Pub Date : 2025-12-25DOI: 10.3724/zdxbyxb-2025-0480
Yuan Yuan, Rui Liu, Hefeng Huang
Primary cilia are microtubule-based organelles that project from the cell surface. They are present in cells from single-celled eukaryotes to vertebrates, including humans. Recent studies have found that primary cilia are also widely distributed in multiple organs and tissues of the reproductive system, where they influence reproductive function by directly participating in or indirectly regulating related signaling pathways, thereby affecting fertility. Primary cilia participate in the regulation of oocyte meiosis and development. They also influence sperm maturation by regulating the homeostatic microenvironment required for spermiogenesis. By mediating Hedgehog (Hh) and Wnt signaling pathways, primary cilia regulate endometrial receptivity and decidual response, thereby influencing the embryo implantation rate. Furthermore, primary cilia control migration, invasion, differentiation, and vascular remodeling of human chorionic villi mesenchymal stromal cells and trophoblasts. Structural or functional impairment of primary cilia may disrupt placental vascular remodeling, leading to placental hypoplasia, potentially through the downregulation of downstream target genes of the Hh signaling pathway. Moreover, primary cilia may be involved in ovarian aging, ovulation, and endocrine function. This article reviews the research progress on the relationship between primary cilia and fertility, explores the potential mechanisms underlying the roles of primary cilia in gamete development, endometrial receptivity, decidualization, placental development, and ovarian reproductive endocrine function, aiming to provide new insights for fertility preservation and the prevention and treatment of human reproductive disorders.
{"title":"[Research progress on the roles of primary cilia in fertility].","authors":"Yuan Yuan, Rui Liu, Hefeng Huang","doi":"10.3724/zdxbyxb-2025-0480","DOIUrl":"10.3724/zdxbyxb-2025-0480","url":null,"abstract":"<p><p>Primary cilia are microtubule-based organelles that project from the cell surface. They are present in cells from single-celled eukaryotes to vertebrates, including humans. Recent studies have found that primary cilia are also widely distributed in multiple organs and tissues of the reproductive system, where they influence reproductive function by directly participating in or indirectly regulating related signaling pathways, thereby affecting fertility. Primary cilia participate in the regulation of oocyte meiosis and development. They also influence sperm maturation by regulating the homeostatic microenvironment required for spermiogenesis. By mediating Hedgehog (Hh) and Wnt signaling pathways, primary cilia regulate endometrial receptivity and decidual response, thereby influencing the embryo implantation rate. Furthermore, primary cilia control migration, invasion, differentiation, and vascular remodeling of human chorionic villi mesenchymal stromal cells and trophoblasts. Structural or functional impairment of primary cilia may disrupt placental vascular remodeling, leading to placental hypoplasia, potentially through the downregulation of downstream target genes of the Hh signaling pathway. Moreover, primary cilia may be involved in ovarian aging, ovulation, and endocrine function. This article reviews the research progress on the relationship between primary cilia and fertility, explores the potential mechanisms underlying the roles of primary cilia in gamete development, endometrial receptivity, decidualization, placental development, and ovarian reproductive endocrine function, aiming to provide new insights for fertility preservation and the prevention and treatment of human reproductive disorders.</p>","PeriodicalId":24007,"journal":{"name":"Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences","volume":" ","pages":"764-771"},"PeriodicalIF":0.0,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12750055/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145453406","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}
Objectives: To investigate the role of PTEN-induced putative kinase 1 (PINK1) in regulating the viability, migration, and apoptosis of colorectal cancer (CRC) cells, and to explore its potential epigenetic mechanisms.
Methods: PINK1 was overex-pressed or knocked down in HCT116 and DLD1 CRC cell lines using lentiviral vectors, with efficiency verified by qRT-PCR and Western blotting. Cell proliferation was assessed using CCK-8 and colony formation. Cell migration was detected using wound healing and Transwell assays. Apoptosis was assessed using Hoechst 33258 staining. Protein levels of apoptosis-related and histone modification-related markers were analyzed by Western blotting. Genome-wide chromatin accessibility was profiled using assay for transposase-accessible chromatin with sequencing (ATAC-seq).
Results: PINK1 expression was significantly downregulated at both mRNA and protein levels in CRC tissues com-pared to normal tissues. PINK1 overexpression inhibited cell proliferation, colony formation, and migration in HCT116 and DLD1 cells (all P<0.05), whereas PINK1 knockdown promoted these malignant phenotypes (all P<0.05). PINK1 overexpression induced apoptosis, associated with decreased levels of anti-apoptotic proteins (MCL-1, BCL-2, BCL-XL) and increased pro-apoptotic BAX (all P<0.05), without altering p53 expression. Mechanistically, PINK1 overexpression reduced histone H3 lysine 9 trimethylation (H3K9me3) and histone H3 lysine 27 trimethylation (H3K27me3), and increased histone H3 lysine 9 acetylation (H3K9ac) and histone H3 lysine 27 acetylation (H3K27ac). It also downregulated key histone-modifying enzymes, including enhancer of Zeste homolog (EZH)2, EZH1, SUZ12, and histone deacetylase 3 (HDAC3) (all P<0.01). ATAC-seq revealed that PINK1 overexpression increased chromatin accessibility, particularly around transcription start sites.
Conclusions: PINK1 acts as a tumor suppressor in colorectal cancer by inhibiting proliferation and migration, promoting apoptosis, and remodeling the epigenetic landscape through altering histone modifications and enhancing chromatin accessibility.
