1.Aging and Regeneration of Hypothalamic Neural Stem Cells
Progress in Biochemistry and Biophysics 2025;52(5):1126-1139
Hypothalamic neural stem cells (htNSCs) are a type of glial-like neural stem cell located in the hypothalamus, possessing unique biological characteristics. They not only have the capacity to proliferate and differentiate but can also migrate into the parenchymal regions of the hypothalamus, further developing into neurons and successfully integrating into neural circuits. HtNSCs play multiple key physiological roles in the adult hypothalamus, including contributing to the formation of the blood-hypothalamic barrier (BHB), which is crucial for maintaining the stability of the hypothalamic environment. Through the BHB, htNSCs facilitate the effective diffusion of small molecules between the blood, cerebrospinal fluid, and hypothalamic parenchyma, thereby ensuring the proper transmission of nutrients and signaling molecules. In addition, htNSCs can sense fluctuations in blood glucose levels and regulate the release of neuropeptides accordingly, thus influencing the body’s energy metabolism and endocrine balance. However, as the body ages, the function of htNSCs gradually declines. Studies have shown that the aging of htNSCs has significant adverse effects on energy metabolism, sex hormone secretion, and overall hypothalamic function. During the aging process, the proliferative and differentiative capacities of htNSCs diminish, leading to reduced neuronal replenishment and subsequently impairing the hypothalamus’s ability to regulate energy balance. Furthermore, aging htNSCs may secrete inflammatory factors that disrupt the endocrine functions of the hypothalamus, thereby affecting sex hormone secretion. This impact extends beyond the hypothalamus itself and may exert widespread effects on the entire endocrine system through pathways such as the hypothalamic-pituitary-gonadal axis. Fortunately, research has found that transplanting young htNSCs can effectively alleviate neurological and skeletal muscle dysfunction associated with aging. This transplantation therapy replenishes active htNSCs, restoring normal hypothalamic function and thereby improving the body’s energy metabolism and neuromuscular function. These findings offer new perspectives and potential therapeutic strategies for anti-aging interventions. In recent years, the role of htNSCs in regulating energy metabolism and promoting aging has attracted significant attention from researchers. Studies have shown that the aging of htNSCs is closely linked to the development of various diseases. For instance, in obesity and metabolic syndrome, htNSC dysfunction may lead to disturbances in energy metabolism. Moreover, the aging of htNSCs has also been associated with the onset of neurodegenerative diseases. Therefore, in-depth research into the mechanisms underlying htNSC aging is crucial for understanding the pathogenesis of these conditions. This article briefly reviews the classification of htNSCs, the impacts of their aging on bodily functions, their relationship with related diseases, and the regulatory mechanisms that promote htNSC regeneration. Some strategies aimed at promoting htNSC regeneration and counteracting their aging appear to influence the overall aging phenotype of organisms. For example, studies have shown that modulating specific signaling pathways or gene expression can promote htNSC regeneration, thereby delaying the aging process. Additionally, certain natural products or pharmacological agents may also influence htNSC aging. Further research on htNSC aging will enhance our understanding of the hypothalamus’s role in systemic aging and elucidate the reasons behind gender differences in aging patterns. Moreover, these studies may offer novel approaches and therapeutic targets for improving energy metabolism disorders and treating diseases associated with gonadal hormone abnormalities. In summary, htNSCs play a vital role in the physiological functions of the hypothalamus and the aging process. Further investigation into the mechanisms and regulatory pathways of htNSC aging will aid in the development of new anti-aging therapies and provide innovative strategies for the treatment of related diseases.
