Jing Feng, Shan Li, Laihua Li, Zhiqiang Du, Guangying Yang, Zhi Zhao, Xueke Fan, Na Wang, Zhigang Zhao
{"title":"AMBRA1 Inhibits Non-Small Cell Lung Cancer Progression Through miR-1178/p53/CDK2-Regulated Cell Cycle Arrest","authors":"Jing Feng, Shan Li, Laihua Li, Zhiqiang Du, Guangying Yang, Zhi Zhao, Xueke Fan, Na Wang, Zhigang Zhao","doi":"10.1111/jcmm.70610","DOIUrl":"https://doi.org/10.1111/jcmm.70610","url":null,"abstract":"<p>AMBRA1 is associated with a variety of pathological processes in cancer cells, but may have different functions in different tumour microenvironments or genetic backgrounds. In this study, the function and regulatory mechanisms of AMBRA1 were explored in the progression of non-small cell lung cancer (NSCLC). The abnormally expressed miRNAs in AMBRA1-overexpressed and differentially expressed genes in miR-1178-knockdown NSCLC cells were validated by RNA sequencing. Cell viability, proliferation, invasion, apoptosis, and cell cycle were tested through Cell Counting Kit-8 (CCK-8), EdU, colony formation, transwell, and flow cytometry. A mouse tumour xenograft model was conducted to assess the roles of the AMBRA1-miR-1178 axis on NSCLC progression in vivo. AMBRA1 overexpression suppressed NSCLC cell proliferation and invasion, while promoting apoptosis and G0/G1 phase cell cycle arrest in vitro, and inhibited tumour growth in vivo. RNA sequencing revealed miR-1178 as a target of AMBRA1. miR-1178 overexpression partially weakened the suppressive function of AMBRA1 on cell malignant biological behaviours. p53 and CDK2 were confirmed as the downstream targets of miR-1178. Silencing p53 or overexpressing CDK2 reversed the repressive effects of AMBRA1 on the development of NSCLC cells. AMBRA1 may suppress the malignant phenotype of NSCLC cells via regulating the miR-1178-p53-CDK2 signalling pathway.</p>","PeriodicalId":101321,"journal":{"name":"JOURNAL OF CELLULAR AND MOLECULAR MEDICINE","volume":"29 11","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jcmm.70610","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144206655","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Dual Targeting EZH2 and Histone Deacetylases in Human Uterine Sarcoma Cells Under Both 2D and 3D Culture Conditions","authors":"Mervat M. Omran, Somayeh Vafaei, Samar Alkhrait, Qiwei Yang, Ayman Al-Hendy","doi":"10.1111/jcmm.70626","DOIUrl":"https://doi.org/10.1111/jcmm.70626","url":null,"abstract":"<p>Uterine sarcoma is strongly associated with poor prognosis. However, its treatment options remain limited. Tazemetostat is a potent and selective EZH2 inhibitor with limited clinical application. Entinostat is one of the strong inhibitors for HDAC1 and HDAC3. This study aimed to assess the effect of dual targeting of EZH2 and HDACs on the phenotype of uterine sarcoma cells in both 2D and 3D culture systems. The uterine sarcoma cell line (MES-SA) was treated with varying concentrations of tazemetostat and/or entinostat for 24, 48 and 72 h. For 3D culture conditions, the cells were combined with Matrigel and seeded in V-bottom plates and incubated for 5 days. Cell proliferation, cell cycle progression and apoptosis were evaluated. Additionally, the RNA expression, IHC staining, wound healing assay, DNMT and HDAC activity measurements were conducted. Our data showed that single-inhibitor treatment with entinostat or tazemetostat significantly increased the cytotoxicity and significantly enhanced apoptosis concomitantly. Furthermore, both inhibitors induced cell cycle arrest in 2D and 3D culture conditions. We also demonstrated that entinostat, but not tazemetostat, suppressed the wound healing in the 2D culture. The combination treatment showed a significantly superior effect compared to single-agent treatment. Our studies demonstrate that treatment with either entinostat or tazemetostat alone showed a potent anti-uterine sarcoma effect in 2D and 3D culture conditions. Importantly, the combination of entinostat and tazemetostat produced superior therapeutic effects, suggesting that dual targeting EZH2 and HDACs may provide a promising treatment option for this aggressive cancer.