{"title":"[PINK1 suppresses colorectal cancer cell growth through epigenetic regulation of histone modifications].","authors":"Meng Wang, Shijia Luan, Xiang Fan, Dong Han, Yuping Zhu","doi":"10.3724/zdxbyxb-2024-0652","DOIUrl":"10.3724/zdxbyxb-2024-0652","url":null,"abstract":"<p><strong>Objectives: </strong>To investigate the role of PTEN-induced putative kinase 1 (PINK1) in regulating the viability, migration, and apoptosis of colorectal cancer (CRC) cells, and to explore its potential epigenetic mechanisms.</p><p><strong>Methods: </strong><i>PINK1</i> was overex-pressed or knocked down in HCT116 and DLD1 CRC cell lines using lentiviral vectors, with efficiency verified by qRT-PCR and Western blotting. Cell proliferation was assessed using CCK-8 and colony formation. Cell migration was detected using wound healing and Transwell assays. Apoptosis was assessed using Hoechst 33258 staining. Protein levels of apoptosis-related and histone modification-related markers were analyzed by Western blotting. Genome-wide chromatin accessibility was profiled using assay for transposase-accessible chromatin with sequencing (ATAC-seq).</p><p><strong>Results: </strong>PINK1 expression was significantly downregulated at both mRNA and protein levels in CRC tissues com-pared to normal tissues. <i>PINK1</i> overexpression inhibited cell proliferation, colony formation, and migration in HCT116 and DLD1 cells (all <i>P</i><0.05), whereas <i>PINK1</i> knockdown promoted these malignant phenotypes (all <i>P</i><0.05). <i>PINK1</i> overexpression induced apoptosis, associated with decreased levels of anti-apoptotic proteins (MCL-1, BCL-2, BCL-XL) and increased pro-apoptotic BAX (all <i>P</i><0.05), without altering p53 expression. Mechanistically, <i>PINK1</i> overexpression reduced histone H3 lysine 9 trimethylation (H3K9me3) and histone H3 lysine 27 trimethylation (H3K27me3), and increased histone H3 lysine 9 acetylation (H3K9ac) and histone H3 lysine 27 acetylation (H3K27ac). It also downregulated key histone-modifying enzymes, including enhancer of Zeste homolog (EZH)2, EZH1, SUZ12, and histone deacetylase 3 (HDAC3) (all <i>P</i><0.01). ATAC-seq revealed that <i>PINK1</i> overexpression increased chromatin accessibility, particularly around transcription start sites.</p><p><strong>Conclusions: </strong>PINK1 acts as a tumor suppressor in colorectal cancer by inhibiting proliferation and migration, promoting apoptosis, and remodeling the epigenetic landscape through altering histone modifications and enhancing chromatin accessibility.</p>","PeriodicalId":24007,"journal":{"name":"Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences","volume":" ","pages":"820-829"},"PeriodicalIF":0.0,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12750058/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145662376","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}
<p><strong>Objectives: </strong>To investigate the molecular mechanism by which He's Yangchao recipe (HSYC) improves ovarian function in a mouse model of premature ovarian insufficiency (POI).</p><p><strong>Methods: </strong>Forty ICR mice were used to establish a POI model via intraperitoneal injection of cyclophosphamide and were randomly assigned to four groups: model control group, low-dose HSYC group, high-dose HSYC group, and estradiol group (positive control). Additionally, 10 age-matched ICR mice were selected as the blank control group. After intragastric intervention, the ovarian index, serum follicle-stimulating hormone (FSH) levels, and ovarian tissue expression of the FSH receptor (FSHR) were measured. A POI cell model was established by treating the human granulosa tumor cell line with 4-hydroxycyclophosphamide. The cells were divided into four groups: solvent control group, HSYC group, inhibitor control group, and inhibitor+HSYC group, which were treated with dimethyl sulfoxide, HSYC-containing serum and 8-oxoguanine DNA glycosylase 1 (OGG1) inhibitor TH5487, respectively. The expressions of OGG1, mitochondrial DNA (mtDNA) oxidative damage markers, and pyroptosis-related proteins were detected by molecular docking, Western blotting, and immunofluorescence, respectively.</p><p><strong>Results: </strong>Compared with the blank control group, the model control group showed a decreased ovarian index (<i>P</i><0.05) and increased serum FSH level (<i>P</i><0.01). The ovarian index was higher in both the low- and high-dose HSYC groups compared with the model control group (both <i>P</i><0.05). FSHR expression in ovarian tissue was lower in the model control group than in the blank control group, but was higher in the high-dose HSYC group compared with the model control group (both <i>P</i><0.05). Molecular docking confirmed strong binding affinity between the active components of HSYC and OGG1 (binding energy: -8.3 to -6.3 kcal/mol). Western blotting analysis revealed that OGG1 protein expression in the ovaries of the model control group was significantly reduced compared with the blank control group, while it increased in the low-dose HSYC group and the estradiol group (all <i>P</i><0.05). Immunofluorescence results demonstrated that the expression levels of mitochondrial transcription factor A (TFAM) and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) decreased in the model control group compared with the blank control group (both <i>P</i><0.01), whereas the expressions were significantly elevated in the high-dose HSYC group and the estradiol group (all <i>P</i><0.01). Cell experiments showed that TH5487 intervention increased the expression of 8-oxoguanine (8-OxoG) (<i>P</i><0.01), while HSYC-containing serum intervention reduced 8-OxoG expression and increased TFAM expression (both <i>P</i><0.01). The expres-sion of pyroptosis-related proteins (GSDMD, N-GSDMD, caspase-1, IL-1β) increased after TH5487 intervention
{"title":"[He<b>'</b>s Yangchao recipe ameliorates premature ovarian insuffi-ciency by regulating 8-oxoguanine DNA glycosylase 1 in mice].","authors":"Renxin Hu, Ying Zhao, Yu Wu, Yuting Zhang, Qing Liu, Fangxuan Lin, Qin Zhang, Chenyun Miao","doi":"10.3724/zdxbyxb-2025-0490","DOIUrl":"10.3724/zdxbyxb-2025-0490","url":null,"abstract":"<p><strong>Objectives: </strong>To investigate the molecular mechanism by which He's Yangchao recipe (HSYC) improves ovarian function in a mouse model of premature ovarian insufficiency (POI).</p><p><strong>Methods: </strong>Forty ICR mice were used to establish a POI model via intraperitoneal injection of cyclophosphamide and were randomly assigned to four groups: model control group, low-dose HSYC group, high-dose HSYC group, and estradiol group (positive control). Additionally, 10 age-matched ICR mice were selected as the blank control group. After intragastric intervention, the ovarian index, serum follicle-stimulating hormone (FSH) levels, and ovarian tissue expression of the FSH receptor (FSHR) were measured. A POI cell model was established by treating the human granulosa tumor cell line with 4-hydroxycyclophosphamide. The cells were divided into four groups: solvent control group, HSYC group, inhibitor control group, and inhibitor+HSYC group, which were treated with dimethyl sulfoxide, HSYC-containing serum and 8-oxoguanine DNA glycosylase 1 (OGG1) inhibitor TH5487, respectively. The expressions of OGG1, mitochondrial DNA (mtDNA) oxidative damage markers, and pyroptosis-related proteins were detected by molecular docking, Western blotting, and immunofluorescence, respectively.