03/27/2025] [Indexed: 04/27/2025] Open
Abstract
BACKGROUND Hypophosphatasia (HPP) is a rare metabolic disorder caused by low tissue-nonspecific alkaline phosphatase (ALP) activity, presenting symptoms from bone demineralization to tooth loss. It affects multiple systems and is diagnosed based on clinical symptoms, radiological findings, and lab tests. This case report emphasizes considering HPP in patients with unexplained bone pain and low ALP levels, especially with underlying osteopenia or osteoporosis. It highlights the importance of genetic testing and counseling for early diagnosis and treatment, aiming to raise clinician awareness.
CASE SUMMARY We present a case of a 65-year-old female patient who was referred to our endocrinology clinic for complaints of generalized bone pain and hypothyroidism. Initial evaluation revealed osteopenia, managed with calcium and vitamin D supplementation. Persistently low ALP levels and elevated vitamin B6 levels led to the diagnosis of HPP, confirmed by genetic testing identifying a pathogenic ALPL gene variant [c.119C>T (p.Ala40Val)]. Despite conservative treatment, her bone density declined, although remaining in the osteopenic range. The Fracture Risk Assessment score indicated a low risk of major osteoporotic and hip fractures, not warranting immediate treatment. Plans are underway to initiate enzyme replacement therapy with asfotase alfa.
CONCLUSION Recognizing HPP is crucial, as early diagnosis and treatment can significantly improve patient outcomes and prevent complications.
Collapse
Affiliation(s)
- Angad Singh Gill
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14221, United States
- Department of Research, American Society for Inclusion, Diversity, and Equity in Healthcare, Lewes, DE 19958, United States
| | - Pallavi Sharma
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14221, United States
- Department of Research, American Society for Inclusion, Diversity, and Equity in Healthcare, Lewes, DE 19958, United States
| | - Mahmoud Nassar
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14221, United States
- Department of Research, American Society for Inclusion, Diversity, and Equity in Healthcare, Lewes, DE 19958, United States
| | - Erlin Marte
- Department of Research, American Society for Inclusion, Diversity, and Equity in Healthcare, Lewes, DE 19958, United States
- Department of Endocrinology, Buffalo Veterans Affairs Medical Center, Buffalo, NY 14215, United States
| |
Collapse
|
31
|
Sakellariou E, Argyropoulou E, Galanis A, Rozis M, Zachariou D, Varsamos I, Parchas N, Kalavrytinos D, Karampinas P, Vasiliadis ES, Vlamis J, Pneumaticos S. Life-threatening vascular injury in an elderly patient with isolated pubic ramus fracture: A case report. World J Clin Cases 2025; 13:103786. [DOI: 10.12998/wjcc.v13.i21.103786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 02/20/2025] [Accepted: 03/24/2025] [Indexed: 04/27/2025] Open
Abstract
BACKGROUND Pubic ramus fractures are generally considered fragility fractures in the elderly population, commonly deriving from a low-impact fall. Treatment is ordinarily conservative and hemodynamic complications are exceedingly infrequent. Notwithstanding, patients with copious comorbidities should be carefully monitored for potential vascular injury.
CASE SUMMARY This case report presents the management of a 75-year-old male patient with a history of diabetes mellitus and arterial hypertension who was admitted to the emergency room with a superior pubic ramus fracture. The patient experienced a significant drop in hematocrit and hemoglobin levels post-admission, necessitating urgent intervention. A computed tomography angiography revealed active bleeding, leading to the embolization of the medial femoral branch. The patient was stabilized hemodynamically and was discharged after 15 days, with recommendations for home-based follow-up care.
CONCLUSION This report denotes the various challenges and strategies in managing simple fractures that are treated conservatively, but need prompt monitoring for occult vascular injuries that can be fatal.
Collapse
Affiliation(s)
- Evangelos Sakellariou
- 3rd Department of Orthopaedic Surgery, KAT General Hospital, National & Kapodistrian University of Athens, Athens 14561, Greece
| | - Evangelia Argyropoulou
- Department of Orthopaedics and Traumatology, University General Hospital of Patras, Patras 26504, Greece
| | - Athanasios Galanis
- 3rd Department of Orthopaedic Surgery, KAT General Hospital, National & Kapodistrian University of Athens, Athens 14561, Greece
| | - Meletis Rozis
- 3rd Department of Orthopaedic Surgery, KAT General Hospital, National & Kapodistrian University of Athens, Athens 14561, Greece
| | - Dimitrios Zachariou
- 3rd Department of Orthopaedic Surgery, KAT General Hospital, National & Kapodistrian University of Athens, Athens 14561, Greece
| | - Iordanis Varsamos
- 3rd Department of Orthopaedic Surgery, KAT General Hospital, National & Kapodistrian University of Athens, Athens 14561, Greece
| | - Nicolaos Parchas
- Department of Orthopaedics and Traumatology, University General Hospital of Patras, Patras 26504, Greece
| | | | - Panagiotis Karampinas
- 3rd Department of Orthopaedic Surgery, KAT General Hospital, National & Kapodistrian University of Athens, Athens 14561, Greece
| | - Elias S Vasiliadis
- 3rd Department of Orthopaedic Surgery, KAT General Hospital, National & Kapodistrian University of Athens, Athens 14561, Greece
| | - John Vlamis
- 3rd Department of Orthopaedic Surgery, KAT General Hospital, National & Kapodistrian University of Athens, Athens 14561, Greece
| | - Spiros Pneumaticos
- 3rd Department of Orthopaedic Surgery, KAT General Hospital, National & Kapodistrian University of Athens, Athens 14561, Greece
| |
Collapse
|
32
|
Zhang W, Chen Y, Wang Y, Zhou Y, Guo H, Xu J. TSH inhibits osteoclast differentiation through AMPK signaling pathway. Gene 2025; 955:149442. [PMID: 40157619 DOI: 10.1016/j.gene.2025.149442] [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: 01/08/2025] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
PURPOSE It is believed that osteoporosis (OP) is associated with hyperthyroidism as a result of the elevation in thyroxine levels. However, patients with subclinical hyperthyroidism, which is characterized by decreased levels of thyroid-stimulating hormone (TSH) alone, are at equal risk of osteoporosis. Research has shown that TSH receptor (TSHR) is expressed on osteoclasts, but whether TSH directly regulates osteoclasts and the underlying mechanisms remain unclear. METHODS In this study, we used osteoclast precursor cell conditional TSHR-knockout (TSHR CKO) mouse to study the effects of TSHR knockout on bone metabolism in mice and the changes in osteoclast differentiation in vitro. Transcriptomics was used to identify differentially expressed genes and signaling pathways. RESULTS In vitro, experiments confirmed that TSH inhibited osteoclast differentiation in mouse RAW264.7 monocyte/macrophage cell line and targeted the key signaling pathway AMPK by RNA-seq sequencing. We found TSHR CKO mice exhibited decreased femoral biomechanics and damaged bone microstructure. The serum levels of bone resorption marker were increased, accompanied by an increase in the number of osteoclasts. CONCLUSION TSH inhibits osteoclast differentiation by activating the AMPK signaling pathway, and exerts an osteoprotective effect. This study will provide guidance for the diagnosis and treatment of osteoporosis. TSH structural analogs or AMPK activators are expected to provide new ideas for the development of drugs to prevent and treat osteoporosis.
