首页 > 最新文献

Stem Cell Research & Therapy最新文献

英文 中文
Exosomal miR-149 from human umbilical cord mesenchymal stem cells attenuates spinal cord injury-induced blood-spinal cord barrier disruption by suppressing the ET-1/PI3K/Akt signaling pathway. 人脐带间充质干细胞外泌体miR-149通过抑制ET-1/PI3K/Akt信号通路减弱脊髓损伤诱导的血脊髓屏障破坏。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-12-31 DOI: 10.1186/s13287-025-04873-7
Chenhui Xue, Xiaochen Qiao, Wenxuan Wang, Zhenwu Gao, Xin Chen, Xihua Yang, Hui Wang, Jiansheng Jing, Haoyu Feng, Hui Zhang, Lin Sun, Xiaoming Guan

Background: Spinal cord injury (SCI) leads to persistent neurological deficits partly by disruption of the blood-spinal cord barrier (BSCB). Small extracellular vesicles (sEVs) from human umbilical cord mesenchymal stem cells (hUC-MSCs) can promote BSCB repair, but their active components remain unclear. This study examined whether miR-149 carried by hUC-MSC-derived sEVs (hUC-MSCs-sEVs) protects the BSCB after SCI by targeting endothelin-1 (ET-1).

Methods: Human brain microvascular endothelial cells (HBMECs) were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to model barrier injury, and rats underwent a thoracic SCI. hUC-MSCs-sEVs were isolated and loaded with miR-149 mimics or inhibitors. Endothelial cell viability, paracellular permeability (FITC-dextran assay), and junction protein levels (ZO-1, Claudin-5, β-Catenin, Occludin) were measured by viability assays, Western blot, and immunofluorescence. ET-1 levels and PI3K/Akt pathway activation were measured by ELISA and Western blot. In SCI rats, sEVs (with or without the miR-149 inhibitor) were injected; motor function (BBB locomotor score), BSCB permeability (Evans blue/FITC-dextran leakage) and spinal cord histology were evaluated.

Results: hUC-MSCs-sEVs were internalized by HBMECs and significantly improved cell survival and barrier function after OGD/R. sEVs treatment restored tight and adherens junction proteins and suppressed OGD/R-induced ET-1 upregulation and PI3K/Akt activation. OGD/R reduced miR-149 expression, which was rescued by sEVs. sEVs loaded with miR-149 mimic further enhanced these protective effects, whereas a miR-149 inhibitor abolished them. Notably, co-administration of an ET-1 receptor antagonist reversed the barrier disruption caused by miR-149 inhibition. In vivo, hUC-MSCs-sEVs treatment improved locomotor recovery and reduced BSCB leakage and tissue damage, whereas miR-149 inhibition abolished these benefits.

Conclusions: hUC-MSC-derived exosomal miR-149 preserves BSCB integrity and promotes functional recovery after SCI by targeting ET-1 and inhibiting the PI3K/Akt pathway, thereby enhancing junctional protein expression. The miR-149/ET-1 axis may represent a promising therapeutic target for SCI.

背景:脊髓损伤(SCI)导致持续的神经功能缺损,部分原因是血脊髓屏障(BSCB)的破坏。来自人脐带间充质干细胞(hUC-MSCs)的小细胞外囊泡(sev)可以促进BSCB修复,但其活性成分尚不清楚。本研究检测了huc - msc衍生的sev (huc - msc - sev)携带的miR-149是否通过靶向内皮素-1 (ET-1)来保护脊髓损伤后的BSCB。方法:采用氧-葡萄糖剥夺/再氧合(OGD/R)方法模拟脑屏障损伤,并对大鼠进行胸椎脊髓损伤。分离huc - mscs - sev并装载miR-149模拟物或抑制剂。内皮细胞活力、细胞旁通透性(fitc -葡聚糖测定)和连接蛋白水平(ZO-1、Claudin-5、β-Catenin、Occludin)通过活力测定、Western blot和免疫荧光测定。ELISA和Western blot检测ET-1水平和PI3K/Akt通路激活情况。在SCI大鼠中,注射sev(含或不含miR-149抑制剂);运动功能(BBB运动评分)、BSCB通透性(Evans蓝/ fitc -葡聚糖渗漏)和脊髓组织学。结果:huc - mscs - sev被hbmec内化,并显著提高OGD/R后的细胞存活率和屏障功能。sev处理恢复紧密和粘附连接蛋白,抑制OGD/ r诱导的ET-1上调和PI3K/Akt活化。OGD/R降低miR-149的表达,通过sev挽救。负载miR-149模拟物的sev进一步增强了这些保护作用,而miR-149抑制剂则消除了它们。值得注意的是,ET-1受体拮抗剂的联合施用逆转了miR-149抑制引起的屏障破坏。在体内,huc - msc - sev治疗改善了运动恢复,减少了BSCB泄漏和组织损伤,而miR-149抑制消除了这些益处。结论:huc - msc衍生的外泌体miR-149通过靶向ET-1和抑制PI3K/Akt通路,从而增强连接蛋白的表达,从而保持BSCB的完整性,促进SCI后功能恢复。miR-149/ET-1轴可能是一个有希望的脊髓损伤治疗靶点。
{"title":"Exosomal miR-149 from human umbilical cord mesenchymal stem cells attenuates spinal cord injury-induced blood-spinal cord barrier disruption by suppressing the ET-1/PI3K/Akt signaling pathway.","authors":"Chenhui Xue, Xiaochen Qiao, Wenxuan Wang, Zhenwu Gao, Xin Chen, Xihua Yang, Hui Wang, Jiansheng Jing, Haoyu Feng, Hui Zhang, Lin Sun, Xiaoming Guan","doi":"10.1186/s13287-025-04873-7","DOIUrl":"https://doi.org/10.1186/s13287-025-04873-7","url":null,"abstract":"<p><strong>Background: </strong>Spinal cord injury (SCI) leads to persistent neurological deficits partly by disruption of the blood-spinal cord barrier (BSCB). Small extracellular vesicles (sEVs) from human umbilical cord mesenchymal stem cells (hUC-MSCs) can promote BSCB repair, but their active components remain unclear. This study examined whether miR-149 carried by hUC-MSC-derived sEVs (hUC-MSCs-sEVs) protects the BSCB after SCI by targeting endothelin-1 (ET-1).</p><p><strong>Methods: </strong>Human brain microvascular endothelial cells (HBMECs) were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to model barrier injury, and rats underwent a thoracic SCI. hUC-MSCs-sEVs were isolated and loaded with miR-149 mimics or inhibitors. Endothelial cell viability, paracellular permeability (FITC-dextran assay), and junction protein levels (ZO-1, Claudin-5, β-Catenin, Occludin) were measured by viability assays, Western blot, and immunofluorescence. ET-1 levels and PI3K/Akt pathway activation were measured by ELISA and Western blot. In SCI rats, sEVs (with or without the miR-149 inhibitor) were injected; motor function (BBB locomotor score), BSCB permeability (Evans blue/FITC-dextran leakage) and spinal cord histology were evaluated.</p><p><strong>Results: </strong>hUC-MSCs-sEVs were internalized by HBMECs and significantly improved cell survival and barrier function after OGD/R. sEVs treatment restored tight and adherens junction proteins and suppressed OGD/R-induced ET-1 upregulation and PI3K/Akt activation. OGD/R reduced miR-149 expression, which was rescued by sEVs. sEVs loaded with miR-149 mimic further enhanced these protective effects, whereas a miR-149 inhibitor abolished them. Notably, co-administration of an ET-1 receptor antagonist reversed the barrier disruption caused by miR-149 inhibition. In vivo, hUC-MSCs-sEVs treatment improved locomotor recovery and reduced BSCB leakage and tissue damage, whereas miR-149 inhibition abolished these benefits.</p><p><strong>Conclusions: </strong>hUC-MSC-derived exosomal miR-149 preserves BSCB integrity and promotes functional recovery after SCI by targeting ET-1 and inhibiting the PI3K/Akt pathway, thereby enhancing junctional protein expression. The miR-149/ET-1 axis may represent a promising therapeutic target for SCI.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145879143","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Exosomes from LPS-pretreated BMSCs treated periodontitis via improving oxidative stress. lps预处理的骨髓间充质干细胞外泌体通过改善氧化应激治疗牙周炎。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-12-31 DOI: 10.1186/s13287-025-04860-y
Chenyu Xu, Hanping Wang, Wenqi Dong, Wen Cheng, Yuran Su, Qiang Yang, Yue Wang, Yanhong Zhao

Background: Research indicates that the occurrence of periodontitis is related to oxidative stress and mitochondrial dysfunction. Alleviating oxidative stress and mitochondrial dysfunction may be a promising treatment strategy for periodontitis. In this study, bone marrow mesenchymal stem cells (BMSCs) were pretreated with lipopolysaccharide (LPS), and their derived exosomes (LPS-BMSCs-Exo) were extracted. In vitro and in vivo experiments were conducted to study the therapeutic effects of alleviating oxidative stress, mitochondrial disorders, and periodontitis.