</p>","PeriodicalId":101321,"journal":{"name":"JOURNAL OF CELLULAR AND MOLECULAR MEDICINE","volume":"29 11","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jcmm.70626","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144206460","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Novel High-Resolution Lipidomes Could Serve as New Biomarkers for Diabetic Retinopathy: A Bidirectional and Mediated Mendelian Randomization Study","authors":"Yuxin Sun, Ziran Zhang, Zejun Chen, Zhengran Li, Zijin Wang, Fanye Wu, Xinyu Ma, Shaoyu Wang, Mingzhe Cao, Guoguo Yi, Min Fu","doi":"10.1111/jcmm.70614","DOIUrl":"https://doi.org/10.1111/jcmm.70614","url":null,"abstract":"<p>Although lipid metabolism is a critical factor in the pathogenesis of diabetic retinopathy (DR), the connection between lipidome and DR is still a subject of debate. We aimed to demonstrate that lipidome could serve as novel biomarkers for DR and elucidate the mediating role of inflammatory factors. Data for our investigation are available from the GWAS catalogue and FinnGen Biobank. The bidirectional Mendelian randomization (MR) analyses were conducted to assess the “total effect” between lipidome and DR and its subtypes. Subsequently, the mediation analyses were performed to explore the involvement of circulating inflammatory proteins in mediating the connection between them. Mediation proportion was calculated to measure the contribution of inflammatory factors to the overall effect. Ultimately, a battery of sensitivity tests proceeded to examine the dependability of the findings. This study has revealed a causal relationship between lipidome and different stages of DR. Additionally, we have successfully discovered a range of new lipids that protect against DR and have the potential to serve as new markers. This study also highlights the important role of inflammatory factors in elucidating the protective mechanisms of lipids against DR and provides new perspectives on lipidomic-based treatments and cytokine-targeted interventions for DR.</p>","PeriodicalId":101321,"journal":{"name":"JOURNAL OF CELLULAR AND MOLECULAR MEDICINE","volume":"29 11","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jcmm.70614","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144206604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Evaluating the Potential of COL8A1 as a Therapeutic Target for Chemoresistance, Disease Progression, and a Prognostic Marker in Gastric Cancer","authors":"Chao Xu, MuZhen He, HongYuan Chen, LiangJie Chi, XiangYu Wang, ShuYuan Li, QingShui Wang, Yao Lin, FangQin Xue","doi":"10.1111/jcmm.70621","DOIUrl":"https://doi.org/10.1111/jcmm.70621","url":null,"abstract":"<p>This study aimed to identify key genes associated with post-chemotherapy recurrence in gastric cancer patients. Gene expression data from multiple cohorts were analysed to determine differentially expressed genes between recurrent and non-recurrent cases. A prognostic risk model incorporating COL8A1, HSPB7 and SLIT2 was developed and validated across six independent cohorts. The risk score demonstrated significant associations with disease-free and overall survival, tumour grade and molecular subtypes. Notably, the risk score showed potential as a predictor of immunotherapy response, outperforming established markers such as microsatellite instability score and Epstein–Barr virus status. Analysis of the tumour immune microenvironment revealed a correlation between risk score and M2 macrophage infiltration. A nomogram integrating the risk score with clinical factors demonstrated high accuracy in predicting patient survival. Further investigation of COL8A1 revealed its significant role in gastric cancer cell proliferation, metastasis, and chemoresistance. In vitro and in vivo experiments showed that COL8A1 knockdown inhibited cancer cell growth, invasion, and metastasis while enhancing chemosensitivity. These findings provide valuable insights into the molecular mechanisms of gastric cancer recurrence and offer potential biomarkers for prognosis and treatment response prediction. The study highlights the importance of integrating genomic data with clinical information to improve patient stratification and personalised treatment strategies in gastric cancer management.</p>","PeriodicalId":101321,"journal":{"name":"JOURNAL OF CELLULAR AND MOLECULAR MEDICINE","volume":"29 11","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jcmm.70621","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144206904","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Chengjie Wei, Xiaomin Hou, Xuelu Jiang, Ming Gao, Yan Gao, Lin Bi, Jisheng Nie, Liangyuan Zhao, Yiwei Shi, Xiaojiang Qin