Collapse
Affiliation(s)
- Wenwen Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of stem cell and Gene Therapy for endocrine Metabolic diseases, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China
| | - Yu Chen
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of stem cell and Gene Therapy for endocrine Metabolic diseases, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China
| | - Yan Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of stem cell and Gene Therapy for endocrine Metabolic diseases, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China
| | - Yanman Zhou
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Honglin Guo
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of stem cell and Gene Therapy for endocrine Metabolic diseases, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Jin Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China; "Chuangxin China" Innovation Base of stem cell and Gene Therapy for endocrine Metabolic diseases, China; Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong 250021, China.
| |
Collapse
|
33
|
Yaseen AA, Tumey LN. Advances and challenges in immunosuppressive antibody drug conjugates. Eur J Med Chem 2025; 291:117576. [PMID: 40186891 DOI: 10.1016/j.ejmech.2025.117576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
Since the approval of Mylotarg™ in 2000 for acute myeloid leukemia, antibody-drug conjugates (ADCs) have significantly advanced precision medicine, particularly for oncology applications. ADCs combine an antibody, a linker, and a payload to result in a targeted therapeutic that minimizes toxicity resulting from systemic drug exposure. This review explores the innovative application of ADC technology towards immunosuppressive therapeutics, primarily focusing on antibody-mediated delivery of glucocorticoids (GCs). Despite their potent anti-inflammatory effects, the clinical use of GCs is limited by adverse systemic effects including osteoporosis, high blood sugar, adrenal insufficiency, weight gain, and glaucoma. Therefore, targeted delivery via ADCs presents a promising strategy to enhance therapeutic efficacy while reducing toxicity. Herein, we review the current status of immune-suppressing ADC technology, starting with early investigations of CD163-targeted dexamethasone and moving to the design of ADCs employing next-generation ultra-potent GCs. Additionally, we will discuss the current status of anti-inflammatory ADCs that employ non-glucocorticoid immune-suppressive medications. Throughout, we will highlight preclinical and clinical data that serves to derisk and drive investment in this new therapeutic class. In parallel, we will focus on ADC design principles that illustrate the importance of careful selection of payload, linker, and conjugation technology in this emerging field.
Collapse
Affiliation(s)
- Aiman A Yaseen
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, PO Box 6000, Binghamton, NY, 13902-6000, USA
| | - L Nathan Tumey
- Binghamton University, School of Pharmacy and Pharmaceutical Sciences, PO Box 6000, Binghamton, NY, 13902-6000, USA.
| |
Collapse
|
34
|
Liu G, Xue J, Zhou X, Gui M, Xia R, Zhang Y, Cai Y, Li S, Shi S, Mao X, Chen Z. The paradigm shifts of periodontal regeneration strategy: From reparative manipulation to developmental engineering. Bioact Mater 2025; 49:418-436. [PMID: 40165829 PMCID: PMC11957753 DOI: 10.1016/j.bioactmat.2025.03.009] [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: 11/02/2024] [Revised: 02/07/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025] Open
Abstract
Ideal periodontal regeneration requires the integration of alveolar bone, periodontal ligament, and cementum, along with Sharpey's fibers for occlusal force resistance. However, physiological regeneration remains rare due to its intricate structure, making clinical regeneration a challenge. Periodontal ligament stem cells (PDLSCs), first isolated in 2004, hold the key to multi-directional differentiation into cementoblasts, fibroblasts, and osteoblasts. While traditional therapies like guided tissue regeneration (GTR) aim to activate PDLSCs, clinical outcomes are inconsistent, suggesting the need for additional strategies to enhance PDLSCs' functions. Advancements in molecular biotechnology have introduced the use of recombinant growth factors for tissue regeneration. However, maintaining their efficacy requires high doses, posing cost and safety issues. Multi-layered scaffolds combined with cell sheet technology offer new insights, but face production, ethical, and survival challenges. Immune regulation plays a crucial role in PDLSC-mediated regeneration. The concept of "coagulo-immunomodulation" has emerged, emphasizing the coupling of blood coagulation and immune responses for periodontal regeneration. Despite its potential, the clinical translation of immune-based strategies remains elusive. The "developmental engineering" approach, which mimics developmental events using embryonic-stage cells and microenvironments, shows promise. Our research group has made initial strides, indicating its potential as a viable solution for periodontal complex regeneration. However, further clinical trials and considerations are needed for successful clinical application. This review aims to summarize the strategic transitions in the development of periodontal regenerative materials and to propose prospective avenues for future development.
