首页 > 最新文献

Stem Cell Research & Therapy最新文献

英文 中文
Correction: Radiochemotherapy-induced DNA repair promotes the biogenesis of gastric cancer stem cells. 更正:放化疗诱导的DNA修复促进了胃癌干细胞的生物生成。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-10-10 DOI: 10.1186/s13287-024-03984-x
Yu Lu, Xiaobo Zhang
{"title":"Correction: Radiochemotherapy-induced DNA repair promotes the biogenesis of gastric cancer stem cells.","authors":"Yu Lu, Xiaobo Zhang","doi":"10.1186/s13287-024-03984-x","DOIUrl":"10.1186/s13287-024-03984-x","url":null,"abstract":"","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"358"},"PeriodicalIF":7.1,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11465732/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142401376","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
Correction: Mir-340-3p-modified bone marrow mesenchymal stem cell-derived exosomes inhibit ferroptosis through METTL3-mediated m6A modification of HMOX1 to promote recovery of injured rat uterus. 更正:Mir-340-3p修饰的骨髓间充质干细胞衍生外泌体通过METTL3介导的HMOX1 m6A修饰抑制铁变态反应,促进受伤大鼠子宫的恢复。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-10-10 DOI: 10.1186/s13287-024-03985-w
Bang Xiao, Yiqing Zhu, Meng Liu, Meiting Chen, Chao Huang, Dabing Xu, Fang Wang, Shuhan Sun, Jinfeng Huang, Ningxia Sun, Fu Yang
{"title":"Correction: Mir-340-3p-modified bone marrow mesenchymal stem cell-derived exosomes inhibit ferroptosis through METTL3-mediated m6A modification of HMOX1 to promote recovery of injured rat uterus.","authors":"Bang Xiao, Yiqing Zhu, Meng Liu, Meiting Chen, Chao Huang, Dabing Xu, Fang Wang, Shuhan Sun, Jinfeng Huang, Ningxia Sun, Fu Yang","doi":"10.1186/s13287-024-03985-w","DOIUrl":"10.1186/s13287-024-03985-w","url":null,"abstract":"","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"357"},"PeriodicalIF":7.1,"publicationDate":"2024-10-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11465922/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142401375","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
The therapeutic use of clonal neural stem cells in experimental Parkinson´s disease. 克隆神经干细胞在实验性帕金森病中的治疗应用。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-10-09 DOI: 10.1186/s13287-024-03965-0
Anna Nelke, Silvia García-López, Javier R Caso, Marta P Pereira

Background: Parkinson´s disease (PD), the second most common neurodegenerative disease in the world, is characterized by the death or impairment of dopaminergic neurons (DAn) in the substantia nigra pars compacta and dopamine depletion in the striatum. Currently, there is no cure for PD, and treatments only help to reduce the symptoms of the disease, and do not repair or replace the DAn damaged or lost in PD. Cell replacement therapy (CRT) seeks to relieve both pathological and symptomatic PD manifestations and has been shown to have beneficial effects in experimental PD models as well as in PD patients, but an apt cell line to be used in the treatment of PD has yet to be established. The purpose of this study was to examine the effects of the transplantation of hVM1 clone 32 cells, a bankable line of human neural stem cells (hNSCs), in a PD mouse model at four months post-transplant.

Methods: Adult (five month-old) C57BL/6JRccHsd male mice were injected with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and subsequently transplanted with hVM1 clone 32 cells, or buffer, in the left striatum. Four months post-transplant, behavioral effects were explored using the open field and paw print tests, and histological analyses were performed.

Results: Transplantation of hVM1 clone 32 cells rescued dopaminergic nigrostriatal populations in adult Parkinsonian mice. Motor and neurological deterioration were observed in buffer-treated mice, the latter of which had a tendency to improve in hNSC-transplanted mice. Detection of mast cell migration to the superficial cervical lymph nodes in cell-transplanted mice denoted a peripheral effect. Transplantation of hNSCs also rescued neuroblast neurogenesis in the subgranular zone, which was correlated with dopaminergic recovery and is indicative of local recovery mechanisms.

Conclusions: In this proof-of-concept study, the transplantation of hVM1 clone 32 cells provided neuroprotection in adult Parkinsonian mice by restoring the dopaminergic nigrostriatal pathway and hippocampal neurogenesis, demonstrating the efficacy of cell replacement therapy as a treatment for PD.