{"title":"Plant-Derived Monomers for Grey Hair Reversal Through Upregulation of Melanogenesis and Tyrosinase Activity","authors":"Chengjie Wei, Xiaomin Hou, Xuelu Jiang, Ming Gao, Yan Gao, Lin Bi, Jisheng Nie, Liangyuan Zhao, Yiwei Shi, Xiaojiang Qin","doi":"10.1111/jcmm.70534","DOIUrl":"https://doi.org/10.1111/jcmm.70534","url":null,"abstract":"<p>Grey hair, a common ageing-associated phenomenon in humans, is mainly attributed to the damage of melanocytes and the absence of melanin. Grey hair has long been treated with traditional medicine, and new research has shown that various plant-derived monomers can increase tyrosinase activity and melanogenesis, indicating that they may have therapeutic value in curing grey hair. In this study, we outlined the role of melanin and pigmentation during hair growth and collected various medicinal plant monomers with the potential value of grey hair reversal. Many active ingredients from medicinal plants, such as fraxinol, tribuloside, morin and naringenin, can upregulate melanogenesis and tyrosinase activity through different signalling pathways. Some of them can promote melanosome quantity, maturation and transportation as well. Monomers isolated from medicinal plants may act as stimulators of melanogenesis. Many plant-derived monomers perform as activators that upregulate melanin synthesis and tyrosinase activity through different signalling pathways. They are of great research value for the treatment of hair greying. Moreover, to further improve experimental effect, safety and reliability, a systematic and comprehensive evaluation system needs to be established in the future before studying their clinical efficacy.</p>","PeriodicalId":101321,"journal":{"name":"JOURNAL OF CELLULAR AND MOLECULAR MEDICINE","volume":"29 11","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jcmm.70534","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144197317","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Exploring the Effect and Mechanism of Liraglutide in Treating Depression Based on Network Pharmacology and Experimental Analysis","authors":"Jiangjin Sun, Xiying Fu, Yaqi Liu, Tian Wang, Xing Zhao, Ranji Cui, Wei Yang","doi":"10.1111/jcmm.70630","DOIUrl":"https://doi.org/10.1111/jcmm.70630","url":null,"abstract":"<p>Depression is a disorder caused by various reasons, with low mood as the main symptom, and it has a serious impact on mental health. Liraglutide (Lir) has been confirmed to alleviate neuroinflammation and depression-like behaviours induced by chronic stress, but its underlying mechanisms remain unclear. This study investigated the regulation of Lir for microglia-associated inflammation in depression through network pharmacology. In vivo experiments demonstrate that Lir reduces depressive-like behaviours by activating Nrf2 and subsequently downregulating HMGB1 expression, while also reducing the generation of pro-inflammatory mediators and oxidative stress damage. In vitro studies confirmed that the downregulation of HMGB1 depends on Nrf2 activation, and Lir activates Nrf2 via the PI3K/AKT pathway. Additionally, indirect co-culture of BV2 and HT22 cells demonstrated Lir's neuroprotective effects against neuronal apoptosis, consistent with findings from in vivo experiments. The study results first demonstrate that Lir exerts antidepressant effects through the PI3K/Nrf2/HMGB1 pathway, which reveals a novel mechanism of action for the antidepressant effects of Lir.</p>","PeriodicalId":101321,"journal":{"name":"JOURNAL OF CELLULAR AND MOLECULAR MEDICINE","volume":"29 11","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jcmm.70630","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144206905","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Multiomics-Driven Drug-Cell Interaction Network for Chemotherapy Sensitivity Prediction in Metabolically Defined Triple-Negative Breast Cancer Subtypes","authors":"Jingyuan Zhang, Xuejun Sun","doi":"10.1111/jcmm.70572","DOIUrl":"https://doi.org/10.1111/jcmm.70572","url":null,"abstract":"<p>Triple-negative breast cancer (TNBC) is associated with a poor prognosis due to insufficient molecular subtyping precision and limited actionable targets. Although metabolic reprogramming underlies TNBC chemotherapy resistance, establishing metabolic subtyping systems and investigating drug