Collapse
Affiliation(s)
- Guanqi Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Junlong Xue
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Xuan Zhou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Mixiao Gui
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Ruidi Xia
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Yanshu Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Yihua Cai
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Shuhua Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| | - Songtao Shi
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- South China Center of Craniofacial Stem Cell Research, Guangzhou, 510055, China
| | - Xueli Mao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- South China Center of Craniofacial Stem Cell Research, Guangzhou, 510055, China
| | - Zetao Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou, 510055, China
| |
Collapse
|
35
|
Chen H, Zhang R, Jin M, Yang J, Chen L, Xie Y. Advances in the mechanism and therapies of achondroplasia. Genes Dis 2025; 12:101436. [PMID: 40256430 PMCID: PMC12008630 DOI: 10.1016/j.gendis.2024.101436] [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: 12/10/2023] [Accepted: 04/24/2024] [Indexed: 04/22/2025] Open
Abstract
Achondroplasia (ACH), is the prevailing type of genetic dwarfism in humans, caused by mutations in fibroblast growth factor receptor 3 (FGFR3) that are inherited in an autosomal dominant manner. FGFR3 is mainly expressed in condensed mesenchyme, chondrocytes, and mature osteoblasts and osteoclasts, in which it regulates the formation, development, growth, and remodeling of the skeletal system. Mutations in FGFR3 causing ACH result in enhanced FGFR3 signaling through combined mechanisms including enhancing FGF dimerization and tyrosine kinase activity and stabilizing FGF receptors. In ACH, suppression of the proliferation and maturation of chondrocytes in the growth plate leads to a notable reduction in growth plate size, trabecular bone volume, and bone elongation through a profound enhancement of FGFR3 signaling. This review aims to comprehensively outline the cellular and molecular mechanisms contributing to the pathological process of ACH and its potential therapeutic interventions.
Collapse
Affiliation(s)
- Hangang Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | - Ruobin Zhang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Jing Yang
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| | - Yangli Xie
- Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma and Chemical Poisoning, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing 400042, China
| |
Collapse
|
36
|
Yao F, Bao Y, Meng Q, Chen Y, Zhao L, Wang P, Zhou B. Periprosthetic osteolysis: Mechanisms and potential treatment strategies. Cell Signal 2025; 131:111758. [PMID: 40132773 DOI: 10.1016/j.cellsig.2025.111758] [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: 12/11/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Periprosthetic osteolysis is a common bone-related disorder that often occurs after total hip arthroplasty. The implants can cause damage to bone and bone-related cells due to mechanical stress and micromotions, resulting in the generation of a large number of wear particles. These wear particles trigger inflammation and oxidative stress in the surrounding tissues, disrupting the delicate balance maintained by osteoblasts and osteoclasts, ultimately leading to bone loss around the implant. Clinical investigations have demonstrated that Epimedium prenylflavonoids, miR-19a-3p, stem cell-derived exosomes, and certain non-PPO category pharmaceuticals have regulatory effects on bone homeostasis through distinct molecular pathways. Notably, this phenomenon reflects inherent biological rationality rather than stochastic occurrence. Extensive research has revealed that multiple natural compounds, non-coding RNAs, exosomes, and non-PPO therapeutics not only exert modulatory influences on critical pathophysiological processes including inflammatory cascades, oxidative stress responses, and tissue regeneration mechanisms, but also effectively regulate bone-related cellular functions to inhibit PPO progression. Therefore, this review comprehensively and systematically summarizes the main pathogenic mechanisms of periprosthetic osteolysis. Furthermore, it delves deeper into the research progress on the applications of currently reported natural products, ncRNAs, exosomes, and non-PPO medications in the treatment of periprosthetic osteolysis. Based on this, we hope that this paper can provide new perspectives and references for the future development of drugs targeting periprosthetic osteolysis.
Collapse
Affiliation(s)
- Fang Yao
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yue Bao
- Department of Nursing, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Qian Meng
- Outpatient Department, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yanrong Chen
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Luxi Zhao
- Department of Anesthesiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Pingmei Wang
- Department of Orthopaedics, The People's Hospital of Shimen County, Shimen 415399, China
| | - Bin Zhou
- Department of Orthopaedics, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
37
|
Han X, Zhang C, Lei Q, Xu J, Zhou Y. Stiffness regulates extracellular matrix synthesis in fibroblasts by DDR1-TGF-β/STAT3 mechanotransduction axis. BIOMATERIALS ADVANCES 2025; 172:214240. [PMID: 40023083 DOI: 10.1016/j.bioadv.2025.214240] [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: 08/21/2024] [Revised: 02/10/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
For a long time, research on atherosclerosis (AS) has mainly focused on endothelial cells (ECs) and smooth muscle cells (SMCs) in blood vessels. Fibroblasts, however, being the major component in adventitia, little is known about their role. Fibroblasts are highly plastic cells, capable of undergoing phenotypic changes in response to various extracellular signals. Once activated, fibroblasts can promote fibrosis by altering the secretion of extracellular matrix (ECM). In this study, the effect of ECM stiffness on fibroblasts was investigated. Polyacrylamide (PA) gels with varying elastic moduli (1 kPa, 20 kPa and 100 kPa) were used as models for matrix stiffness. Human fibroblasts were cultured on these substrates, and their phenotypic and functional changes were examined. The data revealed that a collagen-binding receptor, Discoidin Domain Receptor 1 (DDR1), plays a central role in sensing mechanical stimuli from ECM. Matrix stiffness-induced phosphorylation of DDR1 suppresses the synthesis of ECM proteins in fibroblasts. The expression of ECM proteins on the 1 kPa substrate was significantly higher than that on the 20 kPa and 100 kPa substrates, while the phosphorylation level of DDR1 was notably reduced. After knocking out DDR1, the difference in ECM proteins expression among the three substrates with different stiffness levels disappeared. The signal transduction from DDR1 to ECM synthesis is mediated by the TGF-β/STAT3 signaling axis. Our study reveals how matrix stiffness regulates the synthesis of ECM in fibroblasts and paves the way for understanding the regulation of fibrotic process in the pathogenesis of AS.