Methods: BMSCs were pretreated with LPS, and LPS-BMSCs-Exo were extracted and identified via transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blotting. The biosafety of the exosomes was assessed through CCK-8, migration, and uptake assays. A cell oxidative stress model was established and treated with BMSCs-Exo or LPS-BMSCs-Exo, the following tests were performed: the effects of the two types of exosomes on the oxidative stress of periodontal ligament stem cells (PDLSCs) were determined, the mitochondrial state and the membrane potential were detected, the content of adenosine triphosphate (ATP) was determined, apoptosis was detected, and the effect of the exosomes on the osteogenic ability of the PDLSCs was detected. A periodontitis rat model was established, and PBS, BMSCs-Exo, and LPS-BMSCs-Exo were administered separately. Micro-CT, HE staining, Masson staining, immunohistochemistry, and ROS fluorescence staining were used to evaluate the therapeutic effect of each group on periodontitis in rats.

Results: The proposed LPS-BMSCs-Exo exhibits characteristics similar to those of exosomes, can be successfully taken up and internalized by PDLSCs, and promotes the proliferation and migration of these cells. LPS-BMSCs-Exo can effectively improve the oxidative stress state, alleviate mitochondrial dysfunction in cells, increase membrane potential, enhance ATP content, reduce apoptosis, and improve the osteogenic ability of PDLSCs. Micro-CT data revealed that alveolar bone-related indicators were significantly increased after LPS-BMSCs-Exo treatment, which could reduce the degradation and inflammation of periodontal tissue in rats and alleviate their oxidative stress.

Conclusion: LPS-BMSCs-Exo can significantly alleviate the oxidative stress and mitochondrial dysfunction caused by periodontitis in periodontal tissue, thereby reducing inflammation in periodontal tissue and alveolar bone resorption.

背景:研究表明牙周炎的发生与氧化应激和线粒体功能障碍有关。减轻氧化应激和线粒体功能障碍可能是治疗牙周炎的一种有希望的治疗策略。本研究采用脂多糖(LPS)预处理骨髓间充质干细胞(BMSCs),提取其衍生外泌体(LPS-BMSCs- exo)。体外和体内实验研究其对氧化应激、线粒体疾病和牙周炎的治疗作用。方法:采用脂多糖预处理BMSCs,提取脂多糖-BMSCs- exo,通过透射电镜(TEM)、纳米颗粒跟踪分析(NTA)和Western blotting对其进行鉴定。通过CCK-8、迁移和摄取试验评估外泌体的生物安全性。建立细胞氧化应激模型,分别用BMSCs-Exo或LPS-BMSCs-Exo处理,检测两种外泌体对牙周韧带干细胞(periodontal ligament stem cells, PDLSCs)氧化应激的影响,检测线粒体状态和膜电位,检测三磷酸腺苷(adenosine triphosphate, ATP)含量,检测细胞凋亡,检测外泌体对PDLSCs成骨能力的影响。建立牙周炎大鼠模型,分别给药PBS、BMSCs-Exo和LPS-BMSCs-Exo。采用Micro-CT、HE染色、Masson染色、免疫组化、ROS荧光染色评价各组对大鼠牙周炎的治疗效果。结果:LPS-BMSCs-Exo具有与外泌体相似的特性,可被PDLSCs成功吸收和内化,并促进这些细胞的增殖和迁移。LPS-BMSCs-Exo能有效改善PDLSCs的氧化应激状态,缓解细胞线粒体功能障碍,增加膜电位,提高ATP含量,减少凋亡,提高PDLSCs的成骨能力。Micro-CT数据显示,LPS-BMSCs-Exo处理后,牙槽骨相关指标明显升高,可减轻大鼠牙周组织降解和炎症,减轻氧化应激。结论:LPS-BMSCs-Exo可显著缓解牙周炎引起的牙周组织氧化应激和线粒体功能障碍,从而减少牙周组织炎症和牙槽骨吸收。
{"title":"Exosomes from LPS-pretreated BMSCs treated periodontitis via improving oxidative stress.","authors":"Chenyu Xu, Hanping Wang, Wenqi Dong, Wen Cheng, Yuran Su, Qiang Yang, Yue Wang, Yanhong Zhao","doi":"10.1186/s13287-025-04860-y","DOIUrl":"https://doi.org/10.1186/s13287-025-04860-y","url":null,"abstract":"<p><strong>Background: </strong>Research indicates that the occurrence of periodontitis is related to oxidative stress and mitochondrial dysfunction. Alleviating oxidative stress and mitochondrial dysfunction may be a promising treatment strategy for periodontitis. In this study, bone marrow mesenchymal stem cells (BMSCs) were pretreated with lipopolysaccharide (LPS), and their derived exosomes (LPS-BMSCs-Exo) were extracted. In vitro and in vivo experiments were conducted to study the therapeutic effects of alleviating oxidative stress, mitochondrial disorders, and periodontitis.</p><p><strong>Methods: </strong>BMSCs were pretreated with LPS, and LPS-BMSCs-Exo were extracted and identified via transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blotting. The biosafety of the exosomes was assessed through CCK-8, migration, and uptake assays. A cell oxidative stress model was established and treated with BMSCs-Exo or LPS-BMSCs-Exo, the following tests were performed: the effects of the two types of exosomes on the oxidative stress of periodontal ligament stem cells (PDLSCs) were determined, the mitochondrial state and the membrane potential were detected, the content of adenosine triphosphate (ATP) was determined, apoptosis was detected, and the effect of the exosomes on the osteogenic ability of the PDLSCs was detected. A periodontitis rat model was established, and PBS, BMSCs-Exo, and LPS-BMSCs-Exo were administered separately. Micro-CT, HE staining, Masson staining, immunohistochemistry, and ROS fluorescence staining were used to evaluate the therapeutic effect of each group on periodontitis in rats.</p><p><strong>Results: </strong>The proposed LPS-BMSCs-Exo exhibits characteristics similar to those of exosomes, can be successfully taken up and internalized by PDLSCs, and promotes the proliferation and migration of these cells. LPS-BMSCs-Exo can effectively improve the oxidative stress state, alleviate mitochondrial dysfunction in cells, increase membrane potential, enhance ATP content, reduce apoptosis, and improve the osteogenic ability of PDLSCs. Micro-CT data revealed that alveolar bone-related indicators were significantly increased after LPS-BMSCs-Exo treatment, which could reduce the degradation and inflammation of periodontal tissue in rats and alleviate their oxidative stress.</p><p><strong>Conclusion: </strong>LPS-BMSCs-Exo can significantly alleviate the oxidative stress and mitochondrial dysfunction caused by periodontitis in periodontal tissue, thereby reducing inflammation in periodontal tissue and alveolar bone resorption.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145879156","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comparative evaluation of the therapeutic efficacy between human amniotic epithelial cells and human umbilical cord mesenchymal stem cells in premature ovarian insufficiency. 人羊膜上皮细胞与人脐带间充质干细胞治疗卵巢早衰的疗效比较。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-12-31 DOI: 10.1186/s13287-025-04881-7
Qinyu Zhang, Jie Wang, Zixin Cheng, Wenjiao Cao, Qiuwan Zhang, Dongmei Lai

Background: Premature ovarian insufficiency (POI) is a clinically challenging condition characterized by amenorrhea and infertility in women less than 40 years of age. Although both human amniotic epithelial cells (hAECs) and human umbilical cord mesenchymal stem cells (hUC-MSCs) have shown promise in treating POI, their comparative therapeutic efficacy and mechanisms remain poorly understood.