</p><p><strong>Results: </strong>Compared with the blank control group, the model control group showed a decreased ovarian index (<i>P</i><0.05) and increased serum FSH level (<i>P</i><0.01). The ovarian index was higher in both the low- and high-dose HSYC groups compared with the model control group (both <i>P</i><0.05). FSHR expression in ovarian tissue was lower in the model control group than in the blank control group, but was higher in the high-dose HSYC group compared with the model control group (both <i>P</i><0.05). Molecular docking confirmed strong binding affinity between the active components of HSYC and OGG1 (binding energy: -8.3 to -6.3 kcal/mol). Western blotting analysis revealed that OGG1 protein expression in the ovaries of the model control group was significantly reduced compared with the blank control group, while it increased in the low-dose HSYC group and the estradiol group (all <i>P</i><0.05). Immunofluorescence results demonstrated that the expression levels of mitochondrial transcription factor A (TFAM) and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) decreased in the model control group compared with the blank control group (both <i>P</i><0.01), whereas the expressions were significantly elevated in the high-dose HSYC group and the estradiol group (all <i>P</i><0.01). Cell experiments showed that TH5487 intervention increased the expression of 8-oxoguanine (8-OxoG) (<i>P</i><0.01), while HSYC-containing serum intervention reduced 8-OxoG expression and increased TFAM expression (both <i>P</i><0.01). The expres-sion of pyroptosis-related proteins (GSDMD, N-GSDMD, caspase-1, IL-1β) increased after TH5487 intervention ","PeriodicalId":24007,"journal":{"name":"Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences","volume":" ","pages":"794-804"},"PeriodicalIF":0.0,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12750044/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145482882","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}
<p><strong>Objectives: </strong>To evaluate the strategies and outcomes of assisted reproductive technology (ART) for fertility enhancement and preservation in women with malignant tumors, and to analyze ART outcomes across different tumor types.</p><p><strong>Methods: </strong>We conducted a retrospective analysis of female patients who underwent ART for fertility enhancement and preservation at the Reproductive Medicine Center of the Women's Hospital, Zhejiang University School of Medicine, between January 1, 2018 and December 31, 2023. A total of 163 ART-aided pregnancy patients with malignant tumors were included in the case group, among which 6 patients underwent embryo cryopreservation for fertility preservation before radiotherapy or chemotherapy. Additionally, 11 unmarried women underwent oocyte cryopreservation due to borderline ovarian tumors, ovarian cancer, breast cancer, or hematological malignancies. The control group was selected from women without a history of malignant tumors who received ART treatment during the same period, using propensity score matching at a ratio of 1∶2, resulting in 326 cases. Data were collected through the reproductive medical record system and telephone follow-up (as of October 31, 2024). Baseline characteristics, controlled ovarian hyperstimulation parameters, laboratory indicators, and pregnancy outcomes were compared between case and control groups and among patients with different tumor types, and the tumor recurrence of the patients was followed up.</p><p><strong>Results: </strong>Patients in the case group had significantly lower ovarian reserve (anti-Müllerian hormone, antral follicle count) and a higher proportion of diminished ovarian reserve compared to the control group (all <i>P</i><0.01). Regarding the ovulation induction protocol, the proportion of patients using a minimal stimulation protocol in the case group was significantly higher than that in the control group (29.45% <i>vs</i>. 12.88%, <i>P</i><0.01), and the total dosage of gonadotropins used was lower (<i>P</i><0.01). In terms of ART outcomes, there were no statistically significant differences between the two groups in the number of retrieved oocytes, number of high-quality embryos, fertilization rate, cumulative pregnancy rate, cumulative live birth rate, or miscarriage rate (all <i>P</i>>0.05). However, the number of oocyte retrieval cycles and embryo transfer cycles required to achieve a live birth outcome in the case group were significantly higher than those in the control group (both <i>P</i><0.05). Subgroup analysis showed that there were no significant differences in cumulative pregnancy rate and live birth rate among patients with different tumor types, such as thyroid cancer, reproductive system tumors, breast cancer and lung cancer (all <i>P</i>>0.05). Nevertheless, lung cancer patients had the lowest ovarian reserve and required the most oocyte retrieval cycles due to the older age; breast cancer patients had a relativ
目的:评价辅助生殖技术(ART)对恶性肿瘤女性生育能力保存和促进的策略和结果,并分析不同肿瘤类型的ART结果。方法:回顾性分析2018年1月1日至2023年12月31日期间在浙江大学医学院附属女子医院生殖中心接受ART保存或治疗的女性患者。病例组共纳入163例art辅助妊娠恶性肿瘤患者,其中6例患者在放疗或化疗前行胚胎冷冻保存以保存生育能力。此外,11名未婚女性因交界性卵巢肿瘤、卵巢癌、乳腺癌或血液系统恶性肿瘤接受卵母细胞冷冻保存。对照组选取同期接受ART治疗的无恶性肿瘤病史的女性,采用倾向评分匹配法,按1∶2的比例匹配326例。通过生殖医疗记录系统和电话随访收集数据(截至2024年10月31日)。比较病例组与对照组、不同肿瘤类型患者的基线特征、控制性卵巢过度刺激参数、实验室指标及妊娠结局,并随访患者肿瘤复发情况。结果:病例组患者卵巢储备(AMH、AFC)明显低于对照组,卵巢储备减少比例明显高于对照组(p < 12.88%, p < 0.05)。然而,病例组实现活产结果所需的卵母细胞回收周期和胚胎移植周期数量明显高于对照组(均为p结论:育龄恶性肿瘤妇女存在生育能力下降的风险。)抗逆转录病毒治疗可以有效地保持和促进生育能力,实现良好的妊娠和活产结局。建议放疗/化疗前及时开展多学科评估,制定个体化ART方案,保存或促进生育能力,以实现生殖目标或保障未来生育潜力。
{"title":"[Application of assisted reproductive technology in fertility enhancement and preservation for women with malignant tumors].","authors":"Chunmei Ma, Xiaoling Hu, Shanshan Zhang, Lanfeng Xing, Yingwei Zhang, Yimin Zhu","doi":"10.3724/zdxbyxb-2025-0620","DOIUrl":"10.3724/zdxbyxb-2025-0620","url":null,"abstract":"<p><strong>Objectives: </strong>To evaluate the strategies and outcomes of assisted reproductive technology (ART) for fertility enhancement and preservation in women with malignant tumors, and to analyze ART outcomes across different tumor types.</p><p><strong>Methods: </strong>We conducted a retrospective analysis of female patients who underwent ART for fertility enhancement and preservation at the Reproductive Medicine Center of the Women's Hospital, Zhejiang University School of Medicine, between January 1, 2018 and December 31, 2023. A total of 163 ART-aided pregnancy patients with malignant tumors were included in the case group, among which 6 patients underwent embryo cryopreservation for fertility preservation before radiotherapy or chemotherapy. Additionally, 11 unmarried women underwent oocyte cryopreservation due to borderline ovarian tumors, ovarian cancer, breast cancer, or hematological malignancies. The control group was selected from women without a history of malignant tumors who received ART treatment during the same period, using propensity score matching at a ratio of 1∶2, resulting in 326 cases. Data were collected through the reproductive medical record system and telephone follow-up (as of October 31, 2024). Baseline characteristics, controlled ovarian hyperstimulation parameters, laboratory indicators, and pregnancy outcomes were compared between case and control groups and among patients with different tumor types, and the tumor recurrence of the patients was followed up.