Collapse
Affiliation(s)
- Xiaomei Han
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Chao Zhang
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Qian Lei
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Jin Xu
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China
| | - Yue Zhou
- Department of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, 1954 Huashan Road, Shanghai, PR China.
| |
Collapse
|
38
|
Jiang M, Li H, Zhang Q, Xu T, Huang L, Zhang J, Yu H, Zhang J. The role of RGS12 in tissue repair and human diseases. Genes Dis 2025; 12:101480. [PMID: 40271194 PMCID: PMC12017852 DOI: 10.1016/j.gendis.2024.101480] [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: 02/05/2024] [Revised: 08/05/2024] [Accepted: 11/02/2024] [Indexed: 04/25/2025] Open
Abstract
Regulator of G protein signaling 12 (RGS12) belongs to the superfamily of RGS proteins defined by a conserved RGS domain that canonically binds and deactivates heterotrimeric G-proteins. As the largest family member, RGS12 is widely expressed in many cells and tissues. In the past few decades, it has been found that RGS12 affects the activity of various cells in the human body, participates in many physiological and pathological processes, and plays an important role in the pathogenesis of many diseases. Here, we set out to comprehensively review the role of RGS12 in human diseases and its mechanisms, highlighting the possibility of RGS12 as a therapeutic target for the treatment of human diseases.
Collapse
Affiliation(s)
- Min Jiang
- Department of Geriatric Oncology and Department of Palliative Care, Chongqing University Cancer Hospital, Chongqing 400030, China
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hongmei Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Qiong Zhang
- Institute of Burn Research, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Tongtong Xu
- General Department of Critical Care Medicine, Zhenjiang Traditional Chinese Medicine Hospital, Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Zhenjiang, Jiangsu 212003, China
| | - Le Huang
- Army 72nd Group Military Hospital, Huzhou, Zhejiang 313000, China
| | - Jinghong Zhang
- Department of Plastic Surgery, State Key Laboratory of Trauma, Burns and Combined Injury, Southwest Hospital, The Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Huiqing Yu
- Department of Geriatric Oncology and Department of Palliative Care, Chongqing University Cancer Hospital, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Junhui Zhang
- Department of Geriatric Oncology and Department of Palliative Care, Chongqing University Cancer Hospital, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
39
|
Li G, Wei X, Lv K, Xie D, Liu M, Xu Y, Ma D, Jiao G. Cyclodextrin-based self-assembling hydrogel for Photothermal-controlled nitric oxide release in stage-specific treatment of MRSA-induced arthritis. Carbohydr Polym 2025; 359:123578. [PMID: 40306784 DOI: 10.1016/j.carbpol.2025.123578] [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: 01/13/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
MRSA-induced arthritis is a prevalent and highly debilitating orthopedic condition. The inflammatory response induced by bacterial infection hinders tissue repair and exacerbates bone loss. Traditional antibiotic therapies are limited by low bioavailability, substantial side effects, and narrow efficacy, rendering them inadequate for comprehensive treatment of arthritis. Nitric oxide (NO) has demonstrated considerable potential in overcoming bacterial resistance, modulating immune responses, and facilitating tissue repair. Therefore, a stage-specific NO release strategy, tailored to the distinct phases of bacterial arthritis, is essential for effective treatment. In this study, mesoporous polydopamine nanoparticles were utilized as NO donors (mPDA/NONOate) and encapsulated within a supramolecular hydrogel formed via the host-guest interaction between α-cyclodextrin (α-CD) and Pluronic F127. The injectable nature of the resulting NO/PDA-Gel hydrogel ensured uniform distribution within irregular bone joint infection sites, minimizing NO donor loss and enhancing local bioavailability. Notably, upon near-infrared (NIR) irradiation, the hydrogel induces a rapid increase in local temperature, facilitating rapid NO release. At the same time, the synergistic photothermal effect effectively kills bacteria and rapidly controls the infection. Without light irradiation, NO is sustainably and stably released from the NO/PDA-Gel, modulating the bone immune microenvironment, alleviating inflammation, promoting chondrocyte proliferation and differentiation, and accelerating bone tissue repair, thus significantly shortening the healing time of MRSA-induced arthritis. In conclusion, the injectable self-assembled NO/PDA-Gel offers a precise, stage-matched therapeutic approach for MRSA-induced arthritis and holds promise for the treatment of deep-seated infections caused by other multidrug-resistant pathogens.
Collapse
Affiliation(s)
- Guowei Li
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Xiaohua Wei
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Kai Lv
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Dongna Xie
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Mei Liu
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yi Xu
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Dong Ma
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Genlong Jiao
- Dongguan Key Laboratory of Central Nervous System Injury and Repair, Department of Orthopedic Surgery, The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan 523573, Guangdong, China.
| |
Collapse
|
40
|
Zhang J, Shen X, Wang Z, Yong J, Jiang Z, Yang G. Influences and strategies for bone regeneration based on microenvironment pH adjustment. Bone 2025; 196:117484. [PMID: 40194688 DOI: 10.1016/j.bone.2025.117484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/27/2025] [Accepted: 04/04/2025] [Indexed: 04/09/2025]
Abstract
Bone possesses remarkable endogenous regenerative capacity. Bone regeneration is typically divided into three stages: inflammation, bone formation, and bone remodeling, during which pH is a critical variable. The influence of pH on the bone regeneration process depends on three main factors: (1) the activity and differentiation of cells involved in bone regeneration are affected by pH; (2) protein activity is regulated by pH; and (3) extracellular calcium phosphate precipitates in a pH-dependent manner. The aim of this study is to review the mechanisms by which microenvironment pH affects bone regeneration and to explore specific sites and signaling pathways involved in pH regulation during the bone regeneration process. Therapeutic approaches aimed at enhancing bone regeneration via modulation of microenvironment pH are discussed, including pH adjustment via biological implant materials, pH-responsive material setting, and pH stabilization through anti-inflammatory therapy. Investigating the impact of microenvironment pH on bone regeneration is of considerable clinical importance, as it provides valuable insights for improving the success rates of bone implants and promoting bone healing. This review offers insights into regulatory mechanisms, establishes theoretical foundations, and presents new perspectives for current research on bone defect repair.