背景:帕金森病(Parkinson´s disease,PD)是世界上第二大常见的神经退行性疾病,其特征是黑质中的多巴胺能神经元(dopaminergic neurons,DAn)死亡或受损,纹状体中的多巴胺耗竭。目前,帕金森氏症尚无根治方法,治疗方法只能帮助减轻疾病症状,并不能修复或替代因帕金森氏症而受损或丢失的多巴胺能神经元。细胞替代疗法(CRT)旨在缓解帕金森病的病理和症状表现,已被证明对实验性帕金森病模型和帕金森病患者有益,但用于治疗帕金森病的合适细胞系尚未建立。本研究的目的是研究移植hVM1克隆32细胞(一种可存活的人类神经干细胞(hNSCs)品系)对移植后四个月的帕金森病小鼠模型的影响:成年(五个月大)C57BL/6JRccHsd雄性小鼠注射1-甲基-4-苯基-1,2,3,6-四氢吡啶,随后在左侧纹状体移植hVM1克隆32细胞或缓冲液。移植后四个月,使用开阔地和爪印测试探讨了行为效应,并进行了组织学分析:结果:移植hVM1克隆32细胞可挽救成年帕金森小鼠的多巴胺能黑质群。在缓冲液处理的小鼠中观察到运动和神经功能衰退,而在移植了hNSC的小鼠中,后者有改善的趋势。在细胞移植小鼠的颈浅淋巴结中检测到肥大细胞迁移,这表明了外周效应。移植hNSCs还能挽救粒细胞下区的神经母细胞神经发生,这与多巴胺能的恢复相关,表明了局部恢复机制:在这项概念验证研究中,移植 hVM1 克隆 32 细胞通过恢复多巴胺能黑质通路和海马神经发生,为成年帕金森病小鼠提供了神经保护,证明了细胞替代疗法治疗帕金森病的有效性。
{"title":"The therapeutic use of clonal neural stem cells in experimental Parkinson´s disease.","authors":"Anna Nelke, Silvia García-López, Javier R Caso, Marta P Pereira","doi":"10.1186/s13287-024-03965-0","DOIUrl":"10.1186/s13287-024-03965-0","url":null,"abstract":"<p><strong>Background: </strong>Parkinson´s disease (PD), the second most common neurodegenerative disease in the world, is characterized by the death or impairment of dopaminergic neurons (DAn) in the substantia nigra pars compacta and dopamine depletion in the striatum. Currently, there is no cure for PD, and treatments only help to reduce the symptoms of the disease, and do not repair or replace the DAn damaged or lost in PD. Cell replacement therapy (CRT) seeks to relieve both pathological and symptomatic PD manifestations and has been shown to have beneficial effects in experimental PD models as well as in PD patients, but an apt cell line to be used in the treatment of PD has yet to be established. The purpose of this study was to examine the effects of the transplantation of hVM1 clone 32 cells, a bankable line of human neural stem cells (hNSCs), in a PD mouse model at four months post-transplant.</p><p><strong>Methods: </strong>Adult (five month-old) C57BL/6JRccHsd male mice were injected with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine and subsequently transplanted with hVM1 clone 32 cells, or buffer, in the left striatum. Four months post-transplant, behavioral effects were explored using the open field and paw print tests, and histological analyses were performed.</p><p><strong>Results: </strong>Transplantation of hVM1 clone 32 cells rescued dopaminergic nigrostriatal populations in adult Parkinsonian mice. Motor and neurological deterioration were observed in buffer-treated mice, the latter of which had a tendency to improve in hNSC-transplanted mice. Detection of mast cell migration to the superficial cervical lymph nodes in cell-transplanted mice denoted a peripheral effect. Transplantation of hNSCs also rescued neuroblast neurogenesis in the subgranular zone, which was correlated with dopaminergic recovery and is indicative of local recovery mechanisms.</p><p><strong>Conclusions: </strong>In this proof-of-concept study, the transplantation of hVM1 clone 32 cells provided neuroprotection in adult Parkinsonian mice by restoring the dopaminergic nigrostriatal pathway and hippocampal neurogenesis, demonstrating the efficacy of cell replacement therapy as a treatment for PD.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"356"},"PeriodicalIF":7.1,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11465761/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142393467","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
Exosomes in nanomedicine: a promising cell-free therapeutic intervention in burn wounds. 纳米医学中的外泌体:一种治疗烧伤伤口的前景广阔的无细胞疗法。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-10-09 DOI: 10.1186/s13287-024-03970-3
Tasaduq Manzoor, Nida Farooq, Arushi Sharma, Parvaiz A Shiekh, Amreena Hassan, Lateef Ahmad Dar, Junaid Nazir, Meena Godha, Faheem A Sheikh, Mudasir Bashir Gugjoo, Sahar Saleem, Syed Mudasir Ahmad