sensitivity across distinct metabolic subgroups could provide novel therapeutic avenues for breast cancer management. GSVA (Gene Set Variation Analysis) analysis of metabolic pathways reveals significant differences in TNBC (Triple-Negative Breast Cancer) patients. TNBC patients are classified into four metabolic subtypes through consensus clustering, based on their GSVA values of metabolic pathways. These subtypes are: MS_1, characterised by increased lipogenic activity; MS_2, characterised by increased carbohydrate and nucleotide metabolism; MS_3, a metabolism-active subtype with activation of all types of metabolism; and MS_4, characterised by suppressed metabolic activity across all types of metabolism. We next propose a novel method called MODIN (Multiomics-Driven Drug-Cell Interaction Network), which embeds multi-omics gene information (mRNA expression, copy number variation and DNA methylation) and drug SMILES data into a latent space, and then employs a multi-head attention-based interaction module to accurately predict the LN_IC50 values of 621 drugs in TNBC. Based on MODIN, noteworthy disparities in drug sensitivity emerge between the patient cohorts categorised as MS_2 and MS_3. MS_3 patients show a significantly higher sensitivity to chemotherapy regimens, especially for doxorubicin and docetaxel, while the MS_2 cohort displays marked resistance to these drugs. Our study reveals the metabolic heterogeneity of TNBC, and TNBC patients with increased carbohydrate and nucleotide metabolism exhibit the poorest prognoses and greater resistance to doxorubicin and docetaxel.</p>","PeriodicalId":101321,"journal":{"name":"JOURNAL OF CELLULAR AND MOLECULAR MEDICINE","volume":"29 11","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jcmm.70572","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144197106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Targeted ZnO@CuEA Nanoplatform for Cuproptosis-Based Synergistic Cancer Therapy","authors":"Hao Zhang, Guoyan Liu","doi":"10.1111/jcmm.70636","DOIUrl":"https://doi.org/10.1111/jcmm.70636","url":null,"abstract":"<p>Gastric cancer is a common malignant tumour. Copper-induced cell death, a recently discovered form of metal ion-related cell death, has garnered significant attention from researchers. We synthesised a multifunctional nanoparticle, ZnO@CuEA, and characterised its morphology using transmission electron microscopy. Cytotoxicity was analysed via CCK8 assays and calcein-AM staining. The tumour-targeting capability of ZnO@CuEA was validated using confocal microscopy and in vivo imaging experiments. RNA-seq and proteomic analysis were conducted to assess changes in mRNA and protein expression before and after ZnO@CuEA treatment. Lysosomal β-galactosidase staining was employed for cellular senescence detection, and protein expression levels were analysed via Western blot. Finally, in vivo experiments demonstrated the tumour-inhibitory effect of ZnO@CuEA. ZnO@CuEA is a multifunctional nanoparticle capable of targeting tumour cells and inducing cuproptosis. In vivo experiments showed that ZnO@CuEA exhibits significant antitumor activity. The multifunctional nanoparticles synthesised in this study provide a novel therapeutic approach for cancer treatment.</p>","PeriodicalId":101321,"journal":{"name":"JOURNAL OF CELLULAR AND MOLECULAR MEDICINE","volume":"29 11","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jcmm.70636","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144190833","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Machine Learning–Assisted Analysis of the Oral Cancer Immune Microenvironment: From Single-Cell Level to Prognostic Model Construction","authors":"Ling Yang, Lijuan Guo, Yun Zhu, Zehan Zhang","doi":"10.1111/jcmm.70637","DOIUrl":"https://doi.org/10.1111/jcmm.70637","url":null,"abstract":"<p>Oral cancer is among the most prevalent malignant tumours worldwide; prognosis can be affected by several factors, including molecular subtypes, immune microenvironment and clinical characteristics. In this study, we aimed to apply machine learning methods in conjunction with single-cell sequencing data to characterise the immune microenvironment of oral cancer and build an immune infiltration prediction model to provide a theoretical basis for the personalised therapy and prognosis assessment of oral cancer. Clinico-genomic data were obtained