</p><p><strong>Results: </strong>Patients in the case group had significantly lower ovarian reserve (anti-Müllerian hormone, antral follicle count) and a higher proportion of diminished ovarian reserve compared to the control group (all <i>P</i><0.01). Regarding the ovulation induction protocol, the proportion of patients using a minimal stimulation protocol in the case group was significantly higher than that in the control group (29.45% <i>vs</i>. 12.88%, <i>P</i><0.01), and the total dosage of gonadotropins used was lower (<i>P</i><0.01). In terms of ART outcomes, there were no statistically significant differences between the two groups in the number of retrieved oocytes, number of high-quality embryos, fertilization rate, cumulative pregnancy rate, cumulative live birth rate, or miscarriage rate (all <i>P</i>>0.05). However, the number of oocyte retrieval cycles and embryo transfer cycles required to achieve a live birth outcome in the case group were significantly higher than those in the control group (both <i>P</i><0.05). Subgroup analysis showed that there were no significant differences in cumulative pregnancy rate and live birth rate among patients with different tumor types, such as thyroid cancer, reproductive system tumors, breast cancer and lung cancer (all <i>P</i>>0.05). Nevertheless, lung cancer patients had the lowest ovarian reserve and required the most oocyte retrieval cycles due to the older age; breast cancer patients had a relativ","PeriodicalId":24007,"journal":{"name":"Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences","volume":" ","pages":"718-726"},"PeriodicalIF":0.0,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12750050/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145662368","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}
Extracellular vesicles (EVs) are membrane-bound vesicles secreted by cells, including exosomes, microvesicles, and apoptotic bodies, which play critical roles in intercellular communication, material transport, and signal transduction. In recent years, increasing evidence has highlighted the essential function of EVs in early embryo development. By carrying bioactive molecules such as proteins, nucleic acids (e.g., mRNA and miRNA), and lipids, EVs regulate embryonic gene expression, cell proliferation, differ-entiation, and the microenvironment. Studies have shown that EVs derived from various segments of the female reproductive tract can enhance embryonic developmental potential, improve embryo quality, and facilitate implantation. Additionally, EVs secreted by embryos themselves participate in intercellular communication and play pivotal roles during embryogenesis. This review summarizes recent advances in understanding the functions of EVs in early embryo development, discusses their roles in mediating cell-to-cell com-munication and regulating gene expression, and explores the potential applications in reproductive medicine and clinical practice, offering new perspectives for optimizing assisted reproductive technologies.
{"title":"[The role of extracellular vesicles in early embryo development and their application in assisted reproductive technologies].","authors":"Haichao Wang, Xiaoxuan Li, Hongyan Lan, Xiaomei Tong","doi":"10.3724/zdxbyxb-2025-0085","DOIUrl":"10.3724/zdxbyxb-2025-0085","url":null,"abstract":"<p><p>Extracellular vesicles (EVs) are membrane-bound vesicles secreted by cells, including exosomes, microvesicles, and apoptotic bodies, which play critical roles in intercellular communication, material transport, and signal transduction. In recent years, increasing evidence has highlighted the essential function of EVs in early embryo development. By carrying bioactive molecules such as proteins, nucleic acids (e.g., mRNA and miRNA), and lipids, EVs regulate embryonic gene expression, cell proliferation, differ-entiation, and the microenvironment. Studies have shown that EVs derived from various segments of the female reproductive tract can enhance embryonic developmental potential, improve embryo quality, and facilitate implantation. Additionally, EVs secreted by embryos themselves participate in intercellular communication and play pivotal roles during embryogenesis. This review summarizes recent advances in understanding the functions of EVs in early embryo development, discusses their roles in mediating cell-to-cell com-munication and regulating gene expression, and explores the potential applications in reproductive medicine and clinical practice, offering new perspectives for optimizing assisted reproductive technologies.</p>","PeriodicalId":24007,"journal":{"name":"Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences","volume":" ","pages":"772-784"},"PeriodicalIF":0.0,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12750051/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145193195","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}
Pub Date : 2025-12-25DOI: 10.3724/zdxbyxb-2025-0153
Jian Chen, Chun Feng, Min Jin
With the increasing survival rates of cancer patients, the demand for fertility preservation in women has become increasingly prominent. Ovarian tissue cryopreservation and transplantation (OTCT) is an emerging fertility preservation technique that offers a unique advantage over embryo or oocyte cryopreservation, as it does not require ovarian stimulation. This makes it particularly suitable for prepubertal girls requiring urgent gonadotoxic therapy and reproductive-age women who cannot delay cancer treatment. Clinical evidence confirms that OTCT can effectively restore female fertility-especially the potential for natural conception-and restore ovarian endocrine function. The OTCT process involves key steps such as patient evaluation, tissue processing, cryopreservation, and transplantation. The patient's age at cryopreservation, ovarian reserve status, and prior exposure to gonadotoxic therapy significantly influence the outcomes of fertility preserva-tion. Optimal tissue preparation and the choice of cryopreservation method are critical for preserving ovarian tissue viability. During processing, the size of ovarian tissue fragments must be carefully controlled to balance freezing efficiency and post-transplantation viability, with adjustments based on individual patient factors. Slow freezing remains the mainstream clinical method, while vitrification is still considered experimental, with its efficacy and safety under ongoing investigation. The number, size, and transplantation site of ovarian tissue grafts impact their biological activity and functional outcomes. Both orthotopic and heterotopic transplantation can restore endocrine function, but orthotopic sites are superior for restoring fertility. A major safety concern in OTCT is the potential risk of reintroducing malignant or premalignant cells upon reimplantation. Innovative techniques such as in vitro maturation of oocytes and artificial ovaries are being explored to mitigate this risk. This review summarizes recent clinical advances in OTCT, with a focus on its indications, efficacy, implementation strategies, and safety profile, aiming to provide a reference for further research and clinical practice in this field.