Burn injuries are serious injuries that have a big impact on a person's health and can even cause death. Incurring severe burns can incite an immune response and inflammation within the body, alongside metabolic changes. It is of utmost importance to grasp the fact that the effects of the burn injury extend beyond the body, affecting the mind and overall well-being. Burn injuries cause long-lasting changes that need to be taken care of in order to improve their quality of life. The intricate process of skin regeneration at the site of a burn wound involves a complex and dynamic interplay among diverse cells, growth factors, nerves, and blood vessels. Exciting opportunities have arisen in the field of stem cells and regenerative medicine, allowing us to explore the development of cell-free-based alternatives that can aid in the treatment of burn injuries. These cell-free-based therapies have emerged as a promising facet within regenerative medicine. Exosomes, also referred to as naturally occurring nanoparticles, are small endosome-derived vesicles that facilitate the delivery of molecular cargo between the cells, thus allowing intercellular communication. The knowledge gained in this field has continued to support their therapeutic potential, particularly in the domains of wound healing and tissue regeneration. Notably, exosomes derived from mesenchymal stem cells (MSCs) can be safely administered in the system, which is then adeptly uptaken and internalized by fibroblasts/epithelial cells, subsequently accelerating essential processes such as migration, proliferation, and collagen synthesis. Furthermore, exosomes released by immune cells, specifically macrophages, possess the capability to modulate inflammation and effectively diminish it in adjacent cells. Exosomes also act as carriers when integrated with a scaffold, leading to scarless healing of cutaneous wounds. This comprehensive review examines the role of exosomes in burn wound healing and their potential utility in regeneration and repair.