Collapse
Affiliation(s)
- Jing Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Xinyi Shen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhikang Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Jiawen Yong
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
41
|
Bellinge JW, Sim M, Francis RJ, Lee SC, Chan DC, Girgis CM, Watts GF, Lewis JR, Schultz CJ. The effect of oral colchicine and vitamin K1 on bone metabolism in patients with diabetes mellitus: A post-hoc analysis of a 2 × 2 factorial randomized controlled trial with 18F-sodium fluoride positron emission tomography. Bone 2025; 196:117492. [PMID: 40258481 DOI: 10.1016/j.bone.2025.117492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/25/2025] [Accepted: 04/16/2025] [Indexed: 04/23/2025]
Abstract
PURPOSE Diabetes mellitus (DM) confers an increased risk of fracture. Fracture risk stratification techniques are imperfect, and preventative therapies are sparse. We aimed to describe features associated with a dysfunctional bone metabolism determined by 18F-Sodium Fluoride Positron Emission Tomography (18F-NaF PET) in patients with DM and test the effects of vitamin K1 and colchicine therapy on vertebral 18F-NaF activity. METHODS This is a post-hoc analysis of a 2 × 2 factorial randomized double-blind placebo-controlled trial. Participants aged 50-80 with DM underwent 18F-NaF PET/CT imaging at baseline, 3 months of therapy with vitamin K1 (10mg/daily) or placebo, and colchicine (0.5 mg/day) or placebo and repeat 18F-NaF PET/CT. The 18F-NaF vertebral mean standardized uptake value (SUVmean) and the CT estimated bone mineral density (BMD) (in Hounsfield units) was evaluated from thoracic vertebra. RESULTS In total, 149 individuals (66.4 % male, mean age 65.5 ± 6.8 years) were included. Male sex (β -1.421, 95 % CI [-1.826, -1.016], p < 0.001), duration of DM in years (-0.021 [-0.039, -0.002], p = 0.030) and CT estimated vertebral BMD (0.011 [0.006, 0.015], p < 0.001) were independently associated with the SUVmean. The change in the SUVmean was similar between vitamin K1 or placebo groups (-0.07 ± 0.64 v 0.07 ± 0.69, p = 0.20). Participants receiving colchicine therapy had a greater reduction in the SUVmean, compared with placebo (-0.12 ± 0.72 v 0.11 ± 0.60, p = 0.039). CONCLUSION 18F-NaF PET may be a useful measure of vertebral bone metabolism in people with DM. Three months of oral colchicine reduced the 18F-NaF vertebral SUVmean, whereas Vitamin K1 had no effect. The findings should be considered hypothesis generating. TRIAL REGISTRATION www.anzctr.org.au (ACTRN12616000024448).
Collapse
Affiliation(s)
- Jamie W Bellinge
- Medical School, University of Western Australia, Perth, Western Australia, Australia; Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.
| | - Marc Sim
- Medical School, University of Western Australia, Perth, Western Australia, Australia; Institute for Nutrition Research, School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Roslyn J Francis
- Medical School, University of Western Australia, Perth, Western Australia, Australia; Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Sing Ching Lee
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Dick C Chan
- Medical School, University of Western Australia, Perth, Western Australia, Australia
| | - Christian M Girgis
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Gerald F Watts
- Medical School, University of Western Australia, Perth, Western Australia, Australia; Cardiometabolic service, Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Joshua R Lewis
- Medical School, University of Western Australia, Perth, Western Australia, Australia; Institute for Nutrition Research, School of Medical and Health Sciences, Edith Cowan University, Perth, Australia; Centre for Kidney Research, Children's Hospital Westmead, School of Public Health, University of Sydney, Westmead, New South Wales, Australia
| | - Carl J Schultz
- Medical School, University of Western Australia, Perth, Western Australia, Australia; Department of Cardiology, Royal Perth Hospital, Perth, Western Australia, Australia
| |
Collapse
|
42
|
Weng K, He Y, Weng X, Yuan Y. Exercise alleviates osteoporosis by regulating the secretion of the Senescent Associated Secretory Phenotype. Bone 2025; 196:117485. [PMID: 40216288 DOI: 10.1016/j.bone.2025.117485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/18/2025]
Abstract
As the elderly population grows, the number of patients with metabolic bone diseases such as osteoporosis has increased sharply, posing a significant threat to public health and social economics. Although pharmacological therapies for osteoporosis demonstrate therapeutic benefits, their prolonged use is associated with varying degrees of adverse effects. As a non-pharmacological intervention, exercise is widely recognized for its cost-effectiveness, safety, and lack of toxic side effects, making it a recommended treatment for osteoporosis prevention and management. Previous studies have demonstrated that exercise can improve metabolic bone diseases by modulating the Senescent Associated Secretory Phenotype (SASP). However, the mechanisms through which exercise influences SASP remain unclear. Therefore, this review aims to summarize the effects of exercise on SASP and elucidate the specific mechanisms by which exercise regulates SASP to alleviate osteoporosis, providing a theoretical basis for osteoporosis through exercise and developing targeted therapies.
Collapse
Affiliation(s)
- Kaihong Weng
- Graduate School, Guangzhou Sport University, 510500 Guangzhou, China
| | - Yuting He
- Graduate School, Guangzhou Sport University, 510500 Guangzhou, China
| | - Xiquan Weng
- School of Exercise and Health, Guangzhou Sport University, 510500 Guangzhou, China; Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, 510500 Guangzhou, China.
| | - Yu Yuan
- School of Exercise and Health, Guangzhou Sport University, 510500 Guangzhou, China; Guangdong Provincial Key Laboratory of Physical Activity and Health Promotion, Guangzhou Sport University, 510500 Guangzhou, China.