Methods: hAECs and hUC-MSCs were isolated from human amniotic membrane and umbilical cords, respectively, and characterized using standard protocols. A chemotherapy-induced POI mouse model was established to evaluate follicular development, ovarian fibrosis, and fertility recovery after hAEC and hUC-MSC transplantation. Longitudinal in vivo bioluminescence imaging was used to track and compare the biodistribution and retention rates of the transplanted cells. RNA sequencing and in vitro functional assays under oxidative stress and apoptosis-induced conditions were employed to analyze the differential stress responses of hAECs and hUC-MSCs. Furthermore, cytokine arrays were utilized to profile their secretomes.

Results: In the chemotherapy-induced POI mouse model, both hAECs and hUC-MSCs transplantation improved ovarian function, as evidenced by increased ovarian weight, restored estrous cycle, elevated follicle counts, reduced fibrosis, and enhanced fertility. In vivo imaging revealed that both cell types primarily homed to the lungs, liver, and spleen post-transplantation, with signal intensity declining over time. Quantitative analysis revealed significantly longer in vivo retention of hAECs compare to hUC-MSCs. RNA sequencing and in vitro assays confirmed the superior antioxidant capacity of hAECs under stress conditions. Cytokine profiling showed that hAEC-CM was enriched in pro-angiogenic factors, while hUC-MSC-CM contained higher levels of immunoregulatory cytokines, a functional difference further validated by in vitro experiments.

Conclusion: Our findings demonstrate that both hAECs and hUC-MSCs are effective in restoring ovarian function and fertility in a chemotherapy-induced POI mouse model. However, these two cell types exhibit distinct therapeutic advantages attributable to their differential metabolic kinetics and paracrine profiles. Specifically, hAECs displayed prolonged in vivo retention rates compared to hUC-MSCs, consistent with their enhanced antioxidant capabilities. In terms of secretory function, hAECs demonstrated superior pro-angiogenic activity, while hUC-MSCs exhibited stronger immunomodulatory effects. These distinct properties provide critical insights for cell-type-specific selection in developing targeted therapies for ovarian dysfunction.

背景:卵巢功能不全(POI)是一种临床上具有挑战性的疾病,以40岁以下女性闭经和不孕为特征。虽然人羊膜上皮细胞(hAECs)和人脐带间充质干细胞(hUC-MSCs)都显示出治疗POI的希望,但它们的比较治疗效果和机制仍然知之甚少。方法:分别从人羊膜和脐带中分离hAECs和hUC-MSCs,采用标准方法进行鉴定。建立化疗诱导的POI小鼠模型,评估hAEC和hUC-MSC移植后的卵泡发育、卵巢纤维化和生育恢复情况。采用体内纵向生物发光成像技术跟踪和比较移植细胞的生物分布和保留率。采用RNA测序和氧化应激和凋亡诱导条件下的体外功能分析,分析hAECs和hUC-MSCs的应激反应差异。此外,利用细胞因子阵列分析它们的分泌组。结果:在化疗诱导的POI小鼠模型中,hAECs和hUC-MSCs移植均能改善卵巢功能,表现为卵巢重量增加、排卵周期恢复、卵泡计数升高、纤维化减少、生育能力增强。体内成像显示,移植后两种细胞类型主要归巢于肺、肝和脾,信号强度随时间下降。定量分析显示,与hUC-MSCs相比,haec在体内的滞留时间明显更长。RNA测序和体外实验证实了应激条件下haec具有优越的抗氧化能力。细胞因子分析显示,hAEC-CM富含促血管生成因子,而hUC-MSC-CM含有更高水平的免疫调节因子,体外实验进一步验证了功能差异。结论:我们的研究结果表明,在化疗诱导的POI小鼠模型中,haec和hUC-MSCs都能有效地恢复卵巢功能和生育能力。然而,这两种细胞类型由于其不同的代谢动力学和旁分泌谱表现出明显的治疗优势。具体来说,与hUC-MSCs相比,haec在体内的保留率更长,这与它们增强的抗氧化能力一致。在分泌功能方面,hAECs表现出更强的促血管生成活性,而hUC-MSCs表现出更强的免疫调节作用。这些独特的特性为开发针对卵巢功能障碍的靶向治疗提供了细胞类型特异性选择的关键见解。
{"title":"Comparative evaluation of the therapeutic efficacy between human amniotic epithelial cells and human umbilical cord mesenchymal stem cells in premature ovarian insufficiency.","authors":"Qinyu Zhang, Jie Wang, Zixin Cheng, Wenjiao Cao, Qiuwan Zhang, Dongmei Lai","doi":"10.1186/s13287-025-04881-7","DOIUrl":"https://doi.org/10.1186/s13287-025-04881-7","url":null,"abstract":"<p><strong>Background: </strong>Premature ovarian insufficiency (POI) is a clinically challenging condition characterized by amenorrhea and infertility in women less than 40 years of age. Although both human amniotic epithelial cells (hAECs) and human umbilical cord mesenchymal stem cells (hUC-MSCs) have shown promise in treating POI, their comparative therapeutic efficacy and mechanisms remain poorly understood.</p><p><strong>Methods: </strong>hAECs and hUC-MSCs were isolated from human amniotic membrane and umbilical cords, respectively, and characterized using standard protocols. A chemotherapy-induced POI mouse model was established to evaluate follicular development, ovarian fibrosis, and fertility recovery after hAEC and hUC-MSC transplantation. Longitudinal in vivo bioluminescence imaging was used to track and compare the biodistribution and retention rates of the transplanted cells. RNA sequencing and in vitro functional assays under oxidative stress and apoptosis-induced conditions were employed to analyze the differential stress responses of hAECs and hUC-MSCs. Furthermore, cytokine arrays were utilized to profile their secretomes.</p><p><strong>Results: </strong>In the chemotherapy-induced POI mouse model, both hAECs and hUC-MSCs transplantation improved ovarian function, as evidenced by increased ovarian weight, restored estrous cycle, elevated follicle counts, reduced fibrosis, and enhanced fertility. In vivo imaging revealed that both cell types primarily homed to the lungs, liver, and spleen post-transplantation, with signal intensity declining over time. Quantitative analysis revealed significantly longer in vivo retention of hAECs compare to hUC-MSCs. RNA sequencing and in vitro assays confirmed the superior antioxidant capacity of hAECs under stress conditions. Cytokine profiling showed that hAEC-CM was enriched in pro-angiogenic factors, while hUC-MSC-CM contained higher levels of immunoregulatory cytokines, a functional difference further validated by in vitro experiments.</p><p><strong>Conclusion: </strong>Our findings demonstrate that both hAECs and hUC-MSCs are effective in restoring ovarian function and fertility in a chemotherapy-induced POI mouse model. However, these two cell types exhibit distinct therapeutic advantages attributable to their differential metabolic kinetics and paracrine profiles. Specifically, hAECs displayed prolonged in vivo retention rates compared to hUC-MSCs, consistent with their enhanced antioxidant capabilities. In terms of secretory function, hAECs demonstrated superior pro-angiogenic activity, while hUC-MSCs exhibited stronger immunomodulatory effects. These distinct properties provide critical insights for cell-type-specific selection in developing targeted therapies for ovarian dysfunction.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145879119","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Stem cell therapy for patients with acute myocardial infarction: a systematic review of clinical trials. 急性心肌梗死患者的干细胞治疗:临床试验的系统回顾。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-12-31 DOI: 10.1186/s13287-025-04882-6
Hadiseh Mahram, Vida Khalafi, Ali Arman, Seyed Alireza Mirhosseini, Alireza Hosseinpour, Ali Khani Jeihooni, Armin Attar

Introduction: Stem cell therapy has emerged as a potential regenerative approach for Acute myocardial infarction (AMI). Despite decades of research and advancement in acute myocardial infarction (AMI) management, translating innovative therapies from bench to bedside remains a central challenge. Nonetheless, clinical outcomes exhibit considerable variability. This review provides a comprehensive overview of the clinical landscape of stem cell therapy for AMI, specifically focusing on how variations in cell type, delivery timing, routes, and dosages can affect cell therapy efficacy.