烧伤是一种严重伤害,对人的健康有很大影响,甚至可能导致死亡。严重烧伤会引发免疫反应和体内炎症,同时还会导致新陈代谢发生变化。最重要的是,烧伤的影响超出身体范围,会影响心理和整体健康。烧伤造成的变化是长期的,需要加以护理,以提高他们的生活质量。烧伤部位的皮肤再生过程错综复杂,涉及各种细胞、生长因子、神经和血管之间复杂而动态的相互作用。干细胞和再生医学领域出现了令人兴奋的机遇,使我们能够探索开发无细胞替代品,帮助治疗烧伤。这些不含细胞的疗法已成为再生医学中前景广阔的一个方面。外泌体(Exosomes)也被称为天然纳米颗粒,是一种源自内泌体的小囊泡,有助于在细胞间传递分子货物,从而实现细胞间的交流。这一领域的知识不断支持其治疗潜力,特别是在伤口愈合和组织再生领域。值得注意的是,从间充质干细胞(MSCs)中提取的外泌体可在系统中安全给药,然后被成纤维细胞/上皮细胞吸收和内化,随后加速迁移、增殖和胶原蛋白合成等重要过程。此外,免疫细胞(尤其是巨噬细胞)释放的外泌体具有调节炎症的能力,能有效减轻邻近细胞的炎症。外泌体与支架结合后还可作为载体,实现皮肤伤口的无疤痕愈合。本综述探讨了外泌体在烧伤伤口愈合中的作用及其在再生和修复中的潜在用途。
{"title":"Exosomes in nanomedicine: a promising cell-free therapeutic intervention in burn wounds.","authors":"Tasaduq Manzoor, Nida Farooq, Arushi Sharma, Parvaiz A Shiekh, Amreena Hassan, Lateef Ahmad Dar, Junaid Nazir, Meena Godha, Faheem A Sheikh, Mudasir Bashir Gugjoo, Sahar Saleem, Syed Mudasir Ahmad","doi":"10.1186/s13287-024-03970-3","DOIUrl":"10.1186/s13287-024-03970-3","url":null,"abstract":"<p><p>Burn injuries are serious injuries that have a big impact on a person's health and can even cause death. Incurring severe burns can incite an immune response and inflammation within the body, alongside metabolic changes. It is of utmost importance to grasp the fact that the effects of the burn injury extend beyond the body, affecting the mind and overall well-being. Burn injuries cause long-lasting changes that need to be taken care of in order to improve their quality of life. The intricate process of skin regeneration at the site of a burn wound involves a complex and dynamic interplay among diverse cells, growth factors, nerves, and blood vessels. Exciting opportunities have arisen in the field of stem cells and regenerative medicine, allowing us to explore the development of cell-free-based alternatives that can aid in the treatment of burn injuries. These cell-free-based therapies have emerged as a promising facet within regenerative medicine. Exosomes, also referred to as naturally occurring nanoparticles, are small endosome-derived vesicles that facilitate the delivery of molecular cargo between the cells, thus allowing intercellular communication. The knowledge gained in this field has continued to support their therapeutic potential, particularly in the domains of wound healing and tissue regeneration. Notably, exosomes derived from mesenchymal stem cells (MSCs) can be safely administered in the system, which is then adeptly uptaken and internalized by fibroblasts/epithelial cells, subsequently accelerating essential processes such as migration, proliferation, and collagen synthesis. Furthermore, exosomes released by immune cells, specifically macrophages, possess the capability to modulate inflammation and effectively diminish it in adjacent cells. Exosomes also act as carriers when integrated with a scaffold, leading to scarless healing of cutaneous wounds. This comprehensive review examines the role of exosomes in burn wound healing and their potential utility in regeneration and repair.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"355"},"PeriodicalIF":7.1,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11462792/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142393457","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
SLAMF8 regulates osteogenesis and adipogenesis of bone marrow mesenchymal stem cells via S100A6/Wnt/β-catenin signaling pathway. SLAMF8通过S100A6/Wnt/β-catenin信号通路调控骨髓间充质干细胞的成骨和成脂。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-10-08 DOI: 10.1186/s13287-024-03964-1
Yibo Wang, Kai Hang, Xiaoyong Wu, Li Ying, Zhongxiang Wang, Zemin Ling, Hao Hu, Zhijun Pan, Xuenong Zou

Background: The inflammatory microenvironment plays an essential role in bone healing after fracture. The signaling lymphocytic activation molecule family (SLAMF) members deeply participate in inflammatory response and make a vast difference.

Methods: We identified SLAMF8 in GEO datasets (GSE129165 and GSE176086) and co-expression analyses were performed to define the relationships between SLAMF8 and osteogenesis relative genes (RUNX2 and COL1A1). In vitro, we established SLAMF8 knockdown and overexpression mouse bone marrow mesenchymal stem cells (mBMSCs) lines. qPCR, Western blot, ALP staining, ARS staining, Oil Red O staining and Immunofluorescence analyses were performed to investigate the effect of SLAMF8 in mBMSCs osteogenesis and adipogenesis. In vivo, mice femoral fracture model was performed to explore the function of SLAMF8.

Results: SLAMF8 knockdown significantly suppressed the expression of osteogenesis relative genes (RUNX2, SP7 and COL1A1), ALP activity and mineral deposition, but increased the expression of adipogenesis relative genes (PPARγ and C/EBPα). Additionally, SLAMF8 overexpression had the opposite effects. The role SLAMF8 played in mBMSCs osteogenic and adipogenic differentiation were through S100A6 and Wnt/β-Catenin signaling pathway. Moreover, SLAMF8 overexpression mBMSCs promoted the healing of femoral fracture.

Conclusions: SLAMF8 promotes osteogenesis and inhibits adipogenesis of mBMSCs via S100A6 and Wnt/β-Catenin signaling pathway. SLAMF8 overexpression mBMSCs effectively accelerate the healing of femoral fracture in mice.