2.Cloning, subcellular localization and expression analysis of SmIAA7 gene from Salvia miltiorrhiza
Yu-ying HUANG ; Ying CHEN ; Bao-wei WANG ; Fan-yuan GUAN ; Yu-yan ZHENG ; Jing FAN ; Jin-ling WANG ; Xiu-hua HU ; Xiao-hui WANG
Acta Pharmaceutica Sinica 2025;60(2):514-525
The auxin/indole-3-acetic acid (Aux/IAA) gene family is an important regulator for plant growth hormone signaling, involved in plant growth, development, as well as response to environmental stresses. In the present study, we identified
3.Small Intestine Lipid Absorption and Health: The Improvement Effect of Exercise Under The Challenge of High-fat Diet
Wei-Huan WANG ; Yu-Xi DAI ; Yu-Xiu HE
Progress in Biochemistry and Biophysics 2025;52(6):1560-1573
The two core causes of obesity in modern lifestyle are high-fat diet (HFD) and insufficient physical activity. HFD can lead to disruption of gut microbiota and abnormal lipid metabolism, further exacerbating the process of obesity. The small intestine, as the “first checkpoint” for the digestion and absorption of dietary lipids into the body, plays a pivotal role in lipid metabolism. The small intestine is involved in the digestion, absorption, transport, and synthesis of dietary lipids. The absorption of lipids in the small intestine is a crucial step, as overactive absorption leads to a large amount of lipids entering the bloodstream, which affects the occurrence of obesity. HFD can lead to insulin resistance, disruption of gut microbiota, and inflammatory response in the body, which can further induce lipid absorption and metabolism disorders in the small intestine, thereby promoting the occurrence of chronic metabolic diseases such as obesity. Long term HFD can accelerate pathological structural remodeling and lipid absorption dysfunction of the small intestine: after high-fat diet, the small intestine becomes longer and heavier, with excessive villi elongation and microvilli elongation, thereby increasing the surface area of lipid absorption and causing lipid overload in the small intestine. In addition, overexpression of small intestine uptake transporters, intestinal mucosal damage induced “intestinal leakage”, dysbiosis of intestinal microbiota, ultimately leading to abnormal lipid absorption and chronic inflammation, accelerating lipid accumulation and obesity. Exercise, as one of the important means of simple, economical, and effective proactive health interventions, has always been highly regarded for its role in improving lipid metabolism homeostasis. The effect of exercise on small intestine lipid absorption shows a dose-dependent effect. Moderate to low-intensity aerobic exercise can improve the intestinal microenvironment, regulate the structure and lipid absorption function of the small intestine, promote lipid metabolism and health, while vigorous exercise, excessive exercise, and long-term high-intensity training can cause intestinal discomfort, leading to the destruction of intestinal structure and related symptoms, affecting lipid absorption. Long term regular exercise can regulate the diversity of intestinal microbiota, inhibit inflammatory signal transduction such as NF-κB, enhance intestinal mucosal barrier function, and improve intestinal lipid metabolism disorders, further enhancing the process of small intestinal lipid absorption. Exercise also participates in the remodeling process of small intestinal epithelial cells, regulating epithelial structural homeostasis by activating cell proliferation related pathways such as Wnt/β-catenin. Exercise can regulate the expression of lipid transport proteins CD36, FATP, and NPC1L1, and regulate the function of small intestine lipid absorption. However, the research on the effects of long-term exercise on small intestine structure, villus structure, absorption surface area, and lipid absorption related proteins is not systematic enough, the results are inconsistent, and the relevant mechanisms are not clear. In the future, experimental research can be conducted on the dose-response relationship of different intensities and forms of exercise, exploring the mechanisms of exercise improving small intestine lipid absorption and providing theoretical reference for scientific weight loss. It should be noted that the intestine is an organ that is sensitive to exercise response. How to determine the appropriate range, threshold, and form of exercise intensity to ensure beneficial regulation of intestinal lipid metabolism induced by exercise should become an important research direction in the future.
4.Exercise Improves Metaflammation: The Potential Regulatory Role of BDNF
Yu-Xi DAI ; Wei-Huan WANG ; Yu-Xiu HE
Progress in Biochemistry and Biophysics 2025;52(9):2314-2331