{"title":"[Ovarian tissue cryopreservation and transplantation: a review of clinical progress in fertility preservation].","authors":"Jian Chen, Chun Feng, Min Jin","doi":"10.3724/zdxbyxb-2025-0153","DOIUrl":"10.3724/zdxbyxb-2025-0153","url":null,"abstract":"<p><p>With the increasing survival rates of cancer patients, the demand for fertility preservation in women has become increasingly prominent. Ovarian tissue cryopreservation and transplantation (OTCT) is an emerging fertility preservation technique that offers a unique advantage over embryo or oocyte cryopreservation, as it does not require ovarian stimulation. This makes it particularly suitable for prepubertal girls requiring urgent gonadotoxic therapy and reproductive-age women who cannot delay cancer treatment. Clinical evidence confirms that OTCT can effectively restore female fertility-especially the potential for natural conception-and restore ovarian endocrine function. The OTCT process involves key steps such as patient evaluation, tissue processing, cryopreservation, and transplantation. The patient's age at cryopreservation, ovarian reserve status, and prior exposure to gonadotoxic therapy significantly influence the outcomes of fertility preserva-tion. Optimal tissue preparation and the choice of cryopreservation method are critical for preserving ovarian tissue viability. During processing, the size of ovarian tissue fragments must be carefully controlled to balance freezing efficiency and post-transplantation viability, with adjustments based on individual patient factors. Slow freezing remains the mainstream clinical method, while vitrification is still considered experimental, with its efficacy and safety under ongoing investigation. The number, size, and transplantation site of ovarian tissue grafts impact their biological activity and functional outcomes. Both orthotopic and heterotopic transplantation can restore endocrine function, but orthotopic sites are superior for restoring fertility. A major safety concern in OTCT is the potential risk of reintroducing malignant or premalignant cells upon reimplantation. Innovative techniques such as <i>in vitro</i> maturation of oocytes and artificial ovaries are being explored to mitigate this risk. This review summarizes recent clinical advances in OTCT, with a focus on its indications, efficacy, implementation strategies, and safety profile, aiming to provide a reference for further research and clinical practice in this field.</p>","PeriodicalId":24007,"journal":{"name":"Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences","volume":" ","pages":"727-736"},"PeriodicalIF":0.0,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12750056/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145490329","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}
Pub Date : 2025-12-17DOI: 10.3724/zdxbyxb-2025-0696
Huiying Zhang, Qing Fan, Yihua Zhang, Mengchen Liu, Guozhen Cui
Objectives: Ginkgolide B (GB), a natural bioactive compound, acts on multiple molecular targets; however, its potential multi-target-based therapeutic indications remain unexplored. This study used a network medicine framework to systematically predict novel therapeutic indications for GB, aiming to provide evidence to support its clinical repositioning.
Methods: GB targets were comprehensively identified by integrating data from TTD, CTD, and BindingDB databases, followed by literature-based validation. Disease-gene associations were compiled by integrating GWAS and OMIM datasets. The topological proximity between GB targets and disease modules within the human protein-protein interaction network was quantified using network proximity metrics, with statistical significance evaluated via Z-score and permutation testing. Candidate diseases were classified according to MeSH terms, visualized through network mapping, and further characterized by gene set similarity based on the Jaccard index. Experiments in RAW 264.7 macrophages were conducted to validate the inhibitory effect of GB on pro-inflammatory cytokines in cells.
Results: Twelve experimentally validated GB targets were identified, and a disease-gene set encompassing 680 diseases was established. Network proximity analysis revealed significant associations between GB and 22 diseases (Z<0, P<0.05), of which 11 have prior literature support and 11 represent novel predictions. Disease classification indicated that the predicted indications of GB were primarily enriched in immune and inflammatory disease categories, with notable examples including juvenile idiopathic arthritis (Z=-3.10) and Crohn's disease (Z=-3.05). In vitro experiments with macrophages demonstrated that GB suppresses the production of pro-inflammatory cytokines, thereby supporting its potential as a therapeutic agent for immune-mediated inflammatory diseases. Network analysis suggested that GB may exert pleiotropic regulatory effects across multiple diseases via key targets such as MAPK1. The strongest gene set similarity was observed between juvenile idiopathic arthritis and ankylosing spondylitis (Jaccard index=0.60).
Conclusions: This study demonstrates the utility of a network medicine approach in systematically predicting and partially validating novel indications for GB, particularly within immune-inflammatory disorders, including type 1 diabetes, inflammatory bowel disease, and rheumatoid arthritis. These findings provide a theoretical framework for the clinical repositioning of GB and highlight the potential utility of the network-based strategies in the systematic exploration of natural products.
{"title":"[Predicting and <i>in vitro</i> validating new indications of Ginkgolide B via network-medicine framework].","authors":"Huiying Zhang, Qing Fan, Yihua Zhang, Mengchen Liu, Guozhen Cui","doi":"10.3724/zdxbyxb-2025-0696","DOIUrl":"https://doi.org/10.3724/zdxbyxb-2025-0696","url":null,"abstract":"<p><strong>Objectives: </strong>Ginkgolide B (GB), a natural bioactive compound, acts on multiple molecular targets; however, its potential multi-target-based therapeutic indications remain unexplored. This study used a network medicine framework to systematically predict novel therapeutic indications for GB, aiming to provide evidence to support its clinical repositioning.</p><p><strong>Methods: </strong>GB targets were comprehensively identified by integrating data from TTD, CTD, and BindingDB databases, followed by literature-based validation. Disease-gene associations were compiled by integrating GWAS and OMIM datasets. The topological proximity between GB targets and disease modules within the human protein-protein interaction network was quantified using network proximity metrics, with statistical significance evaluated via Z-score and permutation testing. Candidate diseases were classified according to MeSH terms, visualized through network mapping, and further characterized by gene set similarity based on the Jaccard index. Experiments in RAW 264.7 macrophages were conducted to validate the inhibitory effect of GB on pro-inflammatory cytokines in cells.</p><p><strong>Results: </strong>Twelve experimentally validated GB targets were identified, and a disease-gene set encompassing 680 diseases was established. Network proximity analysis revealed significant associations between GB and 22 diseases (Z<0, <i>P</i><0.05), of which 11 have prior literature support and 11 represent novel predictions. Disease classification indicated that the predicted indications of GB were primarily enriched in immune and inflammatory disease categories, with notable examples including juvenile idiopathic arthritis (Z=-3.10) and Crohn's disease (Z=-3.05). <i>In vitro</i> experiments with macrophages demonstrated that GB suppresses the production of pro-inflammatory cytokines, thereby supporting its potential as a therapeutic agent for immune-mediated inflammatory diseases. Network analysis suggested that GB may exert pleiotropic regulatory effects across multiple diseases via key targets such as MAPK1. The strongest gene set similarity was observed between juvenile idiopathic arthritis and ankylosing spondylitis (Jaccard index=0.60).