| |
Collapse
|
43
|
Ripamonti U. Global morphogenesis regulating tissue architecture and organogenesis. BIOMATERIALS ADVANCES 2025; 172:214262. [PMID: 40054230 DOI: 10.1016/j.bioadv.2025.214262] [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: 12/04/2024] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/17/2025]
Abstract
This perspective article proposes that the induction of bone by recombinant human bone morphogenetic proteins (hBMPs), and by the recombinant human transforming growth factor-β3 (hTGF-β3), the latter only in primates, recapitulates embryonic development, whereby large ossicles de novo form in heterotopic intramuscular sites, where several responding cells are available with marked vascular invasion. The induction of bone initiates with the induction of cartilage' anlages recapitulating development. Selected recombinant hBMPs, hBMP-2 and hOP-1 (hBMP-7) on the other hand, fail to induce significant osteoinduction in orthotopic intraskeletal sites in clinical contexts. This review proposes that the failure of significant clinical bone formation in orthotopic sites is because implantation of hBMP-2 and hOP-1, as well as hTGF-β3 in human mandibular sites, does not proceed via recapitulation of embryonic development, ultimately failing the clinical translation of the "bone induction principle". Biologically, a prerogative of the osteogenic proteins of the TGF-β super family, is the heterotopic induction of bone that initiates via recapitulation of embryonic bone development. Delivery of recombinant hBMP-2 and/or hOP-1 into human bony defects fails because the induction of bone is possible only via embryonic development, which does not occur in orthotopic sites, with limited responding cells and vascular supply. The initiation and assembly of the bone/bone-marrow organ follow a multistep molecular and cellular cascades that ultimately morphogenize the assembly of skeletogenesis masterminding ambulation, body erection, masticatory functions, copulation and the preservation of vital organs and tissues such as the marrow and the cerebral hemispheres. Embryologically, bone forms either via mesenchymal osteogenesis or via endochondral osteogenesis. The former route prevalently sculpts the craniomandibulofacial bones by inducing aggregation of targeted mesenchymal cells to condensate in the primordia of the craniofacial skeleton. A critical step is the condensation of mesenchymal cells that forms around vessels, as the vessels guide the formation of bone. Endochondral osteogenesis initiates by firstly constructing cartilage' anlages that developing bone uses as struts to growth and differentiate. The cartilage anlage is an extraordinary conduit that sets into motion molecular and cellular cell-to-cell, cell-to-receptors cross-talking to initiate angiogenesis and capillary invasion within the hypertrophic cartilage, chondrolysis that initiate osteoblastic cellular differentiation and the deposition of bone, osteoid synthesis, bone marrow development and the induction of a complete mineralized bone-bone marrow organ. The assembled constructs are storage of critical ions, several structural proteins such as osteonectins and fibronectins, biological markers of osteoblast' secretion and activity, such as osteocalcin, together with an array of extraordinary morphogen initiators that de novo set into motion the molecular and cellular cascades inducing bone in heterotopic sites recapitulating embryonic development. In evolutionary molecular biology contexts however, the pleiotropic activities of both proteins' family and the induction of bone formation in heterotopic sites are developmental, and thus not suitable to induce bone when recombinant morphogens are singly implanted in orthotopic skeletal defects, the latter lacking the developmental biological platform.
Collapse
Affiliation(s)
- Ugo Ripamonti
- The Department of internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
44
|
Zojaji M, Yang B, Collins CJ, Crenshaw TD, Ploeg HL. Accurate measurement of a bone surrogate flexural rigidity in three- and four-point bending. J Mech Behav Biomed Mater 2025; 167:106986. [PMID: 40179529 DOI: 10.1016/j.jmbbm.2025.106986] [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: 12/08/2023] [Revised: 02/07/2025] [Accepted: 03/27/2025] [Indexed: 04/05/2025]
Abstract
The mechanical assessment of long bones through bending is an established preclinical approach to evaluate the effectiveness of treatments for osteoporosis and fractures. Three- and four-point bending (3PB and 4PB) tests are the most common methods for mechanical characterization of long bones with Euler-Bernoulli (EB) theory to calculate of bone flexural rigidity (EI). Previous studies demonstrated that EB theory underestimates the EI of long bones due to its reliance on assumptions that are not entirely applicable to long bones. Therefore, the current study aimed to evaluate the factors that affect the percent error (PE) and bias stemming from the omission of contact and shear deflections in the EI estimation using mechanical testing and finite element analysis (FEA). The true EI of a porcine bone surrogate was used to quantify the percent error and bias of EI estimations from three deflection measurement methods and FEA, in 3PB and 4PB. The analysis confirmed that bending was the main component of total deflection, but only contributed to approximately 50 % and 65 % of the total deflection in 3PB and 4PB, respectively. The combined shear and indentation deflections accounted for the remainder of the total deflection. The FEA aligned with 3PB and 4PB tests with less than 10 % deviation. Underestimation of EI was largest with deflection measurements taken from the machine crosshead (PE 74 % in 3PB and 71 % in 4PB). However, these underestimations improved notably when indentation and shear deflections were considered (PE 42 % in 3PB and 39 % in 4PB). The deflection measurements from extensometer and digital image correlation (DIC) underestimated the EI by 66 % and 71 % in 3PB, and 57 % and 59 % in 4PB. When corrected for shear and indentation deflections, the 3PB PE reduced to 16 % and 14 %, respectively. In 4PB, PE reduced to, 10 % and 7 %, respectively, demonstrating the advantage of the 4PB test configuration over 3PB. The bias resulting from shear deflection was not consistent across deflection measurement methods; and therefore, cannot be generalized with a constant bias correction. The current study highlighted that a slight error in deflection measurement can lead to a significant inaccuracy in EI measurements. This sensitivity comes from the hyperbolic relationship between EI and deflection which not only depends on the ratio of support span to diameter of the specimen but also the test configuration and the ratio of the elastic to shear modulus of specimen. In other words, the PE from neglecting shear effects increases as the specimen EI increases. Accuracy with less than 10 % PE in EI estimations can be achieved by: 1. taking deflection measurements with extensometers, DIC, or FEA; 2. testing in 4PB instead of 3PB; and, 3. correcting for indentation and shear deflections.