from patients with oral cancer and single-cell sequencing was utilised to delineate the immune cell composition in the tumour microenvironment. Model construction and immune-related gene screening were performed using machine learning algorithms such as Lasso regression, random forest and gradient boosting machine. We assessed the predictive performance of the model by cross-validation on its training dataset and by testing the model on an independent dataset. Certain subsets of immune cells correlate with the prognosis of patients with oral cancer. C-index (given in supplementary) yielded a good discrimination ability (C-index > 0.75) in the training set and validation set. Moreover, the model-identified immune-related genes presented remarkable expression differences in the two different risk groups and played important roles in the response to immune therapy. By exploring the complexity of the oral cancer immune microenvironment with machine learning techniques, in this study, we build a reliable prognostic model based on immune infiltration. The model could be applied in clinical practice to personalisation treatment decision-making and prognosis evaluation.</p>","PeriodicalId":101321,"journal":{"name":"JOURNAL OF CELLULAR AND MOLECULAR MEDICINE","volume":"29 11","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jcmm.70637","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144197515","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Correction to “The Uyghur Population and Genetic Susceptibility to Type 2 Diabetes: Potential Role for Variants in CAPN10, APM1 and FUT6 Genes”","authors":"","doi":"10.1111/jcmm.70638","DOIUrl":"https://doi.org/10.1111/jcmm.70638","url":null,"abstract":"<p>Zhao F, Mamatyusupu D, Wang Y, Fang H, Wang H, Gao Q, Dong H, Ge S, Yu X, Zhang J, Wu L, Song M, Wang W. The Uyghur Population and Genetic Susceptibility to Type 2 Diabetes: Potential Role for Variants in CAPN10, APM1 and FUT6 genes. <i>J Cell Mol Med</i>. 2016;20(11):2138-2147.</p><p>In the Acknowledgements section, the Australian National Health and Medical Research Council (NHMRC-APP1112767) and the Edith Cowan University Strategic Research Fund (SRF-2015) should be removed.</p><p>The Acknowledgement statement reads:</p><p>This study was supported by research grants from the National Natural Science Foundation of China (81573215, 81273170, 31460285 and 81370083), National 12th Five-Year Major Projects of China (2012BAI37B03), Australian National Health and Medical Research Council and National Natural Science Foundation of China (NHMRC-APP1112767-NSFC 81561128020), Edith Cowan University Strategic Research Fund (SRF-2015), Natural Science Foundation of Xinjiang Uyghur Autonomous Region (2013211A016), and Natural Science Foundation of Capital Medical University, Beijing, China (2014ZR16). Manshu Song was supported by the Importation and Development of High-Calibre Talents Project of Beijing Municipal Institutions (CIT&TCD201404185). The authors thank the Uyghur volunteers and community leaders for their supports and participation. We appreciate the English editing by Eric Adua, School of Medical Sciences and Health, Edith Cowan University, Australia.</p><p>The Acknowledgement statement should read:</p><p>This study was supported by research grants from the National Natural Science Foundation of China (81573215, 81273170, 31460285, 81370083 and 81561128020), National 12th Five-Year Major Projects of China (2012BAI37B03), Natural Science Foundation of Xinjiang Uyghur Autonomous Region (2013211A016) and Natural Science Foundation of Capital Medical University, Beijing, China (2014ZR16). Manshu Song was supported by the Importation and Development of High-Calibre Talents Project of Beijing Municipal Institutions (CIT&TCD201404185). The authors thank the Uyghur volunteers and community leaders for their supports and participation. We appreciate the English editing by Eric Adua, School of Medical Sciences and Health, Edith Cowan University, Australia.</p><p>We apologize for this error.</p>","PeriodicalId":101321,"journal":{"name":"JOURNAL OF CELLULAR AND MOLECULAR MEDICINE","volume":"29 11","pages":""},"PeriodicalIF":5.3,"publicationDate":"2025-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1111/jcmm.70638","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144171545","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}