首页 > 最新文献

STEM CELLS最新文献

英文 中文
Co-culture of RhoA-overexpressed Microtia Chondrocytes and Adipose-Derived Stem Cells in the Construction of Tissue-engineered Ear-shaped Cartilage. RhoA过表达小耳软骨细胞与脂肪来源干细胞共培养构建组织工程耳形软骨
IF 5.2 2区 医学 Q1 Medicine Pub Date : 2024-04-13 DOI: 10.1093/stmcls/sxae026
Yi Wu, Jian Wang, Xiu Li, Kang Wang, Zonglin Huang, Qian Wang, Xin Fu, Haiyue Jiang, Bo Pan, Ran Xiao
Microtia is a congenital auricle dysplasia with a high incidence and tissue engineering technology provides a promising strategy to reconstruct auricles. We previously described that the engineered cartilage constructed from microtia chondrocytes exhibited inferior levels of biochemical and biomechanical properties, which was proposed to be resulted from the decreased migration ability of microtia chondrocytes. In the current study, we found that Rho GTPase members were deficient in microtia chondrocytes. By overexpressing RhoA, Rac1 and CDC42, respectively, we further demonstrated that RhoA took great responsibility for the decreased migration ability of microtia chondrocytes. Moreover, we constructed PGA/PLA scaffold-based cartilages to verify the chondrogenic ability of RhoA overexpressed microtia chondrocytes, and the results showed that overexpressing RhoA was of limited help to improve the quality of microtia chondrocyte engineered cartilage. However, co-culture of ADSCs significantly improved the biochemical and biomechanical property of engineered cartilage. Especially, co-culture of RhoA overexpressed microtia chondrocytes and ADSCs produced an excellent effect on the wet weight, cartilage-specific extracellular matrix and biomechanical property of engineered cartilage. Furthermore, we presented that co-culture of RhoA overexpressed microtia chondrocytes and ADSCs combined with human ear-shaped PGA/PLA scaffold and titanium alloy stent fabricated by CAD/CAM and 3D printing technology effectively constructed and maintained auricle structure in vivo. Collectively, our results provide evidence for the essential role of RhoA in microtia chondrocytes and a developed strategy for the construction of patient-specific tissue-engineered auricular cartilage.
小耳症是一种先天性耳廓发育不良,发病率很高,组织工程技术为重建耳廓提供了一种前景广阔的策略。我们曾描述过,由小耳畸形软骨细胞构建的工程软骨表现出较低水平的生物化学和生物力学特性,这被认为是小耳畸形软骨细胞迁移能力下降的结果。在本研究中,我们发现小耳软骨细胞中缺乏 Rho GTPase 成员。通过分别过表达 RhoA、Rac1 和 CDC42,我们进一步证明 RhoA 对小耳畸形软骨细胞迁移能力的下降负有重要责任。此外,我们还构建了基于PGA/PLA支架的软骨,以验证过表达RhoA的小鼠软骨细胞的软骨生成能力,结果表明过表达RhoA对提高小鼠软骨细胞工程软骨的质量帮助有限。然而,与 ADSCs 共同培养能明显改善工程软骨的生化和生物力学特性。特别是,RhoA 过度表达的微蒂亚软骨细胞与 ADSCs 共同培养对工程软骨的湿重、软骨特异性细胞外基质和生物力学性质产生了很好的影响。此外,我们还发现,RhoA 过度表达的小耳软骨细胞和 ADSCs 与通过 CAD/CAM 和 3D 打印技术制作的人耳形 PGA/PLA 支架和钛合金支架联合培养,能有效地在体内构建和维持耳廓结构。总之,我们的研究结果证明了RhoA在小耳软骨细胞中的重要作用,并为构建患者特异性组织工程耳廓软骨提供了一种新策略。
{"title":"Co-culture of RhoA-overexpressed Microtia Chondrocytes and Adipose-Derived Stem Cells in the Construction of Tissue-engineered Ear-shaped Cartilage.","authors":"Yi Wu, Jian Wang, Xiu Li, Kang Wang, Zonglin Huang, Qian Wang, Xin Fu, Haiyue Jiang, Bo Pan, Ran Xiao","doi":"10.1093/stmcls/sxae026","DOIUrl":"https://doi.org/10.1093/stmcls/sxae026","url":null,"abstract":"Microtia is a congenital auricle dysplasia with a high incidence and tissue engineering technology provides a promising strategy to reconstruct auricles. We previously described that the engineered cartilage constructed from microtia chondrocytes exhibited inferior levels of biochemical and biomechanical properties, which was proposed to be resulted from the decreased migration ability of microtia chondrocytes. In the current study, we found that Rho GTPase members were deficient in microtia chondrocytes. By overexpressing RhoA, Rac1 and CDC42, respectively, we further demonstrated that RhoA took great responsibility for the decreased migration ability of microtia chondrocytes. Moreover, we constructed PGA/PLA scaffold-based cartilages to verify the chondrogenic ability of RhoA overexpressed microtia chondrocytes, and the results showed that overexpressing RhoA was of limited help to improve the quality of microtia chondrocyte engineered cartilage. However, co-culture of ADSCs significantly improved the biochemical and biomechanical property of engineered cartilage. Especially, co-culture of RhoA overexpressed microtia chondrocytes and ADSCs produced an excellent effect on the wet weight, cartilage-specific extracellular matrix and biomechanical property of engineered cartilage. Furthermore, we presented that co-culture of RhoA overexpressed microtia chondrocytes and ADSCs combined with human ear-shaped PGA/PLA scaffold and titanium alloy stent fabricated by CAD/CAM and 3D printing technology effectively constructed and maintained auricle structure in vivo. Collectively, our results provide evidence for the essential role of RhoA in microtia chondrocytes and a developed strategy for the construction of patient-specific tissue-engineered auricular cartilage.","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-04-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140708241","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 epigenetic landscape in intestinal stem cells and its deregulation in colorectal cancer. 肠道干细胞的表观遗传景观及其在结直肠癌中的失调。
IF 5.2 2区 医学 Q1 Medicine Pub Date : 2024-04-10 DOI: 10.1093/stmcls/sxae027
Axelle Larue, Yaser Atlasi
Epigenetic mechanisms play a pivotal role in controlling gene expression and cellular plasticity in both normal physiology and pathophysiological conditions. These mechanisms are particularly important in the regulation of stem cell self-renewal and differentiation, both in embryonic development and within adult tissues. A prime example of this finely tuned epigenetic control is observed in the gastrointestinal lining, where the small intestine undergoes renewal approximately every 3-5 days. How various epigenetic mechanisms modulate chromatin functions in intestinal stem cells (ISCs) is currently an active area of research. In this review, we discuss the main epigenetic mechanisms that control ISC differentiation under normal homeostasis. Furthermore, we explore the dysregulation of these mechanisms in the context of colorectal cancer (CRC) development. By outlining the main epigenetic mechanisms contributing to CRC, we highlight the recent therapeutics development and future directions for colorectal cancer research.