背景:炎症微环境在骨折后骨愈合过程中发挥着至关重要的作用。信号淋巴细胞活化分子家族(SLAMF)成员深度参与了炎症反应并发挥了巨大作用:我们在 GEO 数据集(GSE129165 和 GSE176086)中发现了 SLAMF8,并进行了共表达分析,以确定 SLAMF8 与成骨相关基因(RUNX2 和 COL1A1)之间的关系。在体外,我们建立了SLAMF8基因敲除和过表达的小鼠骨髓间充质干细胞(mBMSCs)株,并进行了qPCR、Western印迹、ALP染色、ARS染色、油红O染色和免疫荧光分析,以研究SLAMF8在mBMSCs成骨和成脂过程中的作用。在体内,通过小鼠股骨骨折模型探讨SLAMF8的功能:结果:SLAMF8敲除可明显抑制成骨相关基因(RUNX2、SP7和COL1A1)、ALP活性和矿物质沉积的表达,但可增加脂肪生成相关基因(PPARγ和C/EBPα)的表达。此外,SLAMF8的过表达具有相反的效果。SLAMF8通过S100A6和Wnt/β-Catenin信号通路在mBMSCs成骨和成脂分化中发挥作用。此外,SLAMF8过表达的mBMSCs能促进股骨骨折的愈合:结论:SLAMF8通过S100A6和Wnt/β-Catenin信号通路促进mBMSCs成骨并抑制脂肪生成。过表达 SLAMF8 的 mBMSCs 能有效加速小鼠股骨骨折的愈合。
{"title":"SLAMF8 regulates osteogenesis and adipogenesis of bone marrow mesenchymal stem cells via S100A6/Wnt/β-catenin signaling pathway.","authors":"Yibo Wang, Kai Hang, Xiaoyong Wu, Li Ying, Zhongxiang Wang, Zemin Ling, Hao Hu, Zhijun Pan, Xuenong Zou","doi":"10.1186/s13287-024-03964-1","DOIUrl":"10.1186/s13287-024-03964-1","url":null,"abstract":"<p><strong>Background: </strong>The inflammatory microenvironment plays an essential role in bone healing after fracture. The signaling lymphocytic activation molecule family (SLAMF) members deeply participate in inflammatory response and make a vast difference.</p><p><strong>Methods: </strong>We identified SLAMF8 in GEO datasets (GSE129165 and GSE176086) and co-expression analyses were performed to define the relationships between SLAMF8 and osteogenesis relative genes (RUNX2 and COL1A1). In vitro, we established SLAMF8 knockdown and overexpression mouse bone marrow mesenchymal stem cells (mBMSCs) lines. qPCR, Western blot, ALP staining, ARS staining, Oil Red O staining and Immunofluorescence analyses were performed to investigate the effect of SLAMF8 in mBMSCs osteogenesis and adipogenesis. In vivo, mice femoral fracture model was performed to explore the function of SLAMF8.</p><p><strong>Results: </strong>SLAMF8 knockdown significantly suppressed the expression of osteogenesis relative genes (RUNX2, SP7 and COL1A1), ALP activity and mineral deposition, but increased the expression of adipogenesis relative genes (PPARγ and C/EBPα). Additionally, SLAMF8 overexpression had the opposite effects. The role SLAMF8 played in mBMSCs osteogenic and adipogenic differentiation were through S100A6 and Wnt/β-Catenin signaling pathway. Moreover, SLAMF8 overexpression mBMSCs promoted the healing of femoral fracture.</p><p><strong>Conclusions: </strong>SLAMF8 promotes osteogenesis and inhibits adipogenesis of mBMSCs via S100A6 and Wnt/β-Catenin signaling pathway. SLAMF8 overexpression mBMSCs effectively accelerate the healing of femoral fracture in mice.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"349"},"PeriodicalIF":7.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11462740/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142393465","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
Serum metabonomics reveal the effectiveness of human placental mesenchymal stem cell therapy for primary sclerosing cholangitis. 血清代谢组学揭示了人胎盘间充质干细胞疗法对原发性硬化性胆管炎的疗效。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-10-08 DOI: 10.1186/s13287-024-03967-y
Yingduo Yu, Qigu Yao, Deying Chen, Zhehua Zhang, Qiaoling Pan, Jiong Yu, Hongcui Cao, Liang Li, Lanjuan Li

Background: The metabolic patterns of human placental-derived mesenchymal stem cell (hP-MSC) treatment for primary sclerosing cholangitis (PSC) remain unclear, and therapeutic effects significantly vary due to individual differences. Therefore, it is crucial to investigate the serological response to hP-MSC transplantation through small molecular metabolites and identify easily detectable markers for efficacy evaluation.