Collapse
Affiliation(s)
- Mahsa Zojaji
- Queen's University, Mechanical and Materials Engineering, Kingston, ON, Canada.
| | - Baixuan Yang
- Queen's University, Mechanical and Materials Engineering, Kingston, ON, Canada
| | - Caitlyn J Collins
- Virginia Polytechnic Institute and State University, Department of Biomedical Engineering and Mechanics, VA, USA
| | - Thomas D Crenshaw
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Heidi-Lynn Ploeg
- Queen's University, Mechanical and Materials Engineering, Kingston, ON, Canada
| |
Collapse
|
45
|
Wang L, Jiang S, Zhou J, Gholipourmalekabadi M, Cao Y, Lin K, Zhuang Y, Yuan C. From hard tissues to beyond: Progress and challenges of strontium-containing biomaterials in regenerative medicine applications. Bioact Mater 2025; 49:85-120. [PMID: 40124596 PMCID: PMC11928986 DOI: 10.1016/j.bioactmat.2025.02.039] [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: 10/20/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Tissue engineering and regenerative medicine have emerged as crucial disciplines focused on the development of new tissues and organs to overcome the limitations of traditional treatments for tissue damage caused by accidents, diseases, or aging. Strontium ion (Sr2+) has garnered significant attention for its multifaceted role in promoting regeneration medicine and therapy, especially in bone tissue regeneration. Recently, numerous studies further confirm that Sr2+ also plays a critical in soft tissue regeneration. This review firstly summarizes the influence of Sr2+ on critical biological processes such as osteogenesis, angiogenesis, immune modulation, matrix synthesis, mineralization, and antioxidative defence mechanisms. Then details the classification, properties, advantages, and limitations of Sr-containing biomaterials (SrBMs). Additionally, this review extends to the current applications of SrBMs in regenerative medicine for diverse tissues, including bone, cartilage, skeletal muscle, dental pulp, cardiac tissue, skin, hair follicles, etc. Moreover, the review addresses the challenges associated with current SrBMs and provides insights for their future designing and applications in regenerative medicine.
Collapse
Affiliation(s)
- Liyun Wang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Shengjie Jiang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Jialiang Zhou
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Mazaher Gholipourmalekabadi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, 1449614535, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Yuan Cao
- Colorado College, 819 N Tejon Street Box 56, Colorado Springs, 80903, Colorado, USA
| | - Kaili Lin
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Yu Zhuang
- Department of Oral and Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Changyong Yuan
- School of Stomatology, Xuzhou Medical University, Affiliated Stomatological Hospital of Xuzhou Medical University, Xuzhou, 221004, China
| |
Collapse
|
46
|
Huang X, Yu J, Gou S, Qin H, Lu WW, Li Z, Tong L, Chen D. CRISPR/CasRx-mediated RNA knockdown targeting β-catenin and Ihh signaling alleviates osteoarthritis. Genes Dis 2025; 12:101468. [PMID: 40290123 PMCID: PMC12033902 DOI: 10.1016/j.gendis.2024.101468] [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: 07/07/2024] [Revised: 09/08/2024] [Accepted: 10/27/2024] [Indexed: 04/30/2025] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative joint disease. Currently, OA is incurable. Abnormal activation of canonical Wnt/β-catenin or Indian hedgehog (Ihh) signaling could lead to OA development and progression. This study aimed to determine if targeting β-catenin and Ihh signaling could yield an effective therapeutic intervention for OA disease. CRISPR/CasRx is a new RNA interference tool that can precisely and efficiently cleave single-strand RNAs. In this study, we screened CRISPR-derived RNA (crRNA) targeting Ctnnb1 and Smo in vitro and selected two optimal crRNAs for each gene. CasRx-mediated Ctnnb1 and Smo knockdown showed high efficiency and specificity with no obvious off-target effects in vitro. We then performed intra-articular injection of selected crRNAs driven by the adeno-associated virus into an OA mouse model. Micro-CT, histological, and histomorphometric analyses were conducted to evaluate the efficacy of CasRx approach on OA treatment. We found that the knockdown of Ctnnb1 and Smo decelerated pathological damage in the keen joint of the experimental OA mouse model. Our findings suggest that CasRx-mediated Ctnnb1 and Smo knockdown could be a potential strategy for OA treatment.
Collapse
Affiliation(s)
- Xingyun Huang
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
| | - Jiamin Yu
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
- University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100049, China
| | - Shixue Gou
- Guangzhou National Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong 510005, China
| | - Hongyu Qin
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
- Division of Spine Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - William W. Lu
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
| | - Zhen Li
- AO Research Institute Davos, Davos 7270, Switzerland
| | - Liping Tong
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
| | - Di Chen
- Research Center for Computer-aided Drug Discovery, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong 518055, China
- Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong 518055, China
| |
Collapse
|
47
|
Ugarteburu M, Doube M, Witek L, Rau C, Cardoso L, Richter CP, Carriero A. Small and porous ossicles, with flat stapes footplate and incudal fractures in the oim mouse model of osteogenesis imperfecta. Bone 2025; 196:117495. [PMID: 40280254 DOI: 10.1016/j.bone.2025.117495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/14/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Hearing loss affects approximately 70% of individuals with osteogenesis imperfecta (OI), a genetic connective tissue disorder characterized by bone fragility and deformities. No effective treatments exist for OI hearing loss, and its etiology is unknown limiting the development of new targeted therapies. This work investigates the impact of OI type I collagen mutations on the ossicle bone properties in the homozygous oim mouse model of severe OI, which is known to exhibit hearing loss. The morphology and porosity of the ossicles of 14-week-old oim and wild-type mice were analyzed using high-resolution synchrotron radiation microtomography. Additionally, the collagen fibers structure, bone tissue composition and mechanical properties were evaluated through second harmonic generation microscopy, Raman spectroscopy, and nanoindentation. The results demonstrated that oim ossicles are small, highly porous with an elevated lacunar number density, a flat stapes footplate and a small malleal processus brevis. One-in-two oim ossicles had incudomalleal joint abnormalities, exhibiting either a localized fracture in the incus head or a joint space widening. No differences were observed in collagen fibers structure, bone tissue composition and mechanical properties. These findings suggest that bone fractures observed in the oim incudes may contribute to their reported hearing loss. However, the underlying mechanism for these fractures' development remains to be investigated, as they do not appear to result from changes in bone tissue properties (collagen fibers organization, tissue composition or mechanical properties). Instead, they may be associated with joint space widening, and possibly altered ossicle chain kinematics.