表观遗传机制在控制正常生理和病理生理条件下的基因表达和细胞可塑性方面发挥着关键作用。在胚胎发育和成人组织中,这些机制对干细胞自我更新和分化的调控尤为重要。胃肠黏膜就是这种精细调整的表观遗传控制的一个典型例子,小肠大约每3-5天进行一次更新。各种表观遗传机制如何调节肠道干细胞(ISCs)的染色质功能,是目前一个活跃的研究领域。在这篇综述中,我们将讨论在正常平衡状态下控制肠干细胞分化的主要表观遗传机制。此外,我们还探讨了这些机制在结直肠癌(CRC)发展过程中的失调。通过概述导致 CRC 的主要表观遗传机制,我们强调了最近的治疗方法发展和结直肠癌研究的未来方向。
{"title":"The epigenetic landscape in intestinal stem cells and its deregulation in colorectal cancer.","authors":"Axelle Larue, Yaser Atlasi","doi":"10.1093/stmcls/sxae027","DOIUrl":"https://doi.org/10.1093/stmcls/sxae027","url":null,"abstract":"Epigenetic mechanisms play a pivotal role in controlling gene expression and cellular plasticity in both normal physiology and pathophysiological conditions. These mechanisms are particularly important in the regulation of stem cell self-renewal and differentiation, both in embryonic development and within adult tissues. A prime example of this finely tuned epigenetic control is observed in the gastrointestinal lining, where the small intestine undergoes renewal approximately every 3-5 days. How various epigenetic mechanisms modulate chromatin functions in intestinal stem cells (ISCs) is currently an active area of research. In this review, we discuss the main epigenetic mechanisms that control ISC differentiation under normal homeostasis. Furthermore, we explore the dysregulation of these mechanisms in the context of colorectal cancer (CRC) development. By outlining the main epigenetic mechanisms contributing to CRC, we highlight the recent therapeutics development and future directions for colorectal cancer research.","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140716318","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
Modulating the Mesenchymal Stromal Cell Microenvironment Alters Exosome RNA Content and Ligament Healing Capacity. 调节间充质基质细胞微环境可改变外泌体 RNA 含量和韧带愈合能力
IF 5.2 2区 医学 Q1 Medicine Pub Date : 2024-04-10 DOI: 10.1093/stmcls/sxae028
C. Chamberlain, Archana Prabahar, J. Kink, Erika Mueller, Yiyao Li, Stephanie Yopp, C. Capitini, P. Hematti, William L. Murphy, Ray Vanderby, Peng Jiang
Although mesenchymal stromal cell (MSC) based therapies hold promise in regenerative medicine, their clinical application remains challenging due to issues such as immunocompatibility. MSC-derived exosomes are a promising off-the-shelf therapy for promoting wound healing in a cell-free manner. However, the potential to customize the content of MSC-exosomes, and understanding how such modifications influence exosome effects on tissue regeneration remain underexplored. In this study, we used an in vitro system to compare the priming of human MSCs by two inflammatory inducers TNF-α and CRX-527 (a highly potent synthetic TLR4 agonist that can be used as a vaccine adjuvant or to induce anti-tumor immunity) on exosome molecular cargo, as well as on an in vivo rat ligament injury model to validate exosome potency. Different microenvironmental stimuli used to prime MSCs in vitro affected their exosomal microRNAs and mRNAs, influencing ligament healing. Exosomes derived from untreated MSCs significantly enhance the mechanical properties of healing ligaments, in contrast to those obtained from MSCs primed with inflammation-inducers, which not only fail to provide any improvement but also potentially deteriorate the mechanical properties. Additionally, a link was identified between altered exosomal microRNA levels and expression changes in microRNA targets in ligaments. These findings elucidate the nuanced interplay between MSCs, their exosomes, and tissue regeneration.