Methods: Using Mdr2-/- mice as a PSC model and Mdr2+/+ mice as controls, the efficacy of hP-MSC treatment was assessed based on liver pathology, liver enzymes, and inflammatory factors. Serum samples were collected for 12C-/13C-dansylation and DmPA labeling LC-MS analysis to investigate changes in metabolic pathways after hP-MSC treatment. Key metabolites and regulatory enzymes were validated by qRT-PCR and Western blotting. Potential biomarkers of hP-MSC efficacy were identified through correlation analysis and machine learning.

Results: Collectively, the results of the liver histology, serum liver enzyme levels, and inflammatory factors supported the therapeutic efficacy of hP-MSC treatment. Based on significant differences, 41 differentially expressed metabolites were initially identified; these were enriched in bile acid, lipid, and hydroxyproline metabolism. After treatment, bile acid transport was accelerated, whereas bile acid production was reduced; unsaturated fatty acid synthesis was upregulated overall, with increased FADS2 and elongase expression and enhanced fatty acid β-oxidation; hepatic proline 4-hydroxylase expression was decreased, leading to reduced hydroxyproline production. Correlation analysis of liver enzymes and metabolites, combined with time trends, identified eight potential biomarkers: 2-aminomuconate semialdehyde, L-1-pyrroline-3-hydroxy-5-carboxylic acid, L-isoglutamine, and maleamic acid were more abundant in model mice but decreased after hP-MSC treatment. Conversely, 15-methylpalmitic, eicosenoic, nonadecanoic, and octadecanoic acids were less abundant in model mice but increased after hP-MSC treatment.

Conclusions: This study revealed metabolic regulatory changes in PSC model mice after hP-MSC treatment and identified eight promising biomarkers, providing preclinical evidence to support therapeutic applications of hP-MSC.