Metaflammation is a crucial mechanism in the onset and advancement of metabolic disorders, primarily defined by the activation of immune cells and increased concentrations of pro-inflammatory substances. The function of brain-derived neurotrophic factor (BDNF) in modulating immune and metabolic processes has garnered heightened interest, as BDNF suppresses glial cell activation and orchestrates inflammatory responses in the central nervous system via its receptor tyrosine kinase receptor B (TrkB), while also diminishing local inflammation in peripheral tissues by influencing macrophage polarization. Exercise, as a non-pharmacological intervention, is extensively employed to enhance metabolic disorders. A crucial mechanism underlying its efficacy is the significant induction of BDNF expression in central (hypothalamus, hippocampus, prefrontal cortex, and brainstem) and peripheral (liver, adipose tissue, intestines, and skeletal muscle) tissues and organs. This induction subsequently regulates inflammatory responses, ameliorates metabolic conditions, and decelerates disease progression. Consequently, BDNF is considered a pivotal molecule in the motor-metabolic regulation axis. Despite prior suggestions that BDNF may have a role in the regulation of exercise-induced inflammation, systematic data remains inadequate. Since that time, the field continues to lack structured descriptions and conversations pertinent to it. As exercise physiology research has advanced, the academic community has increasingly recognized that exercise is a multifaceted activity regulated by various systems, with its effects contingent upon the interplay of elements such as type, intensity, and frequency of exercise. Consequently, it is imperative to transcend the prior study paradigm that concentrated solely on localized effects and singular mechanisms and transition towards a comprehensive understanding of the systemic advantages of exercise. A multitude of investigations has validated that exercise confers health advantages for individuals with metabolic disorders, encompassing youngsters, adolescents, middle-aged individuals, and older persons, and typically enhances health via BDNF secretion. However, exercise is a double-edged sword; the relationship between exercise and health is not linearly positive. Insufficient exercise is ineffective, while excessive exercise can be detrimental to health. Consequently, it is crucial to scientifically develop exercise prescriptions, define appropriate exercise loads, and optimize health benefits to regulate bodily metabolism. BDNF mitigates metaflammation via many pathways during exercise. Initially, BDNF suppresses pro-inflammatory factors and facilitates the production of anti-inflammatory factors by modulating bidirectional transmission between neural and immune cells, therefore diminishing the inflammatory response. Secondly, exercise stimulates the PI3K/Akt, AMPK, and other signaling pathways via BDNF, enhancing insulin sensitivity, reducing lipotoxicity, and fostering mitochondrial production, so further optimizing the body’s metabolic condition. Moreover, exercise-induced BDNF contributes to the attenuation of systemic inflammation by collaborating with several organs, enhancing hepatic antioxidant capacity, regulating immunological response, and optimizing “gut-brain” axis functionality. These processes underscore the efficacy of exercise as a non-pharmacological intervention for enhancing anti-inflammatory and metabolic health. Despite substantial experimental evidence demonstrating the efficacy of exercise in mitigating inflammation and enhancing BDNF levels, numerous limitations persist in the existing studies. Primarily, the majority of studies have concentrated on molecular biology and lack causal experimental evidence that explicitly confirms BDNF as a crucial mediator in the exercise regulation of metaflammation. Furthermore, the outcomes of current molecular investigations are inadequately applicable to clinical practice, and a definitive pathway of “exercise-BDNF-metaflammation” remains unestablished. Moreover, the existing research methodology, reliant on animal models or limited human subject samples, constrains the broad dissemination of the findings. Future research should progressively transition from investigating isolated and localized pathways to a comprehensive multilevel and multidimensional framework that incorporates systems biology and exercise physiology. Practically, there is an immediate necessity to undertake extensive, double-blind, randomized controlled longitudinal human studies utilizing multi-omics technologies (e.g., transcriptomics, proteomics, and metabolomics) to investigate the principal signaling pathways of BDNF-mediated metaflammation and to elucidate the causal relationships and molecular mechanisms involved. Establishing a more comprehensive scientific evidence system aims to furnish a robust theoretical framework and practical guidance for the mechanistic interpretation, clinical application, and pharmaceutical development of exercise in the prevention and treatment of metabolic diseases.
5.Diagnostic Techniques and Risk Prediction for Cardiovascular-kidney-metabolic (CKM) Syndrome
Song HOU ; Lin-Shan ZHANG ; Xiu-Qin HONG ; Chi ZHANG ; Ying LIU ; Cai-Li ZHANG ; Yan ZHU ; Hai-Jun LIN ; Fu ZHANG ; Yu-Xiang YANG
Progress in Biochemistry and Biophysics 2025;52(10):2585-2601