尽管基于间充质基质细胞(MSC)的疗法在再生医学中大有可为,但由于免疫相容性等问题,其临床应用仍面临挑战。间充质干细胞衍生的外泌体是一种很有前景的现成疗法,能以无细胞方式促进伤口愈合。然而,定制间充质干细胞外泌体含量的潜力以及了解这种修饰如何影响外泌体对组织再生的作用仍未得到充分探索。在这项研究中,我们利用体外系统比较了两种炎症诱导剂TNF-α和CRX-527(一种高效的合成TLR4激动剂,可用作疫苗佐剂或诱导抗肿瘤免疫)对人间充质干细胞的引诱作用,以及体内大鼠韧带损伤模型对外泌体分子货物的引诱作用,以验证外泌体的效力。用于体外培养间充质干细胞的不同微环境刺激会影响其外泌体microRNA和mRNA,从而影响韧带愈合。从未经处理的间充质干细胞中提取的外泌体可显著提高愈合韧带的机械性能,而从使用炎症诱导剂的间充质干细胞中提取的外泌体则与之相反,不仅不能改善韧带的机械性能,还有可能恶化韧带的机械性能。此外,研究还发现了外泌体microRNA水平的改变与韧带中microRNA靶点表达变化之间的联系。这些发现阐明了间充质干细胞、其外泌体和组织再生之间微妙的相互作用。
{"title":"Modulating the Mesenchymal Stromal Cell Microenvironment Alters Exosome RNA Content and Ligament Healing Capacity.","authors":"C. Chamberlain, Archana Prabahar, J. Kink, Erika Mueller, Yiyao Li, Stephanie Yopp, C. Capitini, P. Hematti, William L. Murphy, Ray Vanderby, Peng Jiang","doi":"10.1093/stmcls/sxae028","DOIUrl":"https://doi.org/10.1093/stmcls/sxae028","url":null,"abstract":"Although mesenchymal stromal cell (MSC) based therapies hold promise in regenerative medicine, their clinical application remains challenging due to issues such as immunocompatibility. MSC-derived exosomes are a promising off-the-shelf therapy for promoting wound healing in a cell-free manner. However, the potential to customize the content of MSC-exosomes, and understanding how such modifications influence exosome effects on tissue regeneration remain underexplored. In this study, we used an in vitro system to compare the priming of human MSCs by two inflammatory inducers TNF-α and CRX-527 (a highly potent synthetic TLR4 agonist that can be used as a vaccine adjuvant or to induce anti-tumor immunity) on exosome molecular cargo, as well as on an in vivo rat ligament injury model to validate exosome potency. Different microenvironmental stimuli used to prime MSCs in vitro affected their exosomal microRNAs and mRNAs, influencing ligament healing. Exosomes derived from untreated MSCs significantly enhance the mechanical properties of healing ligaments, in contrast to those obtained from MSCs primed with inflammation-inducers, which not only fail to provide any improvement but also potentially deteriorate the mechanical properties. Additionally, a link was identified between altered exosomal microRNA levels and expression changes in microRNA targets in ligaments. These findings elucidate the nuanced interplay between MSCs, their exosomes, and tissue regeneration.","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-04-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140719340","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: 1
Cell-cycle-specific autoencoding improves cluster analysis of cycling cardiomyocytes. 细胞周期特异性自身编码改进了循环心肌细胞的聚类分析。
IF 5.2 2区 医学 Q1 Medicine Pub Date : 2024-04-08 DOI: 10.1093/stmcls/sxae016
Thanh Nguyen, Yuji Nakada, Yalin Wu, Jianli Zhao, Daniel J. Garry, Hesham A Sadek, J. Zhang
BACKGROUNDOur previous analyses of cardiomyocyte single-nucleus RNA sequencing (snRNAseq) data from the hearts of fetal pigs and pigs that underwent apical resection surgery on postnatal day (P) 1 (ARP1), myocardial infarction (MI) surgery on P28 (MIP28), both ARP1 and MIP28 (ARP1MIP28), or controls (no surgical procedure or CTL) identified 10 cardiomyocyte subpopulations (clusters), one of which appeared to be primed to proliferate in response to MI. However, the clusters composed of primarily proliferating cardiomyocytes still contained non-cycling cells, and we were unable to distinguish between cardiomyocytes in different phases of the cell cycle. Here, we improved the precision of our assessments by conducting similar analyses with snRNAseq data for only the 1646 genes included under the Gene Ontology term "cell cycle."METHODSTwo cardiac snRNAseq datasets, one from mice (GEO dataset number GSE130699) and one from pigs (GEO dataset number GSE185289), were evaluated via our cell-cycle-specific analytical pipeline. Cycling cells were identified via the co-expression of five proliferation markers (AURKB, MKI67, INCENP, CDCA8, and BIRC5).RESULTSThe cell-cycle-specific autoencoder (CSA) algorithm identified seven cardiomyocyte clusters in mouse hearts (mCM1 and mCM3-mCM8), including one prominent cluster of cycling cardiomyocytes in animals that underwent MI or Sham surgery on P1. Five cardiomyocyte clusters (pCM1, pCM3-pCM6) were identified in pig hearts, two of which (pCM1 and pCM4) displayed evidence of cell cycle activity; pCM4 was found primarily in hearts from fetal pigs, while pCM1 comprised a small proportion of cardiomyocytes in both fetal hearts and hearts from ARP1MIP28 pigs during the two weeks after MI induction, but was nearly undetectable in all other experimental groups and at all other time points. Furthermore, pseudotime trajectory analysis of snRNAseq data from fetal pig cardiomyocytes identified a pathway that began at pCM3, passed through pCM2, and ended at pCM1, and whereas pCM3 was enriched for the expression of a cell-cycle activator that regulates the G1/S phase transition (Cyclin D2), pCM2 was enriched for an S-phase regulator (CCNE2), and pCM1 was enriched for the expression of a gene that regulates the G2M phase transition and mitosis (Cyclin B2). We also identified four transcription factors (E2F8, FOXM1, GLI3, and RAD51) that were more abundantly expressed in cardiomyocytes from regenerative mouse hearts than from nonregenerative mouse hearts, from the hearts of fetal pigs than from CTL pig hearts, and from ARP1MIP28 pig hearts than from MIP28 pig hearts during the two weeks after MI induction.CONCLUSIONSThe CSA algorithm improved the precision of our assessments of cell cycle activity in cardiomyocyte subpopulations and enabled us to identify a trajectory across three clusters that appeared to track the onset and progression of cell-cycle activity in cardiomyocytes from fetal pigs.