</p><p><strong>Conclusions: </strong>This study demonstrates the utility of a network medicine approach in systematically predicting and partially validating novel indications for GB, particularly within immune-inflammatory disorders, including type 1 diabetes, inflammatory bowel disease, and rheumatoid arthritis. These findings provide a theoretical framework for the clinical repositioning of GB and highlight the potential utility of the network-based strategies in the systematic exploration of natural products.</p>","PeriodicalId":24007,"journal":{"name":"Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences","volume":" ","pages":"1-12"},"PeriodicalIF":0.0,"publicationDate":"2025-12-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145858165","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p><strong>Objectives: </strong>To investigate the protective mechanism of quercetin, the core component of Zuo Gui Wan, against Alzheimer's disease through the PI3K/AKT signaling pathway, based on network pharmacology, molecular docking, and cell experi-ments.</p><p><strong>Methods: </strong>The active components of Zuo Gui Wan were identified by searching TCMSP, PubChem, Swiss Target Prediction, and BATMAN-TCM databases, and their potential targets were predicted. The target information was standardized using Uniprot, and Alzheimer's disease-related target genes were obtained from Drugbank, GeneCards, and OMIM. The intersection of these datasets was used to identify the potential targets of Zuo Gui Wan for treating Alzheimer's disease. Gene Ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. The protein-protein interaction (PPI) network of potential targets was visualized using Cytoscape 3.10.1 software and the STRING database. The key active compounds and core potential targets for treating Alzheimer's disease with Zuo Gui Wan were identified through calculation. Based on the enrichment analysis results and literature, quercetin and the PI3K/AKT pathway were selected for verification. Molecular docking and binding ability prediction between quercetin and the core target AKT were performed using CB-Dock2, and visualization was conducted with AutoDock and PyMOL software. Finally, Aβ<sub>1-42</sub>-induced HT-22 mouse hippocampal neuronal cells were used to construct an Alzheimer's disease cell model. Quercetin, the PI3K inhibitor LY294002, and the activator EGF were used as interventions. The groups were divided as follows: Control, Aβ<sub>1-42</sub>, Aβ<sub>1-42</sub>+Quercetin 2.5 μM, Aβ<sub>1-42</sub>+Quercetin 5 μM, Aβ<sub>1-42</sub>+Quercetin 10 μM, Aβ<sub>1-42</sub>+EGF, and the PI3K/AKT modulation group: Control, LY294002, LY294002+Quercetin 10 μM, LY294002+EGF. CCK-8 assays were performed to detect cell viability, while JC-1, Calcein AM-PI, and Hoechst staining were used to assess cell apoptosis. Western blotting was employed to detect the expression of relevant target proteins.</p><p><strong>Results: </strong>Network pharmacology and cell experiments collectively demonstrate that the key active ingredient of Zuo Gui Wan, quercetin, targets core proteins such as AKT1 and GSK3β through a network-based approach, significantly enriching the PI3K/AKT pathway. Molecular docking results indicate that quercetin has a strong binding affinity with AKT. Experimental validation in the Aβ<sub>1-42</sub> oligomer-induced HT-22 model reveals that quercetin significantly activates the PI3K/AKT signaling pathway, which is inhibited by Aβ<sub>1-42</sub> oligomers, as well as Bcl-2 protein expression. It also suppresses the expression of Cleaved Caspase 3/Caspase 3, BAX, and Cytochrome C proteins. JC-1, Hoechst 33342, and Calcein AM-PI staining results further show that quercet
{"title":"[Protective effects of quercetin, the key component of Zuo Gui Wan, against Alzheimer<b>'</b>s disease via the PI3K/AKT pathway: insights from network pharmacology, molecular docking, and cell experiments].","authors":"Guangya Li, Peize Li, Liuling Huang, Jingwen Zhu, Xiude Qin, Yunwei Lu","doi":"10.3724/zdxbyxb-2025-0661","DOIUrl":"https://doi.org/10.3724/zdxbyxb-2025-0661","url":null,"abstract":"<p><strong>Objectives: </strong>To investigate the protective mechanism of quercetin, the core component of Zuo Gui Wan, against Alzheimer's disease through the PI3K/AKT signaling pathway, based on network pharmacology, molecular docking, and cell experi-ments.</p><p><strong>Methods: </strong>The active components of Zuo Gui Wan were identified by searching TCMSP, PubChem, Swiss Target Prediction, and BATMAN-TCM databases, and their potential targets were predicted. The target information was standardized using Uniprot, and Alzheimer's disease-related target genes were obtained from Drugbank, GeneCards, and OMIM. The intersection of these datasets was used to identify the potential targets of Zuo Gui Wan for treating Alzheimer's disease. Gene Ontology (GO) functional enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed. The protein-protein interaction (PPI) network of potential targets was visualized using Cytoscape 3.10.1 software and the STRING database. The key active compounds and core potential targets for treating Alzheimer's disease with Zuo Gui Wan were identified through calculation. Based on the enrichment analysis results and literature, quercetin and the PI3K/AKT pathway were selected for verification. Molecular docking and binding ability prediction between quercetin and the core target AKT were performed using CB-Dock2, and visualization was conducted with AutoDock and PyMOL software. Finally, Aβ<sub>1-42</sub>-induced HT-22 mouse hippocampal neuronal cells were used to construct an Alzheimer's disease cell model. Quercetin, the PI3K inhibitor LY294002, and the activator EGF were used as interventions. The groups were divided as follows: Control, Aβ<sub>1-42</sub>, Aβ<sub>1-42</sub>+Quercetin 2.5 μM, Aβ<sub>1-42</sub>+Quercetin 5 μM, Aβ<sub>1-42</sub>+Quercetin 10 μM, Aβ<sub>1-42</sub>+EGF, and the PI3K/AKT modulation group: Control, LY294002, LY294002+Quercetin 10 μM, LY294002+EGF. CCK-8 assays were performed to detect cell viability, while JC-1, Calcein AM-PI, and Hoechst staining were used to assess cell apoptosis. Western blotting was employed to detect the expression of relevant target proteins.</p><p><strong>Results: </strong>Network pharmacology and cell experiments collectively demonstrate that the key active ingredient of Zuo Gui Wan, quercetin, targets core proteins such as AKT1 and GSK3β through a network-based approach, significantly enriching the PI3K/AKT pathway. Molecular docking results indicate that quercetin has a strong binding affinity with AKT. Experimental validation in the Aβ<sub>1-42</sub> oligomer-induced HT-22 model reveals that quercetin significantly activates the PI3K/AKT signaling pathway, which is inhibited by Aβ<sub>1-42</sub> oligomers, as well as Bcl-2 protein expression. It also suppresses the expression of Cleaved Caspase 3/Caspase 3, BAX, and Cytochrome C proteins. JC-1, Hoechst 33342, and Calcein AM-PI staining results further show that quercet","PeriodicalId":24007,"journal":{"name":"Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences","volume":" ","pages":"1-12"},"PeriodicalIF":0.0,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145709110","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-08DOI: 10.3724/zdxbyxb-2025-0567