Methods: This study is a systematic review of randomized clinical trials. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed, and the study was conducted in accordance with the Cochrane Handbook for Systematic Reviews of Interventions.

Results: After searching the relevant databases, a total of 5276 studies were assessed, and 43 trials were considered eligible for inclusion in the present systematic review. The safety and efficacy of various types of stem cells, including bone marrow-derived mononuclear cells (BM-MNCs), mesenchymal stem cells (MSCs), cardiac progenitor cells, and, more recently, induced pluripotent stem cells, have been evaluated in numerous clinical trials and meta-analyses. Among these, BM-MNCs and MSCs have been the most extensively studied. Although results vary from trial to trial and can even be contradictory, from frank failures to monumental achievements, overall, the evidence supports modest but statistically significant improvements in surrogate endpoints, such as left ventricular ejection fraction (LVEF), ventricular remodeling, and reduced infarct size.

Conclusion: We have critically reviewed how methodological approaches-especially the definitions of endpoints and clinical outcome measures-have significantly influenced the reported efficacy and direction of the field. The interpretation of clinical trial results in cell therapy for AMI is heavily impacted by the specific metrics used to define success. A key focus is distinguishing between clinical trials on patients with acute and recent myocardial infarction (which is the main focus of this review) and those with chronic ischemic or non-ischemic cardiomyopathies, as they involve different treatment strategies. Patient selection is essential for improving responses in patients with AMI. Those with a severely reduced LVEF (LVEF < 40%) and younger age tend to benefit more. Limiting the transplantation window to the first 3-7 days after AMI may improve the intervention's effectiveness.

干细胞治疗已经成为治疗急性心肌梗死(AMI)的一种潜在的再生方法。尽管在急性心肌梗死(AMI)治疗方面已经有了几十年的研究和进步,但将创新疗法从实验室应用到床边仍然是一个核心挑战。尽管如此,临床结果表现出相当大的可变性。本文综述了AMI干细胞治疗的临床概况,特别关注细胞类型、递送时间、途径和剂量的变化如何影响细胞治疗的疗效。方法:本研究是一项随机临床试验的系统综述。遵循系统评价和荟萃分析首选报告项目(PRISMA)指南,并按照Cochrane干预措施系统评价手册进行研究。结果:在检索相关数据库后,共评估了5276项研究,其中43项试验被认为符合纳入本系统评价的条件。各种类型干细胞的安全性和有效性,包括骨髓源性单核细胞(BM-MNCs)、间充质干细胞(MSCs)、心脏祖细胞,以及最近的诱导多能干细胞,已经在许多临床试验和荟萃分析中得到了评估。其中,BM-MNCs和MSCs的研究最为广泛。尽管不同试验的结果各不相同,甚至可能相互矛盾,从坦率的失败到巨大的成就,总的来说,证据支持在替代终点,如左室射血分数(LVEF)、心室重塑和梗死面积减少方面有适度但统计学上显著的改善。结论:我们批判性地回顾了方法学方法——特别是终点和临床结果测量的定义——如何显著影响该领域报告的疗效和方向。AMI细胞治疗的临床试验结果的解释在很大程度上受到用于定义成功的特定指标的影响。一个关键的焦点是区分急性和近期心肌梗死患者的临床试验(这是本综述的主要焦点)和慢性缺血性或非缺血性心肌病患者的临床试验,因为它们涉及不同的治疗策略。患者选择对于改善AMI患者的反应至关重要。LVEF严重降低的患者(LVEF)
{"title":"Stem cell therapy for patients with acute myocardial infarction: a systematic review of clinical trials.","authors":"Hadiseh Mahram, Vida Khalafi, Ali Arman, Seyed Alireza Mirhosseini, Alireza Hosseinpour, Ali Khani Jeihooni, Armin Attar","doi":"10.1186/s13287-025-04882-6","DOIUrl":"https://doi.org/10.1186/s13287-025-04882-6","url":null,"abstract":"<p><strong>Introduction: </strong>Stem cell therapy has emerged as a potential regenerative approach for Acute myocardial infarction (AMI). Despite decades of research and advancement in acute myocardial infarction (AMI) management, translating innovative therapies from bench to bedside remains a central challenge. Nonetheless, clinical outcomes exhibit considerable variability. This review provides a comprehensive overview of the clinical landscape of stem cell therapy for AMI, specifically focusing on how variations in cell type, delivery timing, routes, and dosages can affect cell therapy efficacy.</p><p><strong>Methods: </strong>This study is a systematic review of randomized clinical trials. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed, and the study was conducted in accordance with the Cochrane Handbook for Systematic Reviews of Interventions.</p><p><strong>Results: </strong>After searching the relevant databases, a total of 5276 studies were assessed, and 43 trials were considered eligible for inclusion in the present systematic review. The safety and efficacy of various types of stem cells, including bone marrow-derived mononuclear cells (BM-MNCs), mesenchymal stem cells (MSCs), cardiac progenitor cells, and, more recently, induced pluripotent stem cells, have been evaluated in numerous clinical trials and meta-analyses. Among these, BM-MNCs and MSCs have been the most extensively studied. Although results vary from trial to trial and can even be contradictory, from frank failures to monumental achievements, overall, the evidence supports modest but statistically significant improvements in surrogate endpoints, such as left ventricular ejection fraction (LVEF), ventricular remodeling, and reduced infarct size.</p><p><strong>Conclusion: </strong>We have critically reviewed how methodological approaches-especially the definitions of endpoints and clinical outcome measures-have significantly influenced the reported efficacy and direction of the field. The interpretation of clinical trial results in cell therapy for AMI is heavily impacted by the specific metrics used to define success. A key focus is distinguishing between clinical trials on patients with acute and recent myocardial infarction (which is the main focus of this review) and those with chronic ischemic or non-ischemic cardiomyopathies, as they involve different treatment strategies. Patient selection is essential for improving responses in patients with AMI. Those with a severely reduced LVEF (LVEF < 40%) and younger age tend to benefit more. Limiting the transplantation window to the first 3-7 days after AMI may improve the intervention's effectiveness.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145865574","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The treatment of acute tendon injury with small extracellular vesicles originating from TNFAIP6- ADSCs subpopoulation both in vitro and in vivo. 体外和体内研究:来自TNFAIP6- ADSCs亚群的细胞外小泡治疗急性肌腱损伤
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-12-30 DOI: 10.1186/s13287-025-04789-2
Hengchen Liu, Aodan Zhang, Manyu Shi, Jingyao Zhang, Tingting Zhang, Wenjun Lu, Mingzhao Zhang, Zenan Zhang, Yang Wu, Yibo Miao, Shuyao Wang, Limin Hou, Qingbo Cui, Zhaozhu Li

Background: Small extracellular vesicles originating from adipose-derived mesenchymal stromal cells (ADSC-sEVs) have excellent therapeutic value in acute tendon injury. However, their mechanism and effects have not been fully elucidated. This study aimed to identify the key subsets and mechanisms of action of ADSC-sEVs involved in the repair of complete tendon tear caused by acute injury.

Methods: Based on our previous research demonstrating that ADSC-sEVs improve the quality of acute tendon injury repair, the present study utilized second-generation sequencing and bioinformatics to predict the key role of the TNFAIP6- ADSC subgroup in acute tendon injury repair. We constructed different ADSC-sEVs through ADSC transfection and treated tendon stem cells for further exploration. EdU, cell scratch, and Transwell assays were used to evaluate cell proliferation and migration in vitro. Western blot and quantitative real-time polymerase chain reaction analyses were performed. Histopathological, immunohistochemical, and biomechanical testing were used for in vivo validation.