背景我们先前对胎儿猪和在出生后第 1 天(P)接受心尖切除手术(ARP1)、在 P28 天接受心肌梗死(MI)手术(MIP28)、ARP1 和 MIP28(ARP1MIP28)或对照组(无手术或 CTL)的心脏的心肌细胞单核 RNA 测序(snRNAseq)数据进行了分析、心肌梗塞(MI)手术(MIP28)、ARP1 和 MIP28(ARP1MIP28)或对照组(无手术过程或 CTL)的心肌细胞序列(snRNAseq)数据确定了 10 个心肌细胞亚群(簇),其中一个亚群似乎对 MI 有增殖反应。然而,主要由增殖的心肌细胞组成的细胞簇仍包含非循环细胞,我们无法区分处于细胞周期不同阶段的心肌细胞。方法通过我们的细胞周期特异性分析管道评估了两个心脏 snRNAseq 数据集,一个来自小鼠(GEO 数据集编号 GSE130699),另一个来自猪(GEO 数据集编号 GSE185289)。结果细胞周期特异性自动编码器(CSA)算法确定了小鼠心脏中的七个心肌细胞簇(mCM1 和 mCM3-mCM8),包括在 P1 期接受 MI 或 Sham 手术的动物中的一个突出的循环心肌细胞簇。在猪心中发现了五个心肌细胞簇(pCM1、pCM3-pCM6),其中两个(pCM1 和 pCM4)显示出细胞周期活动的证据;pCM4 主要存在于胎儿猪的心脏中,而在诱导 MI 后的两周内,pCM1 在胎儿心脏和 ARP1MIP28 猪的心脏中都占一小部分,但在所有其他实验组和所有其他时间点几乎检测不到。此外,通过对胎儿猪心肌细胞的 snRNAseq 数据进行伪时间轨迹分析,发现了一条始于 pCM3、经过 pCM2、止于 pCM1 的通路、pCM3富集了一种细胞周期激活因子的表达,该因子可调控 G1/S 期转变(细胞周期蛋白 D2);pCM2 富集了一种 S 期调控因子(CCNE2);pCM1 则富集了一种基因的表达,该基因可调控 G2M 期转变和有丝分裂(细胞周期蛋白 B2)。我们还发现了四个转录因子(E2F8、FOXM1、GLI3 和 RAD51),在诱导 MI 后的两周内,这些因子在再生小鼠心脏中的表达量高于非再生小鼠心脏,在胎猪心脏中的表达量高于 CTL 猪心脏,在 ARP1MIP28 猪心脏中的表达量高于 MIP28 猪心脏。结论CSA 算法提高了我们对心肌细胞亚群细胞周期活动评估的精确度,并使我们能够识别出三个群组的轨迹,这三个群组似乎追踪了胎猪心肌细胞细胞周期活动的开始和进展。
{"title":"Cell-cycle-specific autoencoding improves cluster analysis of cycling cardiomyocytes.","authors":"Thanh Nguyen, Yuji Nakada, Yalin Wu, Jianli Zhao, Daniel J. Garry, Hesham A Sadek, J. Zhang","doi":"10.1093/stmcls/sxae016","DOIUrl":"https://doi.org/10.1093/stmcls/sxae016","url":null,"abstract":"BACKGROUND\u0000Our previous analyses of cardiomyocyte single-nucleus RNA sequencing (snRNAseq) data from the hearts of fetal pigs and pigs that underwent apical resection surgery on postnatal day (P) 1 (ARP1), myocardial infarction (MI) surgery on P28 (MIP28), both ARP1 and MIP28 (ARP1MIP28), or controls (no surgical procedure or CTL) identified 10 cardiomyocyte subpopulations (clusters), one of which appeared to be primed to proliferate in response to MI. However, the clusters composed of primarily proliferating cardiomyocytes still contained non-cycling cells, and we were unable to distinguish between cardiomyocytes in different phases of the cell cycle. Here, we improved the precision of our assessments by conducting similar analyses with snRNAseq data for only the 1646 genes included under the Gene Ontology term \"cell cycle.\"\u0000\u0000\u0000METHODS\u0000Two cardiac snRNAseq datasets, one from mice (GEO dataset number GSE130699) and one from pigs (GEO dataset number GSE185289), were evaluated via our cell-cycle-specific analytical pipeline. Cycling cells were identified via the co-expression of five proliferation markers (AURKB, MKI67, INCENP, CDCA8, and BIRC5).\u0000\u0000\u0000RESULTS\u0000The cell-cycle-specific autoencoder (CSA) algorithm identified seven cardiomyocyte clusters in mouse hearts (mCM1 and mCM3-mCM8), including one prominent cluster of cycling cardiomyocytes in animals that underwent MI or Sham surgery on P1. Five cardiomyocyte clusters (pCM1, pCM3-pCM6) were identified in pig hearts, two of which (pCM1 and pCM4) displayed evidence of cell cycle activity; pCM4 was found primarily in hearts from fetal pigs, while pCM1 comprised a small proportion of cardiomyocytes in both fetal hearts and hearts from ARP1MIP28 pigs during the two weeks after MI induction, but was nearly undetectable in all other experimental groups and at all other time points. Furthermore, pseudotime trajectory analysis of snRNAseq data from fetal pig cardiomyocytes identified a pathway that began at pCM3, passed through pCM2, and ended at pCM1, and whereas pCM3 was enriched for the expression of a cell-cycle activator that regulates the G1/S phase transition (Cyclin D2), pCM2 was enriched for an S-phase regulator (CCNE2), and pCM1 was enriched for the expression of a gene that regulates the G2M phase transition and mitosis (Cyclin B2). We also identified four transcription factors (E2F8, FOXM1, GLI3, and RAD51) that were more abundantly expressed in cardiomyocytes from regenerative mouse hearts than from nonregenerative mouse hearts, from the hearts of fetal pigs than from CTL pig hearts, and from ARP1MIP28 pig hearts than from MIP28 pig hearts during the two weeks after MI induction.\u0000\u0000\u0000CONCLUSIONS\u0000The CSA algorithm improved the precision of our assessments of cell cycle activity in cardiomyocyte subpopulations and enabled us to identify a trajectory across three clusters that appeared to track the onset and progression of cell-cycle activity in cardiomyocytes from fetal pigs.","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-04-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140728563","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
Human iPSC-Based Model of COPD to Investigate Disease Mechanisms, Predict SARS-COV-2 Outcome, and Test Preventive Immunotherapy. 基于人类 iPSC 的慢性阻塞性肺病模型,用于研究疾病机制、预测 SARS-COV-2 的结果和测试预防性免疫疗法。
IF 5.2 2区 医学 Q1 Medicine Pub Date : 2024-03-14 DOI: 10.1093/stmcls/sxad094