Zirong Lu, Yufeng Lu, Ji Zhou, Yichao Zhu
<p><strong>Objectives: </strong>To investigate the expression patterns and prognostic value of lipid metabolism-related genes in breast cancer.</p><p><strong>Methods: </strong>RNA sequencing data and clinical information were obtained from The Cancer Genome Atlas breast cancer (TCGA-BRCA) dataset, including 1100 breast cancer tissue samples (18 paired with adjacent tissues) and 112 normal breast tissue samples. Differentially expressed lipid metabolism-related genes were screened from a predefined set of 2043 genes using Bioconductor in R, with a false discovery rate <0.05 and |log2(fold change)|>2. Eligible samples were randomly divided into a training cohort (<i>n</i>=651) and a validation cohort (<i>n</i>=431) at a 6∶4 ratio. Prognostic lipid metabolism-related genes were identified using univariate Cox regression (<i>P</i><0.005) and further refined via LASSO regression. A risk score model was constructed using multivariate Cox regression, and patients were stratified into high- and low-risk groups based on the median risk score. The model's performance was evaluated using Kaplan-Meier survival analysis with the log-rank test and time-dependent receiver operating characteristic (ROC) curves. A nomogram integrating age, TNM stage, clinical grade, and risk score was developed and validated using calibration curves and the concordance index. Immune cell infiltration was quantified using an immune scoring algorithm, and weighted gene co-expression network analysis (WGCNA) was applied to identify key modules associated with immune cell infiltration. Finally, to validate the function of the key gene <i>ALDH2</i>, small interfering RNA targeting <i>ALDH2</i> was transfected into breast cancer cells, and its effects on invasion and migration were assessed using Transwell invasion and wound healing assays.</p><p><strong>Results: </strong>A total of 185 differentially expressed lipid metabolism-related genes were identified. Univariate Cox and LASSO regression analyses identified three genes-<i>ALDH2, CYP21A2,</i> and <i>IL24</i>-which were incorporated into the multivariate Cox model. The prognostic model based on these genes demonstrated good predictive performance in both cohorts: patients in the high-risk group had significantly shorter overall survival (<i>P</i><0.01), and the area under the ROC curve for predicting 1-, 3-, and 5-year survival rates was above 0.64. Analysis of the tumor microenvironment revealed an immunosuppressive phenotype in the high-risk group, characterized by reduced infiltration of several anti-tumor immune cells and downregulation of key immune checkpoint molecules such as PDCD1 and CTLA4. WGCNA suggested an association between <i>ALDH2</i> and immune cell infiltration. Functional experi-ments confirmed that <i>ALDH2</i> knockdown significantly enhanced the migration and invasion abilities of breast cancer cells.</p><p><strong>Conclusions: </strong>This study established and validated a pro-gnostic model for breast cancer
目的:探讨脂质代谢相关基因在乳腺癌中的表达规律及预后价值。方法:从The Cancer Genome Atlas breast Cancer (TCGA-BRCA)数据集中获取RNA测序数据和临床信息,包括1100例乳腺癌组织样本(18例与邻近组织配对)和112例正常乳腺组织样本。使用Bioconductor软件从预定义的2043个基因中筛选差异表达的脂质代谢相关基因,错误发现率为2。符合条件的样本按6∶4的比例随机分为训练组(n=651)和验证组(n=431)。采用单因素Cox回归(单变量Cox regression, PALDH2)鉴定预后脂质代谢相关基因,将靶向ALDH2的小干扰RNA转染到乳腺癌细胞中,并通过Transwell侵袭和伤口愈合试验评估其对侵袭和迁移的影响。结果:共鉴定出185个脂质代谢相关差异表达基因。单因素Cox和LASSO回归分析确定了三个基因- aldh2, CYP21A2和il24,并将其纳入多因素Cox模型。基于这些基因的预后模型在两个队列中都显示出良好的预测性能:高危组患者的总生存期(PALDH2和免疫细胞浸润)明显较短。功能实验证实,ALDH2敲低可显著增强乳腺癌细胞的迁移和侵袭能力。结论:本研究建立并验证了基于脂质代谢相关基因的乳腺癌预测模型。结果表明,ALDH2低表达与乳腺癌预后不良和免疫抑制密切相关,提示其作为乳腺癌预后生物标志物和治疗靶点的潜力。
{"title":"[Construction of a prognostic model for breast cancer based on lipid metabolism-related genes and functional verification of <i>ALDH2</i>].","authors":"Zirong Lu, Yufeng Lu, Ji Zhou, Yichao Zhu","doi":"10.3724/zdxbyxb-2025-0567","DOIUrl":"https://doi.org/10.3724/zdxbyxb-2025-0567","url":null,"abstract":"<p><strong>Objectives: </strong>To investigate the expression patterns and prognostic value of lipid metabolism-related genes in breast cancer.</p><p><strong>Methods: </strong>RNA sequencing data and clinical information were obtained from The Cancer Genome Atlas breast cancer (TCGA-BRCA) dataset, including 1100 breast cancer tissue samples (18 paired with adjacent tissues) and 112 normal breast tissue samples. Differentially expressed lipid metabolism-related genes were screened from a predefined set of 2043 genes using Bioconductor in R, with a false discovery rate <0.05 and |log2(fold change)|>2. Eligible samples were randomly divided into a training cohort (<i>n</i>=651) and a validation cohort (<i>n</i>=431) at a 6∶4 ratio. Prognostic lipid metabolism-related genes were identified using univariate Cox regression (<i>P</i><0.005) and further refined via LASSO regression. A risk score model was constructed using multivariate Cox regression, and patients were stratified into high- and low-risk groups based on the median risk score. The model's performance was evaluated using Kaplan-Meier survival analysis with the log-rank test and time-dependent receiver operating characteristic (ROC) curves. A nomogram integrating age, TNM stage, clinical grade, and risk score was developed and validated using calibration curves and the concordance index. Immune cell infiltration was quantified using an immune scoring algorithm, and weighted gene co-expression network analysis (WGCNA) was applied to identify key modules associated with immune cell infiltration. Finally, to validate the function of the key gene <i>ALDH2</i>, small interfering RNA targeting <i>ALDH2</i> was transfected into breast cancer cells, and its effects on invasion and migration were assessed using Transwell invasion and wound healing assays.</p><p><strong>Results: </strong>A total of 185 differentially expressed lipid metabolism-related genes were identified. Univariate Cox and LASSO regression analyses identified three genes-<i>ALDH2, CYP21A2,</i> and <i>IL24</i>-which were incorporated into the multivariate Cox model. The prognostic model based on these genes demonstrated good predictive performance in both cohorts: patients in the high-risk group had significantly shorter overall survival (<i>P</i><0.01), and the area under the ROC curve for predicting 1-, 3-, and 5-year survival rates was above 0.64. Analysis of the tumor microenvironment revealed an immunosuppressive phenotype in the high-risk group, characterized by reduced infiltration of several anti-tumor immune cells and downregulation of key immune checkpoint molecules such as PDCD1 and CTLA4. WGCNA suggested an association between <i>ALDH2</i> and immune cell infiltration. Functional experi-ments confirmed that <i>ALDH2</i> knockdown significantly enhanced the migration and invasion abilities of breast cancer cells.</p><p><strong>Conclusions: </strong>This study established and validated a pro-gnostic model for breast cancer","PeriodicalId":24007,"journal":{"name":"Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences","volume":" ","pages":"1-10"},"PeriodicalIF":0.0,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145708815","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-11-20DOI: 10.3724/zdxbyxb-2025-0625
Zhiqiang Xiang, Yue Wu, Kaiyu Huang, Fuqiang Wu, Ju Lin, Lieyong Sang, Liming Yang
Objectives: To characterize the biological properties of double-negative T (DNT) cells isolated from leukoreduction filter residues.