Results: TNFAIP6- ADSC-sEVs significantly improved the therapeutic effect of ADSC-sEVs on acute tendon injury, which was related to the high expression of let-7c-5p. Application of different ADSC-sEVs in vitro and in vivo identified CRCT1/JAK2/STAT3 as a key downstream signaling pathway regulated by let-7c-5p.

Conclusions: Our findings enhance the current understanding of how TNFAIP6- ADSC-sEVs exert healing properties in acute tendon injury through the let-7c-5p/CRCT1/JAK2/STAT3 signaling pathway. Furthermore, this study proposes a concept for constructing conditional ADSC-sEVs to enhance their inherent therapeutic effects.

背景:源自脂肪源性间充质基质细胞(adsc - sev)的细胞外小泡在急性肌腱损伤中具有良好的治疗价值。然而,它们的机制和作用尚未完全阐明。本研究旨在确定adsc - sev参与急性损伤完全性肌腱撕裂修复的关键亚群及其作用机制。方法:基于我们之前的研究表明ADSC- sev提高了急性肌腱损伤修复的质量,本研究利用第二代测序和生物信息学来预测TNFAIP6- ADSC亚群在急性肌腱损伤修复中的关键作用。我们通过ADSC转染构建了不同的ADSC- sev,并对肌腱干细胞进行了处理,以进一步探索。EdU、细胞划痕法和Transwell法评价细胞在体外的增殖和迁移。Western blot和实时定量聚合酶链反应分析。组织病理学、免疫组织化学和生物力学测试用于体内验证。结果:TNFAIP6- adsc - sev显著提高了adsc - sev对急性肌腱损伤的治疗效果,这与let-7c-5p的高表达有关。不同adsc - sev在体外和体内的应用鉴定出CRCT1/JAK2/STAT3是let-7c-5p调控的关键下游信号通路。结论:我们的研究结果增强了目前对TNFAIP6- adsc - sev如何通过let-7c-5p/CRCT1/JAK2/STAT3信号通路在急性肌腱损伤中发挥愈合特性的理解。此外,本研究提出了构建条件adsc - sev的概念,以增强其固有的治疗效果。
{"title":"The treatment of acute tendon injury with small extracellular vesicles originating from TNFAIP6<sup>-</sup> ADSCs subpopoulation both in vitro and in vivo.","authors":"Hengchen Liu, Aodan Zhang, Manyu Shi, Jingyao Zhang, Tingting Zhang, Wenjun Lu, Mingzhao Zhang, Zenan Zhang, Yang Wu, Yibo Miao, Shuyao Wang, Limin Hou, Qingbo Cui, Zhaozhu Li","doi":"10.1186/s13287-025-04789-2","DOIUrl":"https://doi.org/10.1186/s13287-025-04789-2","url":null,"abstract":"<p><strong>Background: </strong>Small extracellular vesicles originating from adipose-derived mesenchymal stromal cells (ADSC-sEVs) have excellent therapeutic value in acute tendon injury. However, their mechanism and effects have not been fully elucidated. This study aimed to identify the key subsets and mechanisms of action of ADSC-sEVs involved in the repair of complete tendon tear caused by acute injury.</p><p><strong>Methods: </strong>Based on our previous research demonstrating that ADSC-sEVs improve the quality of acute tendon injury repair, the present study utilized second-generation sequencing and bioinformatics to predict the key role of the TNFAIP6<sup>-</sup> ADSC subgroup in acute tendon injury repair. We constructed different ADSC-sEVs through ADSC transfection and treated tendon stem cells for further exploration. EdU, cell scratch, and Transwell assays were used to evaluate cell proliferation and migration in vitro. Western blot and quantitative real-time polymerase chain reaction analyses were performed. Histopathological, immunohistochemical, and biomechanical testing were used for in vivo validation.</p><p><strong>Results: </strong>TNFAIP6<sup>-</sup> ADSC-sEVs significantly improved the therapeutic effect of ADSC-sEVs on acute tendon injury, which was related to the high expression of let-7c-5p. Application of different ADSC-sEVs in vitro and in vivo identified CRCT1/JAK2/STAT3 as a key downstream signaling pathway regulated by let-7c-5p.</p><p><strong>Conclusions: </strong>Our findings enhance the current understanding of how TNFAIP6<sup>-</sup> ADSC-sEVs exert healing properties in acute tendon injury through the let-7c-5p/CRCT1/JAK2/STAT3 signaling pathway. Furthermore, this study proposes a concept for constructing conditional ADSC-sEVs to enhance their inherent therapeutic effects.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145865490","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Routinely collected data: a path to assess safety of human mesenchymal stromal cell transplantation in randomized controlled trials. 常规收集的数据:在随机对照试验中评估人间充质间质细胞移植安全性的途径。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-12-30 DOI: 10.1186/s13287-025-04814-4
Amir Hossein Norooznezhad

Human mesenchymal stromal cells (hMSCs) are currently at the center of interest in many randomized and non-randomized clinical trials. According to the data, the number of trials on hMSCs has increased rapidly over time. However, the safety of this treatment, despite some available evidence, remains questionable. Routinely collected data (RCD) has become a helpful approach for gathering clinical data, especially in clinical trials. This method of data collection has helped investigators overcome the limitations of randomized controlled trials (RCTs), such as ensuring long-term follow-ups. Herein, the potential role of RCD in investigating the safety of hMSCs in RCTs, particularly concerns about their possible pro-tumorigenic potential, is discussed. The patterns of recent trials in this field suggest high feasibility and the potential for using RCD for this purpose.

人间充质间质细胞(hMSCs)目前是许多随机和非随机临床试验的研究重点。数据显示,随着时间的推移,对骨髓间充质干细胞的试验数量迅速增加。然而,尽管有一些现有的证据,这种治疗的安全性仍然值得怀疑。常规收集数据(RCD)已成为收集临床数据的一种有用方法,特别是在临床试验中。这种数据收集方法帮助研究人员克服了随机对照试验(RCTs)的局限性,如确保长期随访。本文讨论了RCD在随机对照试验中研究hMSCs安全性的潜在作用,特别是对其可能的致瘤潜能的关注。最近在这一领域的试验模式表明,将RCD用于这一目的具有很高的可行性和潜力。
{"title":"Routinely collected data: a path to assess safety of human mesenchymal stromal cell transplantation in randomized controlled trials.","authors":"Amir Hossein Norooznezhad","doi":"10.1186/s13287-025-04814-4","DOIUrl":"10.1186/s13287-025-04814-4","url":null,"abstract":"<p><p>Human mesenchymal stromal cells (hMSCs) are currently at the center of interest in many randomized and non-randomized clinical trials. According to the data, the number of trials on hMSCs has increased rapidly over time. However, the safety of this treatment, despite some available evidence, remains questionable. Routinely collected data (RCD) has become a helpful approach for gathering clinical data, especially in clinical trials. This method of data collection has helped investigators overcome the limitations of randomized controlled trials (RCTs), such as ensuring long-term follow-ups. Herein, the potential role of RCD in investigating the safety of hMSCs in RCTs, particularly concerns about their possible pro-tumorigenic potential, is discussed. The patterns of recent trials in this field suggest high feasibility and the potential for using RCD for this purpose.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"701"},"PeriodicalIF":7.3,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12754955/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145865559","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Single-cell RNA sequencing identifies PD-L1 + mesenchymal stem cells with enhanced immunomodulatory capacity and alleviated the degree of ectopic new bone formation in ankylosing spondylitis. 单细胞RNA测序发现PD-L1 +间充质干细胞具有增强的免疫调节能力,减轻强直性脊柱炎异位新骨形成的程度。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-12-29 DOI: 10.1186/s13287-025-04701-y
Xiqing Luo, Liuzhong Zhou, Xianghui Wen, Jinwei Li, Dong Liu, Budian Liu, Shenghui Wen, Jieruo Gu

Backgroud: This study systematically evaluated the immunomodulatory function of PD-L1-positive mesenchymal stem cells (PD-L1(+) MSCs) using single-cell RNA sequencing (scRNA-seq) and investigated their roles in suppressing inflammation and regulating pathological bone formation in curdlan-induced SKG ankylosing spondylitis (AS) mouse models.