Rania Dagher, Aigul Moldobaeva, Elise Gubbins, Sydney Clark, Mia Madel Alfajaro, Craig B Wilen, Finn Hawkins, Xiaotao Qu, Chia Chien Chiang, Yang Li, Lori Clarke, Yasuhiro Ikeda, Charles Brown, Roland Kolbeck, Qin Ma, Mauricio Rojas, Jonathan L Koff, Mahboobe Ghaedi

Chronic inflammation and dysregulated repair mechanisms after epithelial damage have been implicated in chronic obstructive pulmonary disease (COPD). However, the lack of ex vivo-models that accurately reflect multicellular lung tissue hinders our understanding of epithelial-mesenchymal interactions in COPD. Through a combination of transcriptomic and proteomic approaches applied to a sophisticated in vitro iPSC-alveolosphere with fibroblasts model, epithelial-mesenchymal crosstalk was explored in COPD and following SARS-CoV-2 infection. These experiments profiled dynamic changes at single-cell level of the SARS-CoV-2-infected alveolar niche that unveiled the complexity of aberrant inflammatory responses, mitochondrial dysfunction, and cell death in COPD, which provides deeper insights into the accentuated tissue damage/inflammation/remodeling observed in patients with SARS-CoV-2 infection. Importantly, this 3D system allowed for the evaluation of ACE2-neutralizing antibodies and confirmed the potency of this therapy to prevent SARS-CoV-2 infection in the alveolar niche. Thus, iPSC-alveolosphere cultured with fibroblasts provides a promising model to investigate disease-specific mechanisms and to develop novel therapeutics.

慢性阻塞性肺病与上皮损伤后的慢性炎症和修复机制失调有关。然而,缺乏能准确反映多细胞肺组织的体外模型阻碍了我们对慢性阻塞性肺病中上皮-间充质相互作用的了解。通过将转录组学和蛋白质组学方法结合应用于复杂的体外 iPSC-肺泡与成纤维细胞模型,探索了慢性阻塞性肺病和 SARS-CoV-2 感染后上皮-间充质相互作用。这些实验剖析了受 SARS-CoV-2 感染的肺泡生态位单细胞水平的动态变化,揭示了慢性阻塞性肺病中异常炎症反应、线粒体功能障碍和细胞死亡的复杂性,为深入了解 SARS-CoV-2 感染患者中观察到的组织损伤/炎症/重塑现象提供了依据。重要的是,这种三维系统允许对 ACE2 中和抗体进行评估,并证实了这种疗法在肺泡龛中预防 SARS-CoV-2 感染的有效性。因此,与成纤维细胞一起培养的iPSC-肺泡层为研究疾病特异性机制和开发新型疗法提供了一个前景广阔的模型。
{"title":"Human iPSC-Based Model of COPD to Investigate Disease Mechanisms, Predict SARS-COV-2 Outcome, and Test Preventive Immunotherapy.","authors":"Rania Dagher, Aigul Moldobaeva, Elise Gubbins, Sydney Clark, Mia Madel Alfajaro, Craig B Wilen, Finn Hawkins, Xiaotao Qu, Chia Chien Chiang, Yang Li, Lori Clarke, Yasuhiro Ikeda, Charles Brown, Roland Kolbeck, Qin Ma, Mauricio Rojas, Jonathan L Koff, Mahboobe Ghaedi","doi":"10.1093/stmcls/sxad094","DOIUrl":"10.1093/stmcls/sxad094","url":null,"abstract":"<p><p>Chronic inflammation and dysregulated repair mechanisms after epithelial damage have been implicated in chronic obstructive pulmonary disease (COPD). However, the lack of ex vivo-models that accurately reflect multicellular lung tissue hinders our understanding of epithelial-mesenchymal interactions in COPD. Through a combination of transcriptomic and proteomic approaches applied to a sophisticated in vitro iPSC-alveolosphere with fibroblasts model, epithelial-mesenchymal crosstalk was explored in COPD and following SARS-CoV-2 infection. These experiments profiled dynamic changes at single-cell level of the SARS-CoV-2-infected alveolar niche that unveiled the complexity of aberrant inflammatory responses, mitochondrial dysfunction, and cell death in COPD, which provides deeper insights into the accentuated tissue damage/inflammation/remodeling observed in patients with SARS-CoV-2 infection. Importantly, this 3D system allowed for the evaluation of ACE2-neutralizing antibodies and confirmed the potency of this therapy to prevent SARS-CoV-2 infection in the alveolar niche. Thus, iPSC-alveolosphere cultured with fibroblasts provides a promising model to investigate disease-specific mechanisms and to develop novel therapeutics.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139105617","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
Cancer Stem Cells as a Therapeutic Target: Current Clinical Development and Future Prospective. 作为治疗靶点的癌症干细胞:当前的临床发展和未来展望。
IF 5.2 2区 医学 Q1 Medicine Pub Date : 2024-03-14 DOI: 10.1093/stmcls/sxad092
Alex Philchenkov, Anna Dubrovska

The key role of cancer stem cells (CSCs) in tumor development and therapy resistance makes them essential biomarkers and therapeutic targets. Numerous agents targeting CSCs, either as monotherapy or as part of combination therapy, are currently being tested in clinical trials to treat solid tumors and hematologic malignancies. Data from ongoing and future clinical trials testing novel approaches to target tumor stemness-related biomarkers and pathways may pave the way for further clinical development of CSC-targeted treatments and CSC-guided selection of therapeutic regimens. In this concise review, we discuss recent progress in developing CSC-directed treatment approaches, focusing on clinical trials testing CSC-directed therapies. We also consider the further development of CSC-assay-guided patient stratification and treatment personalization.