Methods: Leukoreduction filters containing residues from 400 mL whole blood units (n=6) were collected from a blood center. Filters were back-flushed with normal saline, and the eluate was concentrated to obtain leukoreduction filter residues. Leukocytes in the residues were counted by dual-fluorescence staining. DNT cells were then isolated from the residues using antibody-mediated adsorption and density gradient centrifugation. Both cryopreserved and fresh unstimulated DNT cells derived from the residues were subjected to in vitro culture. Following culture, cells were assessed for expansion fold, viability, immunophenotype, differentiation status, and cytotoxicity against target cells using dual-fluorescence staining and flow cytometry, with comparisons made to DNT cells derived from whole blood.
Results: The leukocyte recovery rate achieved through reverse flushing of the leukocyte reduction filter was (41.9±14.7)%. Compared to whole blood, the DNT cell starting material obtained from filter residues showed no significant difference in total T-cell content (P>0.05). However, the viability and purity of the resulting DNT cell starting materials were significantly lower (both P<0.05). After 17 days of culture, DNT cells from filter residues and whole blood showed no significant differences in expansion fold, immunophenotype, differentiation status, or cytotoxicity toward target cells (all P>0.05). However, the viability of DNT cells from residues was significantly lower than that of whole blood-derived DNT cells [(86.0±4.2)% vs. (92.2±1.2)%, P<0.05]. After thawing (post 3 or 15 days of cryopreservation) and 17 days of culture, DNT cell starting materials from residues showed comparable immunophenotype, expansion fold, and differentiation status to their non-cryopreserved counterparts from the same source (all P>0.05). However, the viability of DNT cells cryopreserved for 3 days [92.4% (91.8%, 92.8%)] and the cytotoxicity against target cells of those cryopreserved for 15 days [91.3% (89.4%, 95.1%)] were significantly higher than those of non-cryopreserved DNT cells [87.8% (82.0%, 89.0%) and 70.9% (67.3%, 80.2%), respectively] (P<0.05).
Conclusions: DNT cells derived from leukoreduction filter residues exhibited highly comparable characteristics to those from whole blood in terms of expansion, purity, differentiation, and biological potency. Furthermore, their biological activity post-cryopreservation and revival remained largely similar to non-cryopreserved cells. These findings suggest that leukoreduction filter residues represent a promising alternative source of starting material for manufacturing off-the-shelf, allogeneic DNT cell therapeutics.
{"title":"[Functional characterization of double-negative T cells isolated from leukoreduction filter residues].","authors":"Zhiqiang Xiang, Yue Wu, Kaiyu Huang, Fuqiang Wu, Ju Lin, Lieyong Sang, Liming Yang","doi":"10.3724/zdxbyxb-2025-0625","DOIUrl":"https://doi.org/10.3724/zdxbyxb-2025-0625","url":null,"abstract":"<p><strong>Objectives: </strong>To characterize the biological properties of double-negative T (DNT) cells isolated from leukoreduction filter residues.</p><p><strong>Methods: </strong>Leukoreduction filters containing residues from 400 mL whole blood units (<i>n</i>=6) were collected from a blood center. Filters were back-flushed with normal saline, and the eluate was concentrated to obtain leukoreduction filter residues. Leukocytes in the residues were counted by dual-fluorescence staining. DNT cells were then isolated from the residues using antibody-mediated adsorption and density gradient centrifugation. Both cryopreserved and fresh unstimulated DNT cells derived from the residues were subjected to <i>in vitro</i> culture. Following culture, cells were assessed for expansion fold, viability, immunophenotype, differentiation status, and cytotoxicity against target cells using dual-fluorescence staining and flow cytometry, with comparisons made to DNT cells derived from whole blood.</p><p><strong>Results: </strong>The leukocyte recovery rate achieved through reverse flushing of the leukocyte reduction filter was (41.9±14.7)%. Compared to whole blood, the DNT cell starting material obtained from filter residues showed no significant difference in total T-cell content (<i>P</i>>0.05). However, the viability and purity of the resulting DNT cell starting materials were significantly lower (both <i>P</i><0.05). After 17 days of culture, DNT cells from filter residues and whole blood showed no significant differences in expansion fold, immunophenotype, differentiation status, or cytotoxicity toward target cells (all <i>P</i>>0.05). However, the viability of DNT cells from residues was significantly lower than that of whole blood-derived DNT cells [(86.0±4.2)% <i>vs.</i> (92.2±1.2)%, <i>P</i><0.05]. After thawing (post 3 or 15 days of cryopreservation) and 17 days of culture, DNT cell starting materials from residues showed comparable immunophenotype, expansion fold, and differentiation status to their non-cryopreserved counterparts from the same source (all <i>P</i>>0.05). However, the viability of DNT cells cryopreserved for 3 days [92.4% (91.8%, 92.8%)] and the cytotoxicity against target cells of those cryopreserved for 15 days [91.3% (89.4%, 95.1%)] were significantly higher than those of non-cryopreserved DNT cells [87.8% (82.0%, 89.0%) and 70.9% (67.3%, 80.2%), respectively] (<i>P</i><0.05).</p><p><strong>Conclusions: </strong>DNT cells derived from leukoreduction filter residues exhibited highly comparable characteristics to those from whole blood in terms of expansion, purity, differentiation, and biological potency. Furthermore, their biological activity post-cryopreservation and revival remained largely similar to non-cryopreserved cells. These findings suggest that leukoreduction filter residues represent a promising alternative source of starting material for manufacturing off-the-shelf, allogeneic DNT cell therapeutics.</p>","PeriodicalId":24007,"journal":{"name":"Zhejiang da xue xue bao. Yi xue ban = Journal of Zhejiang University. Medical sciences","volume":" ","pages":"1-9"},"PeriodicalIF":0.0,"publicationDate":"2025-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145589032","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}