Methods: scRNA-seq identified MSC subpopulations with high immunomodulatory capacity and key biomarker PD-L1 for subpopulation classification. In vitro co-culture experiments were conducted to evaluate the effects of MSC subpopulations on T-cell proliferation and TNF-α levels. In vivo experiments were performed in forty-eight SKG mouse models to analyze the effects of MSC subpopulations on joint inflammation scores, T-cell subset proportions, inflammatory cytokines, histopathology, and pathological bone formation.

Results: scRNA-seq revealed significant heterogeneity in MSCs under inflammatory stimulation, with the immunomodulatory subpopulation exhibiting high expression of PD-L1 and IDO. In vitro experiments demonstrated that PD-L1(+) MSCs significantly suppressed T-cell proliferation and reduced TNF-α levels. Joint redness and swelling scores showed that the PD-L1(+) MSC group exhibited the most significant improvement in arthritis, while the IL-17Ai, PD-L1(-) MSC, and MSC groups also effectively reduced inflammation, with significantly lower scores than the model control(MC) group. Histological analysis revealed severe inflammatory cell infiltration in the MC group, while the IL-17Ai, PD-L1(+) MSC, and MSC groups exhibited reduced infiltration. Immunohistochemical analysis further confirmed these findings, with PD-L1(+) MSCs exhibiting a significant reduction in TNF-α and IL-17A-positive cells (P < 0.0001 and P < 0.01, respectively).PD-L1(+) MSCs regulated immune responses by reducing Th17 cell proportions, increasing Th2 and Treg cell proportions, and significantly lowering pro-inflammatory cytokines IFN-γ, IL-17A, and TNF-α. MicroCT analysis indicated that the PD-L1(+) MSC, MSC, and IL-17Ai group effectively suppressed pathological bone formation through immunomodulation, whereas the PD-L1(-) MSC group showed weaker effects, underscoring the importance of PD-L1 in regulating bone formation.

Conclusion: hUC-MSCs demonstrated significant therapeutic effects in the AS mouse model, particularly the PD-L1(+) MSCs, which inhibited joint inflammation and pathological new bone formation through immunomodulatory mechanisms. These findings provide valuable insights into the therapeutic mechanisms of AS treatment.

背景:本研究采用单细胞RNA测序(scRNA-seq)技术系统评估PD-L1阳性间充质干细胞(PD-L1(+) MSCs)的免疫调节功能,并研究其在curdlan诱导的SKG强直性脊柱炎(AS)小鼠模型中抑制炎症和调节病理性骨形成的作用。方法:scRNA-seq鉴定具有高免疫调节能力的MSC亚群和关键生物标志物PD-L1进行亚群分类。体外共培养实验评估MSC亚群对t细胞增殖和TNF-α水平的影响。在48只SKG小鼠模型中进行了体内实验,分析MSC亚群对关节炎症评分、t细胞亚群比例、炎症因子、组织病理学和病理性骨形成的影响。结果:scRNA-seq显示炎症刺激下MSCs的异质性显著,免疫调节亚群PD-L1和IDO高表达。体外实验表明,PD-L1(+) MSCs可显著抑制t细胞增殖,降低TNF-α水平。关节红肿评分显示,PD-L1(+) MSC组对关节炎的改善最为显著,而IL-17Ai、PD-L1(-) MSC和MSC组也能有效减轻炎症,评分明显低于模型对照组(MC)组。组织学分析显示,MC组有严重的炎症细胞浸润,而IL-17Ai、PD-L1(+) MSC和MSC组的浸润减少。免疫组织化学分析进一步证实了这些发现,PD-L1(+) MSCs显示出TNF-α和il - 17a阳性细胞的显著减少(P结论:hUC-MSCs在AS小鼠模型中显示出显著的治疗作用,特别是PD-L1(+) MSCs通过免疫调节机制抑制关节炎症和病理性新骨形成。这些发现为AS治疗机制提供了有价值的见解。
{"title":"Single-cell RNA sequencing identifies PD-L1 + mesenchymal stem cells with enhanced immunomodulatory capacity and alleviated the degree of ectopic new bone formation in ankylosing spondylitis.","authors":"Xiqing Luo, Liuzhong Zhou, Xianghui Wen, Jinwei Li, Dong Liu, Budian Liu, Shenghui Wen, Jieruo Gu","doi":"10.1186/s13287-025-04701-y","DOIUrl":"10.1186/s13287-025-04701-y","url":null,"abstract":"<p><strong>Backgroud: </strong>This study systematically evaluated the immunomodulatory function of PD-L1-positive mesenchymal stem cells (PD-L1(+) MSCs) using single-cell RNA sequencing (scRNA-seq) and investigated their roles in suppressing inflammation and regulating pathological bone formation in curdlan-induced SKG ankylosing spondylitis (AS) mouse models.</p><p><strong>Methods: </strong>scRNA-seq identified MSC subpopulations with high immunomodulatory capacity and key biomarker PD-L1 for subpopulation classification. In vitro co-culture experiments were conducted to evaluate the effects of MSC subpopulations on T-cell proliferation and TNF-α levels. In vivo experiments were performed in forty-eight SKG mouse models to analyze the effects of MSC subpopulations on joint inflammation scores, T-cell subset proportions, inflammatory cytokines, histopathology, and pathological bone formation.</p><p><strong>Results: </strong>scRNA-seq revealed significant heterogeneity in MSCs under inflammatory stimulation, with the immunomodulatory subpopulation exhibiting high expression of PD-L1 and IDO. In vitro experiments demonstrated that PD-L1(+) MSCs significantly suppressed T-cell proliferation and reduced TNF-α levels. Joint redness and swelling scores showed that the PD-L1(+) MSC group exhibited the most significant improvement in arthritis, while the IL-17Ai, PD-L1(-) MSC, and MSC groups also effectively reduced inflammation, with significantly lower scores than the model control(MC) group. Histological analysis revealed severe inflammatory cell infiltration in the MC group, while the IL-17Ai, PD-L1(+) MSC, and MSC groups exhibited reduced infiltration. Immunohistochemical analysis further confirmed these findings, with PD-L1(+) MSCs exhibiting a significant reduction in TNF-α and IL-17A-positive cells (P < 0.0001 and P < 0.01, respectively).PD-L1(+) MSCs regulated immune responses by reducing Th17 cell proportions, increasing Th2 and Treg cell proportions, and significantly lowering pro-inflammatory cytokines IFN-γ, IL-17A, and TNF-α. MicroCT analysis indicated that the PD-L1(+) MSC, MSC, and IL-17Ai group effectively suppressed pathological bone formation through immunomodulation, whereas the PD-L1(-) MSC group showed weaker effects, underscoring the importance of PD-L1 in regulating bone formation.</p><p><strong>Conclusion: </strong>hUC-MSCs demonstrated significant therapeutic effects in the AS mouse model, particularly the PD-L1(+) MSCs, which inhibited joint inflammation and pathological new bone formation through immunomodulatory mechanisms. These findings provide valuable insights into the therapeutic mechanisms of AS treatment.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"684"},"PeriodicalIF":7.3,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12750679/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145857997","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Enhancing cardiomyocyte reprogramming efficiency by targeting cellular senescence is mediated via Rb1 gene. Rb1基因介导心肌细胞衰老重编程效率提高。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-12-29 DOI: 10.1186/s13287-025-04776-7
Juntao Fang, Qiangbing Yang, Renée G C Maas, Pieter Vader, Michal Mokry, Noortje A M van den Dungen, Li Qian, Junjie Xiao, Raymond Schiffelers, Zhiyong Lei, Joost P G Sluijter

Introduction: Direct reprogramming of fibroblasts into cardiomyocytes by overexpressing cardiac transcription factors Gata4, Mef2c, and Tbx5 (GMT) is a promising way for cardiac repair, however, the low reprogramming efficiency remains a significant challenge. Cellular senescence, an irreversible cell-cycle arrest occurring in mitotic cells, has been reported to influence the efficiency of induced pluripotent stem cell (iPSC) reprogramming.