Cardiovascular disease (CVD), chronic kidney disease (CKD), and metabolic disorders are the 3 major chronic diseases threatening human health, which are closely related and often coexist, significantly increasing the difficulty of disease management. In response, the American Heart Association (AHA) proposed a novel disease concept of “cardiovascular-kidney-metabolic (CKM) syndrome” in October 2023, which has triggered widespread concern about the co-treatment of heart and kidney diseases and the prevention and treatment of metabolic disorders around the world. This review posits that effectively managing CKM syndrome requires a new and multidimensional paradigm for diagnosis and risk prediction that integrates biological insights, advanced technology and social determinants of health (SDoH). We argue that the core pathological driver is a “metabolic toxic environment”, fueled by adipose tissue dysfunction and characterized by a vicious cycle of systemic inflammation and oxidative stress, which forms a common pathway to multi-organ injury. The at-risk population is defined not only by biological characteristics but also significantly impacted by adverse SDoH, which can elevate the risk of advanced CKM by a factor of 1.18 to 3.50, underscoring the critical need for equity in screening and care strategies. This review systematically charts the progression of diagnostic technologies. In diagnostics, we highlight a crucial shift from single-marker assessments to comprehensive multi-marker panels. The synergistic application of traditional biomarkers like NT-proBNP (reflecting cardiac stress) and UACR (indicating kidney damage) with emerging indicators such as systemic immune-inflammation index (SII) and Klotho protein facilitates a holistic evaluation of multi-organ health. Furthermore, this paper explores the pivotal role of non-invasive monitoring technologies in detecting subclinical disease. Techniques like multi-wavelength photoplethysmography (PPG) and impedance cardiography (ICG) provide a real-time window into microcirculatory and hemodynamic status, enabling the identification of early, often asymptomatic, functional abnormalities that precede overt organ failure. In imaging, progress is marked by a move towards precise, quantitative evaluation, exemplified by artificial intelligence-powered quantitative computed tomography (AI-QCT). By integrating AI-QCT with clinical risk factors, the predictive accuracy for cardiovascular events within 6 months significantly improves, with the area under the curve (AUC) increasing from 0.637 to 0.688, demonstrating its potential for reclassifying risk in CKM stage 3. In the domain of risk prediction, we trace the evolution from traditional statistical tools to next-generation models. The new PREVENT equation represents a major advancement by incorporating key kidney function markers (eGFR, UACR), which can enhance the detection rate of CKD in primary care by 20%-30%. However, we contend that the future lies in dynamic, machine learning-based models. Algorithms such as XGBoost have achieved an AUC of 0.82 for predicting 365-day cardiovascular events, while deep learning models like KFDeep have demonstrated exceptional performance in predicting kidney failure risk with an AUC of 0.946. Unlike static calculators, these AI-driven tools can process complex, multimodal data and continuously update risk profiles, paving the way for truly personalized and proactive medicine. In conclusion, this review advocates for a paradigm shift toward a holistic and technologically advanced framework for CKM management. Future efforts must focus on the deep integration of multimodal data, the development of novel AI-driven biomarkers, the implementation of refined SDoH-informed interventions, and the promotion of interdisciplinary collaboration to construct an efficient, equitable, and effective system for CKM screening and intervention.
6.Analysis of factors influencing clinical outcomes in the first frozen-thawed embryo transfer cycles
Kaixuan SUN ; Yinling XIU ; Yinghua WANG ; Yitong ZHANG ; Xiaoli LU ; Jing ZHOU ; Yuexin YU
Journal of China Medical University 2024;53(9):793-797
Objective To analyze the influencing factors of clinical pregnancy and live birth rates in patients undergoing frozen-thawed embryo transfer(FET)for the first time.Methods The clinical data of 1 458 patients who underwent FET cycle-assisted pregnancy for the first time were retrospectively analyzed and divided into four groups according to clinical pregnancy and live bith outcomes.The clini-cal data were compared to analyze the factors affecting clinical pregnancy and live birth rates in FET cycles that were included in multiple logistic regression analysis.Results Of the 1458 cycles,the clinical pregnancy and live birth rates were 44.0% and 34.0%,respectively.The mean age of the clinical pregnancy and live birth groups was lower than that in non-clinical pregnancy and stillbirth groups(P<0.05).The clinical pregnancy and live birth rates of patients aged<35 years were higher than those aged≥35 years(P<0.05).The clinical preg-nancy and live birth rates of patients with≥8 mm endometrial thickness were higher than those with<8 mm endometrial thickness(P<0.05).The clinical pregnancy rate of natural cycles of endometrial preparation regimen was higher than that of HRT cycles(P<0.05).The clinical pregnancy and live birth rates of double-embryo transfers were higher than that of single-embryo transfers(P<0.05).The clinical pregnancy and live birth rates of blastocyst transfers were higher than those of cleavage stage(P<0.05).Conclusion Age,endometrial thickness,number of transplanted embryos,and embryo morphology were the independent factors influencing clinical pregnancy and live birth outcomes during FET cycle transplantation.