癌症干细胞(CSCs)在肿瘤发生发展和耐药性治疗中起着关键作用,因此成为重要的生物标志物和治疗靶点。目前,许多靶向CSCs的药物,无论是作为单一疗法还是作为联合疗法的一部分,都在治疗实体瘤和血液恶性肿瘤的临床试验中接受测试。正在进行的临床试验和其他临床试验中测试靶向肿瘤干细胞相关生物标志物和通路的新方法的数据,可能为进一步临床开发针对CSC的治疗方法和CSC指导下的治疗方案选择铺平道路。在这篇简明综述中,我们讨论了开发造血干细胞导向治疗方法的最新进展,重点关注测试造血干细胞导向疗法的临床试验。我们还考虑了在造血干细胞检测指导下对患者进行分层和个性化治疗的进一步发展。
{"title":"Cancer Stem Cells as a Therapeutic Target: Current Clinical Development and Future Prospective.","authors":"Alex Philchenkov, Anna Dubrovska","doi":"10.1093/stmcls/sxad092","DOIUrl":"10.1093/stmcls/sxad092","url":null,"abstract":"<p><p>The key role of cancer stem cells (CSCs) in tumor development and therapy resistance makes them essential biomarkers and therapeutic targets. Numerous agents targeting CSCs, either as monotherapy or as part of combination therapy, are currently being tested in clinical trials to treat solid tumors and hematologic malignancies. Data from ongoing and future clinical trials testing novel approaches to target tumor stemness-related biomarkers and pathways may pave the way for further clinical development of CSC-targeted treatments and CSC-guided selection of therapeutic regimens. In this concise review, we discuss recent progress in developing CSC-directed treatment approaches, focusing on clinical trials testing CSC-directed therapies. We also consider the further development of CSC-assay-guided patient stratification and treatment personalization.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138794456","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
Aberrant Lipid Metabolic Signatures in Acute Myeloid Leukemia. 急性髓性白血病中异常的脂质代谢特征
IF 5.2 2区 医学 Q1 Medicine Pub Date : 2024-03-14 DOI: 10.1093/stmcls/sxad095
Pooja Singh, Roopak Murali, Sri Gayathri Shanmugam, Steve Thomas, Julius Scott, Sudha Warrier, Frank Arfuso, Arun Dharmarajan, Rajesh Kumar Gandhirajan

Leukemogenesis is a complex process that involves multiple stages of mutation in either hematopoietic stem or progenitor cells, leading to cancer development over time. Acute myeloid leukemia (AML) is an aggressive malignancy that affects myeloid cells. The major disease burden is caused by immature blast cells, which are eliminated using conventional chemotherapies. Unfortunately, relapse is a leading cause of death in AML patients, with 30%-80% experiencing it within 2 years of initial treatment. The dominant cause of relapse in leukemia is the presence of therapy-resistant leukemic stem cells (LSCs). These cells express genes related to stemness that are frequently difficult to eradicate and tend to survive standard treatments. Studies have demonstrated that by targeting the metabolic pathways of LSCs, it is possible to improve outcomes and extend the survival of those afflicted by leukemia. The overwhelming evidence suggests that lipid metabolism is reprogrammed in LSCs, leading to an increase in fatty acid uptake and de novo lipogenesis. Genes regulating this process also play a crucial role in therapy evasion. In this concise review, we summarize the lipid metabolism in normal hematopoietic cells, AML blast cells, and AML LSCs. We also compare the lipid metabolic signatures in de novo versus therapy-resistant AML blast and LSCs. We further discuss the metabolic switches, cellular crosstalk, potential targets, and inhibitors of lipid metabolism that could alleviate treatment resistance and relapse.

白血病的发生是一个复杂的过程,涉及造血干细胞或祖细胞多个阶段的突变,随着时间的推移导致癌症的发展。急性髓系白血病(AML)是一种影响髓系细胞的侵袭性恶性肿瘤。主要的疾病负担是由未成熟的爆炸细胞造成的,传统的化疗方法可以消除这些细胞。不幸的是,复发是导致急性髓细胞白血病患者死亡的主要原因,其中 30% 至 80% 的患者在接受初始治疗后两年内复发。白血病复发的主要原因是存在抗药性白血病干细胞(LSCs)。这些细胞表达与干性有关的基因,通常难以根除,而且往往能在标准治疗中存活下来。研究表明,通过针对白血病干细胞的代谢途径,有可能改善治疗效果,延长白血病患者的生存期。大量证据表明,脂质代谢在 LSCs 中发生了重编程,导致脂肪酸摄取和新脂肪生成增加。调节这一过程的基因在逃避治疗方面也起着至关重要的作用。在这篇简明综述中,我们总结了正常造血细胞、急性髓细胞性白血病爆破细胞和急性髓细胞性白血病造血干细胞的脂质代谢。我们还比较了新生与耐药急性髓细胞白血病细胞和白血病细胞间充质干细胞的脂质代谢特征。我们还进一步讨论了脂质代谢的代谢开关、细胞交叉对话、潜在靶点和抑制剂,它们可以减轻治疗耐药性和复发。
{"title":"Aberrant Lipid Metabolic Signatures in Acute Myeloid Leukemia.","authors":"Pooja Singh, Roopak Murali, Sri Gayathri Shanmugam, Steve Thomas, Julius Scott, Sudha Warrier, Frank Arfuso, Arun Dharmarajan, Rajesh Kumar Gandhirajan","doi":"10.1093/stmcls/sxad095","DOIUrl":"10.1093/stmcls/sxad095","url":null,"abstract":"<p><p>Leukemogenesis is a complex process that involves multiple stages of mutation in either hematopoietic stem or progenitor cells, leading to cancer development over time. Acute myeloid leukemia (AML) is an aggressive malignancy that affects myeloid cells. The major disease burden is caused by immature blast cells, which are eliminated using conventional chemotherapies. Unfortunately, relapse is a leading cause of death in AML patients, with 30%-80% experiencing it within 2 years of initial treatment. The dominant cause of relapse in leukemia is the presence of therapy-resistant leukemic stem cells (LSCs). These cells express genes related to stemness that are frequently difficult to eradicate and tend to survive standard treatments. Studies have demonstrated that by targeting the metabolic pathways of LSCs, it is possible to improve outcomes and extend the survival of those afflicted by leukemia. The overwhelming evidence suggests that lipid metabolism is reprogrammed in LSCs, leading to an increase in fatty acid uptake and de novo lipogenesis. Genes regulating this process also play a crucial role in therapy evasion. In this concise review, we summarize the lipid metabolism in normal hematopoietic cells, AML blast cells, and AML LSCs. We also compare the lipid metabolic signatures in de novo versus therapy-resistant AML blast and LSCs. We further discuss the metabolic switches, cellular crosstalk, potential targets, and inhibitors of lipid metabolism that could alleviate treatment resistance and relapse.