Methods: We established an inducible GMT expression system in mouse embryonic fibroblasts (MEFs) and human fetal cardiac fibroblasts (hFCFs) using the PiggyBac transposon system. RNA sequencing was performed to identify genes associated with cellular senescence during reprogramming. Selected senescence-related genes were knocked down using shRNA, and their impact on reprogramming efficiency was assessed via flow cytometry, gene expression analysis, and staining for senescence and apoptosis markers.

Results: Direct cardiac reprogramming induced cellular senescence and apoptosis, evidenced by enhanced β-Gal staining, elevated expression of senescence markers P16 and GLB1, and increased apoptosis rates. RNA sequencing and gene set enrichment analysis (GSEA) revealed significant upregulation of senescence-related genes (RB1, RBBP4, RBBP7, CBX8, and CDKN1B). Knockdown of these genes, particularly RB1, significantly enhanced reprogramming efficiency, increasing the proportion of GFP + cells in MEFs and α-actinin + cells in hFCFs. RB1 inhibition also reduced senescence marker levels and upregulated endogenous cardiac transcription factors GATA4 and MEF2C.

Conclusions: Our findings demonstrate that cellular senescence might serves as a barrier to direct cardiac reprogramming and offer novel insights into the regulatory mechanisms involved in this process.

通过过度表达心脏转录因子Gata4、Mef2c和Tbx5 (GMT)将成纤维细胞直接重编程为心肌细胞是一种很有前途的心脏修复方法,然而,低重编程效率仍然是一个重大挑战。细胞衰老是一种发生在有丝分裂细胞中不可逆的细胞周期阻滞,据报道,它会影响诱导多能干细胞(iPSC)重编程的效率。方法:利用PiggyBac转座子系统在小鼠胚胎成纤维细胞(MEFs)和人胎儿心脏成纤维细胞(hffs)中建立了GMT诱导表达系统。进行RNA测序以鉴定重编程过程中与细胞衰老相关的基因。使用shRNA敲除选定的衰老相关基因,并通过流式细胞术、基因表达分析以及衰老和凋亡标记物染色来评估其对重编程效率的影响。结果:直接心脏重编程诱导细胞衰老和凋亡,β-Gal染色增强,衰老标志物P16和GLB1表达升高,凋亡率增加。RNA测序和基因集富集分析(GSEA)显示,衰老相关基因(RB1、RBBP4、RBBP7、CBX8和CDKN1B)显著上调。敲低这些基因,尤其是RB1,可显著提高重编程效率,增加MEFs中GFP +细胞和hffs中α-肌动蛋白+细胞的比例。RB1抑制还降低了衰老标志物水平,上调了内源性心脏转录因子GATA4和MEF2C。结论:我们的研究结果表明,细胞衰老可能是直接心脏重编程的障碍,并为这一过程中涉及的调节机制提供了新的见解。
{"title":"Enhancing cardiomyocyte reprogramming efficiency by targeting cellular senescence is mediated via Rb1 gene.","authors":"Juntao Fang, Qiangbing Yang, Renée G C Maas, Pieter Vader, Michal Mokry, Noortje A M van den Dungen, Li Qian, Junjie Xiao, Raymond Schiffelers, Zhiyong Lei, Joost P G Sluijter","doi":"10.1186/s13287-025-04776-7","DOIUrl":"10.1186/s13287-025-04776-7","url":null,"abstract":"<p><strong>Introduction: </strong>Direct reprogramming of fibroblasts into cardiomyocytes by overexpressing cardiac transcription factors Gata4, Mef2c, and Tbx5 (GMT) is a promising way for cardiac repair, however, the low reprogramming efficiency remains a significant challenge. Cellular senescence, an irreversible cell-cycle arrest occurring in mitotic cells, has been reported to influence the efficiency of induced pluripotent stem cell (iPSC) reprogramming.</p><p><strong>Methods: </strong>We established an inducible GMT expression system in mouse embryonic fibroblasts (MEFs) and human fetal cardiac fibroblasts (hFCFs) using the PiggyBac transposon system. RNA sequencing was performed to identify genes associated with cellular senescence during reprogramming. Selected senescence-related genes were knocked down using shRNA, and their impact on reprogramming efficiency was assessed via flow cytometry, gene expression analysis, and staining for senescence and apoptosis markers.</p><p><strong>Results: </strong>Direct cardiac reprogramming induced cellular senescence and apoptosis, evidenced by enhanced β-Gal staining, elevated expression of senescence markers P16 and GLB1, and increased apoptosis rates. RNA sequencing and gene set enrichment analysis (GSEA) revealed significant upregulation of senescence-related genes (RB1, RBBP4, RBBP7, CBX8, and CDKN1B). Knockdown of these genes, particularly RB1, significantly enhanced reprogramming efficiency, increasing the proportion of GFP + cells in MEFs and α-actinin + cells in hFCFs. RB1 inhibition also reduced senescence marker levels and upregulated endogenous cardiac transcription factors GATA4 and MEF2C.</p><p><strong>Conclusions: </strong>Our findings demonstrate that cellular senescence might serves as a barrier to direct cardiac reprogramming and offer novel insights into the regulatory mechanisms involved in this process.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"685"},"PeriodicalIF":7.3,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12751188/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145857982","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigenetic memory as a readout of environmental susceptibility defines cell manufacturability in mesenchymal stem cell production. 表观遗传记忆作为环境敏感性的读出定义了间充质干细胞生产中的细胞可制造性。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-12-29 DOI: 10.1186/s13287-025-04887-1
Mee-Hae Kim, Masahiro Kino-Oka

Background: Mesenchymal stem cells (MSCs) are highly sensitive to fluctuations in culture process parameters (CPPs), which remain a major barrier to consistent product quality in cell manufacturing. A mechanistic understanding of how cells respond to and encode these variations is essential to enable standardization under a quality-by-design paradigm.

Methods: To address this, we propose the concept of cell manufacturability, defined as the intrinsic ability of cells to maintain their functional phenotype in response to variable inputs. Drawing inspiration from the Japanese concept of yuragi (gentle, adaptive fluctuation), we profiled histone modifications (H3K4me3 and H3K27me3) at the promoters of critical quality attribute (CQA) genes using chromatin immunoprecipitation followed by quantitative PCR (ChIP-qPCR). We established a cell potency index based on the H3K4me3/H3K27me3 ratio. Weighted principal component analysis (PCA) was applied to derive two composite indices: the Cell Susceptibility Index (CSI), indicating environmental responsiveness, and the Cell Comparability Index (CCI), representing inter-donor and process consistency.

Results: The CSI and CCI captured distinct, condition-dependent patterns. Under low-stress conditions (e.g., early passages and low seeding density), a positive correlation between CSI and CCI reflected reproducible adaptive plasticity. Conversely, high-stress cultures exhibited a strong negative correlation, which was indicative of unstable epigenetic responses. These patterns were consistently observed across different MSC sources, underscoring the generalizability of the framework.

Conclusions: This study highlights CSI and CCI as quantitative, chromatin-based metrics that offer a mechanistic basis for characterizing MSC plasticity and manufacturing robustness. Integration of these indices into the evaluation of cell manufacturability offers a predictive and scalable approach to enhance standardization and batch comparability in MSC production processes.