Collapse
Affiliation(s)
- Maialen Ugarteburu
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Michael Doube
- Department of Infectious Diseases and Public Health, City University of Hong Kong, Hong Kong, China
| | - Lukasz Witek
- Biomaterials and Regenerative Biology Division, NYU College of Dentistry, New York, NY, USA; Hansjörg Wyss Department of Plastic Surgery, NYU Grossman School of Medicine, New York, NY, USA; Department of Biomedical Engineering, New York University, New York, NY, USA
| | - Christoph Rau
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK
| | - Luis Cardoso
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - Claus-Peter Richter
- Department of Otolaryngology, Northwestern University, Chicago, IL, USA; Department of Biomedical Engineering, Northwestern University, Evanston, IL, USA; Department of Communication Sciences and Disorders, Northwestern University, Evanston, IL, USA; The Hugh Knowles Center, Northwestern University, Evanston, IL, USA
| | - Alessandra Carriero
- Department of Biomedical Engineering, The City College of New York, New York, NY, USA.
| |
Collapse
|
48
|
Horasan M, Verner KA, Main RP, Nauman EA. Characterization of strains induced by in vivo locomotion and axial tibiotarsal loading in a chukar partridge model. Bone 2025; 196:117497. [PMID: 40280253 DOI: 10.1016/j.bone.2025.117497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Rodent models have offered valuable insights into the mechanobiological mechanisms that regulate bone adaptation responses to dynamic mechanical stimuli. However, using avian models may provide new insights into the mechanisms of bone adaptation to dynamic loads, as bird bones have distinct features that differ from mammalian bones. This paper illuminates these aspects by evaluating the mechanical environment in a novel avian, chukar partridge tibiotarsus (TBT), during fast locomotion and in cortical and cancellous tissue under in vivo dynamic compressive loading within the TBT. We measured in vivo mechanical strains at the TBT midshaft on the anterior, medial, and posterior surfaces during locomotion at various treadmill speeds. The mean in vivo strains measured on the anterior, medial, and posterior surfaces of the TBT midshaft were 154 με, -397 με, and -438 με, respectively, at a treadmill speed of 2 m/s. The mean experimentally measured strains on the anterior, medial, and posterior surfaces of the TBT were 114.7 με, -952.6 με, and -593.7 με under an in vivo dynamic compressive load of 130 N. The study, which employs a micro-computed tomography (microCT) based finite element model in combination with diaphyseal strain gauge measures, found that cancellous strains were greater than those in the midshaft cortical bone. Sensitivity analyses revealed that the material property of cortical bone was the most significant model parameter. In the midshaft cortical volume of interest (VOI), daily dynamic loading increased the maximum moment of inertia and reduced the bone area in the loaded limb compared to the contralateral control limb after three weeks of loading. Despite the strong correlations between the computationally modeled strains and experimentally measured strains at the medial and posterior gauge sites, no correlations existed between the computationally modeled strains and strain gradients, and histologically measured bone formation thickness at the mid-diaphyseal cross-section of the TBT.
Collapse
Affiliation(s)
- Murat Horasan
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA; Department of Mechanical Engineering, Aksaray University, Aksaray, Turkey.
| | - Kari A Verner
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Explico Engineering, Denver, CO, USA.
| | - Russell P Main
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, USA
| | - Eric A Nauman
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, USA; Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
49
|
Sharma A, Ojha S, Shelke A, Habib A. Scanning acoustic microscopy for biomechanical characterization of reindeer antler using singular spectral analysis. Bone 2025; 196:117475. [PMID: 40209972 DOI: 10.1016/j.bone.2025.117475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/23/2025] [Accepted: 03/27/2025] [Indexed: 04/12/2025]
Abstract
Scanning Acoustic Microscopy (SAM) has become a vital tool in materials science and biology, allowing for non-destructive and non-invasive analysis of biological specimens and bio-inspired materials. Its deep-penetrating imaging capabilities enable a broad range of applications. This study combines SAM with Singular Spectral Analysis (SSA) to enhance signal processing and extract key data, particularly acoustic impedance. Reindeer antlers, known for their rapid growth and unique mechanical properties, were chosen as a focus for this method. SAM was used to quantify the specific acoustic impedance, longitudinal stiffness, bulk modulus, and Young's modulus of the material at three orientations (0°, 45°, and 90°). This analysis provides a comprehensive understanding of the directional dependence of its structural behavior, highlighting its orthotropic nature. By analyzing cross-sections along three axes, this study reveals the orthotropic biomechanical properties of reindeer antlers, offering a systematic approach to characterizing biological materials. Their unique strength, resilience, and rapid growth highlight their potential as a sustainable and innovative biomaterial for bioengineering and advanced composites.
Collapse
Affiliation(s)
- Adarsh Sharma
- Department of Physics, Indian Institute of Technology Guwahati, India
| | - Shivam Ojha
- Department of Civil Engineering, Indian Institute of Technology Guwahati, 781039 Guwahati, Assam, India
| | - Amit Shelke
- Department of Civil Engineering, Indian Institute of Technology Guwahati, 781039 Guwahati, Assam, India
| | - Anowarul Habib
- Department of Physics and Technology, UiT The Arctic University of Norway, 9037 Tromsø, Norway.
| |
Collapse
|
50
|
Furrer R, Handschin C. Biomarkers of aging: from molecules and surrogates to physiology and function. Physiol Rev 2025; 105:1609-1694. [PMID: 40111763 DOI: 10.1152/physrev.00045.2024] [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: 10/30/2024] [Revised: 01/10/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Many countries face an unprecedented challenge in aging demographics. This has led to an exponential growth in research on aging, which, coupled to a massive financial influx of funding in the private and public sectors, has resulted in seminal insights into the underpinnings of this biological process. However, critical validation in humans has been hampered by the limited translatability of results obtained in model organisms, additionally confined by the need for extremely time-consuming clinical studies in the ostensible absence of robust biomarkers that would allow monitoring in shorter time frames. In the future, molecular parameters might hold great promise in this regard. In contrast, biomarkers centered on function, resilience, and frailty are available at the present time, with proven predictive value for morbidity and mortality. In this review, the current knowledge of molecular and physiological aspects of human aging, potential antiaging strategies, and the basis, evidence, and potential application of physiological biomarkers in human aging are discussed.
Collapse
|