7.miR-23a targets the XIAP-caspase-3 signaling pathway to contribute to the effects of resveratrol on ovarian function in mice
Yinling XIU ; Yuexin YU ; Kaixuan SUN ; Jing ZHOU ; Panpan ZHAO ; Jinlong XU
Journal of China Medical University 2024;53(11):1031-1035
Objective To investigate the effect of resveratrol(RES)on ovarian function in mice and elucidate the potential mechanism involving miR-23a.Methods Thirty mice were randomly divided into control,premature ovarian failure(POF)model,and treatment(RES)groups,with n=10 per group.The body weight and ovarian mass of the mice were measured,and the ovarian index was calculated.Hematoxylin and eosin staining was performed to observe pathological changes in mouse ovarian tissue.Real-time quantitative PCR and Western blotting were performed to measure the mRNA and protein levels of miR-23a,X-linked inhibitor of apoptosis protein(XIAP),and caspase-3 in the ovarian tissue.Results Compared with the control group,the POF group exhibited significant decreases in ovarian mass(P<0.05),ovarian index(P<0.05),number of primary follicles,and XIAP mRNA expression(P<0.05),alongside significant increases in miR-23a and caspase-3 mRNA expression(P<0.05).Compared with the POF group,the RES group exhibited significant increases in the ovarian mass(P<0.05),ovarian index(P<0.05),number of primary follicles,and XIAP mRNA expression(P<0.05),as well as significant decreases in miR-23a and caspase-3 mRNA expression(P<0.05).XIAP protein expression was significantly lower and caspase-3 protein expression was significantly higher in the POF group than in the control group(P<0.05).Conversely,XIAP protein expression was significantly higher and caspase-3 protein expression was significantly lower in the RES group than in the POF group(P<0.05).Conclusion RES exerts a protective effect on weakened ovarian function in mice,potentially mediated through its effect on miR-23a targeting the XIAP-caspase-3 signaling pathway.
8.Effects of total flavone extract from Ampelopsis megalophylla mediated by autophagy inhibitor 3-MA on proliferation and apoptosis of human breast cancer cells
Shi-Yi XU ; Si-Yu LIAO ; Tian-Xu ZHANG ; Xue ZOU ; Chun GUI ; Xiu-Qiao ZHANG
Chinese Pharmacological Bulletin 2024;40(6):1115-1123
Aim To explore the effect of total flavonoid extract(TFE)of Ampelopsis megalophylla on the pro-liferation and apoptosis of human breast cancer cells and its mechanism in autophagy inhibition.Methods For human cervical cancer cell Hela,human lung cancer cell A549,human liver cancer cell SMMC-7721,human breast cancer cell MCF-7,MDA-MB-231 and human normal liver cell L-02,MTT method was used to select sensitive cell lines.The inhibitory effect of TFE combined with autophagy inhibitor 3-methylade-nine(3-MA)on sensitive cell proliferation was detec-ted using MTT assay.The morphological changes of cells were observed using transmission electron micros-copy and Hoechst 33258 single staining method.The changes in cell apoptosis rate were detected using An-nexin V-FITC/PI dual staining method.The expression levels of apoptosis related proteins and pathway pro-teins(death receptor pathway,mitochondrial pathway,endoplasmic reticulum stress pathway)were detected uisng Western blot.The expression of the key protein Cyt-c in mitochondrial pathway was determiend by im-munofluorescence,and the autophagy agonist rapamy-cin was selected for reverse validation.Results TFE could inhibit the proliferation of human breast cancer cells in a concentration-dependent manner,and MCF-7 cells were sensitive cell lines.Compared with the TFE group,the TFE+3-MA group significantly increased the inhibition rate of MCF-7 cells at 24,48,and 72 h(P<0.01).The number of cells decreased,the gap increased,the number of apoptotic bodies increased,and the apoptosis rate increased(P<0.01).The ex-pression levels of Bax/Bcl-2(P<0.01),cleaved-caspase3(P<0.01),Cyt-c(P<0.05),FADD,and cleaved-caspase-12 all increased,and the expres-sion of apoptotic protein Cyt-c in nucleus increased.The fluorescence of the TFE+RA group decreased,re-versing the mitochondrial pathway apoptosis induced by TFE.Conclusions TFE can significantly inhibit the proliferation of human breast cancer cells.When inhib-iting autophagy,it may promote the apoptosis of MCF-7 cells through the mitochondrial pathway,and activa-ting autophagy can reverse apoptosis.