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139085276","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
Therapeutic Effects of Hematopoietic Stem Cell Derived From Gene-Edited Mice on β654-Thalassemia. 基因编辑小鼠的造血干细胞对β654-地中海贫血症的治疗效果。
IF 5.2 2区 医学 Q1 Medicine Pub Date : 2024-03-14 DOI: 10.1093/stmcls/sxad096
Dan Lu, Xiuli Gong, Xinbing Guo, Yanwen Chen, Yiwen Zhu, Yudan Fang, Qin Cai, Miao Xu, Hua Yang, Dali Li, Yitao Zeng, Fanyi Zeng

β-thalassemia is an inherited blood disease caused by reduced or inadequate β-globin synthesis due to β-globin gene mutation. Our previous study developed a gene-edited mice model (β654-ER mice) by CRISPR/Cas9-mediated genome editing, targeting both the βIVS2-654 (C > T) mutation site and the 3' splicing acceptor site at 579 and corrected abnormal β-globin mRNA splicing in the β654-thalassemia mice. Herein, we further explored the therapeutic effect of the hematopoietic stem cells (HSCs) from β654-ER mice on β-thalassemia by consecutive HSC transplantation. The results indicated that HSC transplantation derived from gene-edited mice can significantly improve the survival rate of mice after lethal radiation doses and effectively achieve hematopoietic reconstruction and long-term hematopoiesis. Clinical symptoms, including hematologic parameters and tissue pathology of transplanted recipients, were significantly improved compared to the non-transplanted β654 mice. The therapeutic effect of gene-edited HSC transplantation demonstrated no significant difference in hematological parameters and tissue pathology compared with wild-type mouse-derived HSCs. Our data revealed that HSC transplantation from gene-edited mice completely recovered the β-thalassemia phenotype. Our study systematically investigated the therapeutic effect of HSCs derived from β654-ER mice on β-thalassemia and further confirmed the efficacy of our gene-editing approach. Altogether, it provided a reference and primary experimental data for the clinical usage of such gene-edited HSCs in the future.

β地中海贫血是一种遗传性血液病,由β-球蛋白基因突变导致β-球蛋白合成减少或不足引起。我们之前的研究通过CRISPR/Cas9介导的基因组编辑,针对βIVS2-654(C>T)突变位点和579处的3'剪接接受位点,建立了基因编辑小鼠模型(β654-ER小鼠),并纠正了β654-地中海贫血小鼠β-球蛋白mRNA剪接异常。在此,我们进一步探讨了β654-ER小鼠造血干细胞连续移植对β地中海贫血的治疗效果。结果表明,基因编辑小鼠的造血干细胞移植能显著提高致死剂量辐射后小鼠的存活率,有效实现造血重建和长期造血。与未移植的β654小鼠相比,移植受体的血液学指标和组织病理学等临床症状明显改善。与野生型小鼠来源的造血干细胞相比,基因编辑造血干细胞移植的治疗效果在血液学指标和组织病理学方面无明显差异。我们的数据显示,基因编辑小鼠的造血干细胞移植完全恢复了β地中海贫血的表型。我们的研究系统地探讨了β654-ER小鼠来源的造血干细胞对β地中海贫血的治疗效果,并进一步证实了我们的基因编辑方法的有效性。总之,该研究为今后此类基因编辑造血干细胞的临床应用提供了参考和原始实验数据。
{"title":"Therapeutic Effects of Hematopoietic Stem Cell Derived From Gene-Edited Mice on β654-Thalassemia.","authors":"Dan Lu, Xiuli Gong, Xinbing Guo, Yanwen Chen, Yiwen Zhu, Yudan Fang, Qin Cai, Miao Xu, Hua Yang, Dali Li, Yitao Zeng, Fanyi Zeng","doi":"10.1093/stmcls/sxad096","DOIUrl":"10.1093/stmcls/sxad096","url":null,"abstract":"<p><p>β-thalassemia is an inherited blood disease caused by reduced or inadequate β-globin synthesis due to β-globin gene mutation. Our previous study developed a gene-edited mice model (β654-ER mice) by CRISPR/Cas9-mediated genome editing, targeting both the βIVS2-654 (C > T) mutation site and the 3' splicing acceptor site at 579 and corrected abnormal β-globin mRNA splicing in the β654-thalassemia mice. Herein, we further explored the therapeutic effect of the hematopoietic stem cells (HSCs) from β654-ER mice on β-thalassemia by consecutive HSC transplantation. The results indicated that HSC transplantation derived from gene-edited mice can significantly improve the survival rate of mice after lethal radiation doses and effectively achieve hematopoietic reconstruction and long-term hematopoiesis. Clinical symptoms, including hematologic parameters and tissue pathology of transplanted recipients, were significantly improved compared to the non-transplanted β654 mice. The therapeutic effect of gene-edited HSC transplantation demonstrated no significant difference in hematological parameters and tissue pathology compared with wild-type mouse-derived HSCs. Our data revealed that HSC transplantation from gene-edited mice completely recovered the β-thalassemia phenotype. Our study systematically investigated the therapeutic effect of HSCs derived from β654-ER mice on β-thalassemia and further confirmed the efficacy of our gene-editing approach. Altogether, it provided a reference and primary experimental data for the clinical usage of such gene-edited HSCs in the future.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138883779","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