背景:间充质干细胞(MSCs)对培养工艺参数(CPPs)的波动高度敏感,这仍然是细胞制造中保持产品质量一致的主要障碍。对细胞如何对这些变化作出反应和编码的机制理解对于实现设计质量范式下的标准化至关重要。方法:为了解决这个问题,我们提出了细胞可制造性的概念,定义为细胞在响应变量输入时维持其功能表型的内在能力。受日本“yuragi”(温和、适应性波动)概念的启发,我们利用染色质免疫沉淀和定量PCR (ChIP-qPCR)分析了关键质量属性(CQA)基因启动子上的组蛋白修饰(H3K4me3和H3K27me3)。我们建立了基于H3K4me3/H3K27me3比值的细胞效价指数。应用加权主成分分析(PCA)得出两个复合指数:细胞敏感性指数(CSI),表示环境响应性;细胞可比性指数(CCI),表示供体间和过程一致性。结果:CSI和CCI捕获了不同的条件依赖模式。在低胁迫条件下(如传代早、播种密度低),CSI与CCI呈正相关,反映了可复制的适应可塑性。相反,高应激培养表现出强烈的负相关,这表明不稳定的表观遗传反应。这些模式在不同的MSC来源中一致观察到,强调了框架的普遍性。结论:本研究强调CSI和CCI是定量的、基于染色质的指标,为表征MSC可塑性和制造稳健性提供了机制基础。将这些指标整合到细胞可制造性评估中,提供了一种预测性和可扩展的方法,以增强MSC生产过程的标准化和批次可比性。
{"title":"Epigenetic memory as a readout of environmental susceptibility defines cell manufacturability in mesenchymal stem cell production.","authors":"Mee-Hae Kim, Masahiro Kino-Oka","doi":"10.1186/s13287-025-04887-1","DOIUrl":"https://doi.org/10.1186/s13287-025-04887-1","url":null,"abstract":"<p><strong>Background: </strong>Mesenchymal stem cells (MSCs) are highly sensitive to fluctuations in culture process parameters (CPPs), which remain a major barrier to consistent product quality in cell manufacturing. A mechanistic understanding of how cells respond to and encode these variations is essential to enable standardization under a quality-by-design paradigm.</p><p><strong>Methods: </strong>To address this, we propose the concept of cell manufacturability, defined as the intrinsic ability of cells to maintain their functional phenotype in response to variable inputs. Drawing inspiration from the Japanese concept of yuragi (gentle, adaptive fluctuation), we profiled histone modifications (H3K4me3 and H3K27me3) at the promoters of critical quality attribute (CQA) genes using chromatin immunoprecipitation followed by quantitative PCR (ChIP-qPCR). We established a cell potency index based on the H3K4me3/H3K27me3 ratio. Weighted principal component analysis (PCA) was applied to derive two composite indices: the Cell Susceptibility Index (CSI), indicating environmental responsiveness, and the Cell Comparability Index (CCI), representing inter-donor and process consistency.</p><p><strong>Results: </strong>The CSI and CCI captured distinct, condition-dependent patterns. Under low-stress conditions (e.g., early passages and low seeding density), a positive correlation between CSI and CCI reflected reproducible adaptive plasticity. Conversely, high-stress cultures exhibited a strong negative correlation, which was indicative of unstable epigenetic responses. These patterns were consistently observed across different MSC sources, underscoring the generalizability of the framework.</p><p><strong>Conclusions: </strong>This study highlights CSI and CCI as quantitative, chromatin-based metrics that offer a mechanistic basis for characterizing MSC plasticity and manufacturing robustness. Integration of these indices into the evaluation of cell manufacturability offers a predictive and scalable approach to enhance standardization and batch comparability in MSC production processes.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145858014","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Carnosic acid serves as a dual Nrf2 activator and PTEN/AKT suppressor to inhibit traumatic heterotopic ossification. 鼠尾草酸可作为Nrf2激活剂和PTEN/AKT抑制剂抑制外伤性异位骨化。
IF 7.3 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-12-29 DOI: 10.1186/s13287-025-04886-2
Donglei Wei, Dezhi Song, Hui Wang, Yuangang Su, Jiamin Liang, Jiake Xu, Jinmin Zhao, Qian Liu

Background: Heterotopic ossification (HO) pathogenesis involves ROS-driven stem cell differentiation. Carnosic acid (CA), a natural antioxidant, remains unexplored for HO.

Methods: In vitro, tendon-derived stem cells (TDSCs) were stimulated with IL-1β, and CA was used for intervention to assess its effects on differentiation and ROS production via real-time quantitative PCR (qPCR), western blotting (WB), and immunofluorescence. Additionally, a burn and Achilles tendon transection-induced mouse model of traumatic HO was established to evaluate the therapeutic potential of CA.

Results: In vitro, CA activated nuclear factor erythroid 2-related factor 2 (Nrf2) and inhibited nicotinamide adenine dinucleotide phosphate oxidase 1 (NOX1), leading to increased antioxidant enzyme activity and reduced intracellular ROS levels. CA also regulated the PTEN/AKT signaling pathway, suppressing osteogenic and chondrogenic differentiation of TDSCs. In vivo, micro-computed tomography (Micro-CT) and histological analyses demonstrated that CA activated Nrf2 and enhanced antioxidant enzyme expression, thereby inhibiting osteogenic and chondrogenic factor expression in Achilles tendon tissue and reducing HO formation.

Conclusions: CA is a novel HO therapeutic by dual targeting of oxidative stress and differentiation pathways.

背景:异位骨化(HO)的发病机制涉及ros驱动的干细胞分化。鼠尾草酸(CA)是一种天然抗氧化剂,对HO的作用仍未被发现。方法:体外用IL-1β刺激肌腱源性干细胞(tdsc),采用CA进行干预,通过实时定量PCR (qPCR)、western blotting (WB)和免疫荧光法评估其对肌腱源性干细胞(tdsc)分化和ROS生成的影响。结果:CA在体外激活核因子-红系2相关因子2 (Nrf2),抑制烟酰胺腺嘌呤二核苷酸磷酸氧化酶1 (NOX1),导致抗氧化酶活性升高,细胞内ROS水平降低。CA还调节PTEN/AKT信号通路,抑制tdsc的成骨和软骨分化。在体内,微计算机断层扫描(Micro-CT)和组织学分析表明,CA激活Nrf2,增强抗氧化酶的表达,从而抑制跟腱组织中成骨和软骨因子的表达,减少HO的形成。结论:CA是一种新型的氧化应激和分化双重靶向治疗HO的药物。
{"title":"Carnosic acid serves as a dual Nrf2 activator and PTEN/AKT suppressor to inhibit traumatic heterotopic ossification.","authors":"Donglei Wei, Dezhi Song, Hui Wang, Yuangang Su, Jiamin Liang, Jiake Xu, Jinmin Zhao, Qian Liu","doi":"10.1186/s13287-025-04886-2","DOIUrl":"https://doi.org/10.1186/s13287-025-04886-2","url":null,"abstract":"<p><strong>Background: </strong>Heterotopic ossification (HO) pathogenesis involves ROS-driven stem cell differentiation. Carnosic acid (CA), a natural antioxidant, remains unexplored for HO.</p><p><strong>Methods: </strong>In vitro, tendon-derived stem cells (TDSCs) were stimulated with IL-1β, and CA was used for intervention to assess its effects on differentiation and ROS production via real-time quantitative PCR (qPCR), western blotting (WB), and immunofluorescence. Additionally, a burn and Achilles tendon transection-induced mouse model of traumatic HO was established to evaluate the therapeutic potential of CA.</p><p><strong>Results: </strong>In vitro, CA activated nuclear factor erythroid 2-related factor 2 (Nrf2) and inhibited nicotinamide adenine dinucleotide phosphate oxidase 1 (NOX1), leading to increased antioxidant enzyme activity and reduced intracellular ROS levels. CA also regulated the PTEN/AKT signaling pathway, suppressing osteogenic and chondrogenic differentiation of TDSCs. In vivo, micro-computed tomography (Micro-CT) and histological analyses demonstrated that CA activated Nrf2 and enhanced antioxidant enzyme expression, thereby inhibiting osteogenic and chondrogenic factor expression in Achilles tendon tissue and reducing HO formation.</p><p><strong>Conclusions: </strong>CA is a novel HO therapeutic by dual targeting of oxidative stress and differentiation pathways.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":" ","pages":""},"PeriodicalIF":7.3,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145858016","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Stem Cell Research & Therapy
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1