9.Effects of astragalus angelica on apoptosis and expression of Bax and caspase-3/9 in rabbit chondrocytes after fresh osteochondral allograft
Wan-Tao DONG ; Pan YANG ; Xiu-Juan YANG ; Shi-Ming QIU ; Peng YUAN ; Jing-Yi LIU ; Jiu-Mei HUANG ; Yu ZHOU
Chinese Pharmacological Bulletin 2024;40(12):2288-2294
Aim To observe the effect of Astragalus membranaceus and Angelica sinensis on the apoptosis of chondrocytes,and to investigate the effect of Astrag-alus membranaceus and Angelica sinensis on the sur-vival of fresh ostecartilage allograft.Methods Forty-eight 4-month-old New Zealand white rabbits,half male and half female,were randomly divided into sham operation group,model group,positive group and As-tragalus and Angelica 5∶1 group.In addition to the sham operation group,the other groups were both male and female donors and recipients for knee joint osteo-cartilage cross transplantation modeling.After 8 weeks of drug intervention,samples were taken for general observation,HE staining,saffrane-O staining,immu-nohistochemical staining,qPCR and Western blot de-tection.Results Compared with model group,As-tragalus and Angelica 5∶1 group and positive group,the repair site healed better,the morphology of osteo-chondrocytes tended to be normal,and the division and proliferation were obvious.Proteoglycan deposition in-creased and type Ⅱ collagen content was higher,the differences were statistically significant(P<0.05).qPCR and Western blot results showed that compared with model group,the mRNA and protein expressions of Bax,caspase-3 and caspase-9 in other groups were significantly decreased(P<0.05).Conclusion As-tragalus and Angelica can promote the survival of fresh osteochondral allograft,and its mechanism may be re-lated to promoting collagen production,promoting chondrocyte proliferation and inhibiting chondrocyte apoptosis.
10.Analysis of the therapeutic effect of trochanteric flip osteotomy combined with Kocher-Langenbeck approach for high acetabular posterior wall fracture
Xiao-Pan WANG ; Xiao-Tian CHEN ; Ren-Jie LI ; Le-Yu LIU ; Xiu-Song DAI ; Jian-Zhong GUAN ; Min WU ; Xiao-Dong CHEN
China Journal of Orthopaedics and Traumatology 2024;37(7):706-712
Objective Evaluation of the clinical efficacy of f trochanteric flip osteotomy combined with Kocher-Langen-beck approach for high acetabular posterior wall fracture.Methods Between January 2020 and December 2022,20 patients with high acetabular posterior wall fractures were retrospectively analyzed,including 12 males and 8 females,aged 18 to 75 years old.They were divided into two groups according to the different surgical methods.Ten patients were treated with greater trochanteric osteotomy combined with Kocher-Langenbeck approach as the observation group,including 5 males and 5 fe-males,aged from 18 to 75 years old.Ten patients were treated with Kocher-Langenbeck approach alone as the control group,including 7 males and 3 females,aged from 18 to 71 years old.Matta reduction criteria were used to evaluate the reduction quality of the two groups,and Harris score was used to compare the hip function of the two groups at the latest follow-up.The operation time,blood loss and postoperative complications of the two groups were analyzed.Results All patients were followed up for 10 to 24 months.According to the Matta fracture reduction quality evaluation criteria,the observation group achieved anatomical reduction in 6 cases,satisfactory reduction in 3 cases,and unsatisfactory reduction in 1 case,while the control group only achieved anatomical reduction in 3 cases,satisfactory reduction in 3 cases,and unsatisfactory reduction in 4 cases.At the final follow-up,the Harris hip score ranged from 71.4 to 96.6 in the observation group and 65.3 to 94.5 in the control group.According to the results of Harris score.The hip joint function of the observation group was excellent in 6 cases,good in 3 cases,and fair in 1 case.The hip joint function of the control group was excellent in 2 cases,good in 3 cases,fair in 3 cases,and poor in 2 cases.In the observation group,the intraoperative blood loss ranged from 300 to 700 ml,and the operation dura-tion ranged from 120 to 180 min;in the control group,the intraoperative blood loss ranged from 300 to 650 ml,and the opera-tion duration ranged from 100 to 180 min.Complications in the observation group included 1 case of traumatic arthritis and 1 case of heterotopic ossification,while complications in the control group included 3 cases of traumatic arthritis,3 cases of het-erotopic ossification and 1 case of hip abduction weakness.Conclusions Trochanteric flip osteotomy combined with the Kocher-Langenbeck approach significantly improved anatomical fracture reduction rates,enhanced excellent and good hip joint function outcomes,and reduced surgical complication incidence compared to the Kocher-Langenbeck approach alone.Clinical application of this combined approach is promising,although larger studies are needed for further validation.

Result Analysis
Print
Save
E-mail