Application Potential of Extracellular Vesicles Derived From Mesenchymal Stem Cells in Renal Diseases. 间充质干细胞细胞外囊泡在肾脏疾病中的应用潜力。
IF 5.2 2区 医学 Q1 Medicine Pub Date : 2024-03-14 DOI: 10.1093/stmcls/sxad089
Enhui Li, Jia Xu, Ning Liu, Qi Xiong, Weiwei Zhang, Yizi Gong, Linlin Zhang, Yikai He, Huipeng Ge, Xiangcheng Xiao

The high prevalence and complex etiology of renal diseases already impose a heavy disease burden on patients and society. In certain kidney diseases such as acute kidney injury and chronic kidney disease, current treatments are limited to slowing rather than stabilizing or reversing disease progression. Therefore, it is crucial to study the pathological mechanisms of kidney disease and discover new therapeutic targets and effective therapeutic drugs. As cell-free therapeutic strategies are continually being developed, extracellular vesicles derived from mesenchymal stem cells (MSC-EVs) have emerged as a hot topic for research in the field of renal diseases. Studies have demonstrated that MSC-EVs not only reproduce the therapeutic effects of MSCs but also localize to damaged kidney tissue. Compared to MSCs, MSC-EVs have several advantages, including ease of preservation, low immunogenicity, an inability to directly form tumors, and ease of artificial modification. Exploring the detailed mechanisms of MSC-EVs by developing standardized culture, isolation, purification, and drug delivery strategies will help facilitate their clinical application in kidney diseases. Here, we provide a comprehensive overview of studies about MSC-EVs in kidney diseases and discuss their limitations at the human nephrology level.

肾脏疾病的高患病率和复杂的病因已经给患者和社会造成了沉重的疾病负担。在某些肾脏疾病,如急性肾损伤(AKI)和慢性肾脏疾病(CKD),目前的治疗仅限于减缓而不是稳定或逆转疾病进展。因此,研究肾脏疾病的病理机制,发现新的治疗靶点和有效的治疗药物至关重要。随着无细胞治疗策略的不断发展,间充质干细胞(msc - ev)衍生的细胞外囊泡已成为肾脏疾病领域的研究热点。研究表明,间充质干细胞- ev不仅能复制间充质干细胞的治疗效果,还能定位于受损的肾组织。与间充质干细胞相比,间充质干细胞- ev具有易于保存、免疫原性低、不能直接形成肿瘤和易于人工修饰等优点。通过建立规范化培养、分离、纯化和给药策略,探索msc - ev的具体机制,将有助于其在肾脏疾病中的临床应用。在这里,我们提供了关于msc - ev在肾脏疾病中的研究的全面概述,并讨论了它们在人类肾脏学水平上的局限性。
{"title":"Application Potential of Extracellular Vesicles Derived From Mesenchymal Stem Cells in Renal Diseases.","authors":"Enhui Li, Jia Xu, Ning Liu, Qi Xiong, Weiwei Zhang, Yizi Gong, Linlin Zhang, Yikai He, Huipeng Ge, Xiangcheng Xiao","doi":"10.1093/stmcls/sxad089","DOIUrl":"10.1093/stmcls/sxad089","url":null,"abstract":"<p><p>The high prevalence and complex etiology of renal diseases already impose a heavy disease burden on patients and society. In certain kidney diseases such as acute kidney injury and chronic kidney disease, current treatments are limited to slowing rather than stabilizing or reversing disease progression. Therefore, it is crucial to study the pathological mechanisms of kidney disease and discover new therapeutic targets and effective therapeutic drugs. As cell-free therapeutic strategies are continually being developed, extracellular vesicles derived from mesenchymal stem cells (MSC-EVs) have emerged as a hot topic for research in the field of renal diseases. Studies have demonstrated that MSC-EVs not only reproduce the therapeutic effects of MSCs but also localize to damaged kidney tissue. Compared to MSCs, MSC-EVs have several advantages, including ease of preservation, low immunogenicity, an inability to directly form tumors, and ease of artificial modification. Exploring the detailed mechanisms of MSC-EVs by developing standardized culture, isolation, purification, and drug delivery strategies will help facilitate their clinical application in kidney diseases. Here, we provide a comprehensive overview of studies about MSC-EVs in kidney diseases and discuss their limitations at the human nephrology level.</p>","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138456744","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
Correction to: Nicotinamide Riboside Modulates HIF-1 Signaling to Maintain and Enhance Odontoblastic Differentiation in Human Dental Pulp Stem Cells. 更正:烟酰胺核苷调节 HIF-1 信号以维持和增强人牙髓干细胞的牙髓分化。
IF 5.2 2区 医学 Q1 Medicine Pub Date : 2024-03-14 DOI: 10.1093/stmcls/sxad093
{"title":"Correction to: Nicotinamide Riboside Modulates HIF-1 Signaling to Maintain and Enhance Odontoblastic Differentiation in Human Dental Pulp Stem Cells.","authors":"","doi":"10.1093/stmcls/sxad093","DOIUrl":"10.1093/stmcls/sxad093","url":null,"abstract":"","PeriodicalId":231,"journal":{"name":"STEM CELLS","volume":null,"pages":null},"PeriodicalIF":5.2,"publicationDate":"2024-03-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139072831","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 CELLS
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:481959085
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