背景:人胎盘间充质干细胞(hP-MSC)治疗原发性硬化性胆管炎(PSC)的代谢模式仍不清楚,而且由于个体差异,治疗效果也大不相同。因此,通过小分子代谢物研究血清学对hP-间充质干细胞移植的反应,并为疗效评估确定易于检测的标记物至关重要:方法:以 Mdr2-/-小鼠为 PSC 模型,以 Mdr2+/+ 小鼠为对照,根据肝脏病理学、肝酶和炎症因子评估 hP-MSC 治疗的疗效。收集血清样本进行12C-/13C-丹酰化和DmPA标记LC-MS分析,以研究hP-间充质干细胞治疗后代谢途径的变化。关键代谢物和调节酶通过 qRT-PCR 和 Western 印迹进行了验证。通过相关性分析和机器学习,确定了 hP-MSC 疗效的潜在生物标志物:结果:肝组织学、血清肝酶水平和炎症因子的综合结果支持了 hP-MSC 治疗的疗效。根据显着差异,初步确定了 41 个差异表达的代谢物;这些代谢物富含胆汁酸、脂质和羟脯氨酸代谢。治疗后,胆汁酸转运加快,而胆汁酸生成减少;不饱和脂肪酸合成总体上调,FADS2和伸长酶表达增加,脂肪酸β氧化增强;肝脏脯氨酸4-羟化酶表达减少,导致羟脯氨酸生成减少。通过对肝酶和代谢物进行相关分析,并结合时间趋势,确定了八种潜在的生物标记物:2-氨基琥珀酸半醛、L-1-吡咯啉-3-羟基-5-羧酸、L-异谷氨酰胺和马来酰胺酸在模型小鼠中含量较高,但在 hP-MSC 治疗后含量下降。相反,15-甲基棕榈酸、二十烯酸、壬癸酸和十八烷酸在模型小鼠体内含量较少,但在 hP-MSC 治疗后含量增加:这项研究揭示了 hP-MSC 治疗后 PSC 模型小鼠体内的代谢调节变化,并确定了 8 种有前景的生物标志物,为 hP-MSC 的治疗应用提供了临床前证据。
{"title":"Serum metabonomics reveal the effectiveness of human placental mesenchymal stem cell therapy for primary sclerosing cholangitis.","authors":"Yingduo Yu, Qigu Yao, Deying Chen, Zhehua Zhang, Qiaoling Pan, Jiong Yu, Hongcui Cao, Liang Li, Lanjuan Li","doi":"10.1186/s13287-024-03967-y","DOIUrl":"10.1186/s13287-024-03967-y","url":null,"abstract":"<p><strong>Background: </strong>The metabolic patterns of human placental-derived mesenchymal stem cell (hP-MSC) treatment for primary sclerosing cholangitis (PSC) remain unclear, and therapeutic effects significantly vary due to individual differences. Therefore, it is crucial to investigate the serological response to hP-MSC transplantation through small molecular metabolites and identify easily detectable markers for efficacy evaluation.</p><p><strong>Methods: </strong>Using Mdr2<sup>-/-</sup> mice as a PSC model and Mdr2<sup>+/+</sup> mice as controls, the efficacy of hP-MSC treatment was assessed based on liver pathology, liver enzymes, and inflammatory factors. Serum samples were collected for <sup>12</sup>C-/<sup>13</sup>C-dansylation and DmPA labeling LC-MS analysis to investigate changes in metabolic pathways after hP-MSC treatment. Key metabolites and regulatory enzymes were validated by qRT-PCR and Western blotting. Potential biomarkers of hP-MSC efficacy were identified through correlation analysis and machine learning.</p><p><strong>Results: </strong>Collectively, the results of the liver histology, serum liver enzyme levels, and inflammatory factors supported the therapeutic efficacy of hP-MSC treatment. Based on significant differences, 41 differentially expressed metabolites were initially identified; these were enriched in bile acid, lipid, and hydroxyproline metabolism. After treatment, bile acid transport was accelerated, whereas bile acid production was reduced; unsaturated fatty acid synthesis was upregulated overall, with increased FADS2 and elongase expression and enhanced fatty acid β-oxidation; hepatic proline 4-hydroxylase expression was decreased, leading to reduced hydroxyproline production. Correlation analysis of liver enzymes and metabolites, combined with time trends, identified eight potential biomarkers: 2-aminomuconate semialdehyde, L-1-pyrroline-3-hydroxy-5-carboxylic acid, L-isoglutamine, and maleamic acid were more abundant in model mice but decreased after hP-MSC treatment. Conversely, 15-methylpalmitic, eicosenoic, nonadecanoic, and octadecanoic acids were less abundant in model mice but increased after hP-MSC treatment.</p><p><strong>Conclusions: </strong>This study revealed metabolic regulatory changes in PSC model mice after hP-MSC treatment and identified eight promising biomarkers, providing preclinical evidence to support therapeutic applications of hP-MSC.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"346"},"PeriodicalIF":7.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11462665/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142393463","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 dissection reveals promotive role of ENO1 in leukemia stem cell self-renewal and chemoresistance in acute myeloid leukemia. 单细胞分析揭示了ENO1在急性髓性白血病的白血病干细胞自我更新和化疗抵抗中的促进作用。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-10-08 DOI: 10.1186/s13287-024-03969-w
Yun Tian, Jiafan Guo, Lipeng Mao, Zhixi Chen, Xingwei Zhang, Yangqiu Li, Yikai Zhang, Xianfeng Zha, Oscar Junhong Luo

Background: Quiescent self-renewal of leukemia stem cells (LSCs) and resistance to conventional chemotherapy are the main factors leading to relapse of acute myeloid leukemia (AML). Alpha-enolase (ENO1), a key glycolytic enzyme, has been shown to regulate embryonic stem cell differentiation and promote self-renewal and malignant phenotypes in various cancer stem cells. Here, we sought to test whether and how ENO1 influences LSCs renewal and chemoresistance within the context of AML.

Methods: We analyzed single-cell RNA sequencing data from bone marrow samples of 8 relapsed/refractory AML patients and 4 healthy controls using bioinformatics and machine learning algorithms. In addition, we compared ENO1 expression levels in the AML cohort with those in 37 control subjects and conducted survival analyses to correlate ENO1 expression with clinical outcomes. Furthermore, we performed functional studies involving ENO1 knockdown and inhibition in AML cell line.

Results: We used machine learning to model and infer malignant cells in AML, finding more primitive malignant cells in the non-response (NR) group. The differentiation capacity of LSCs and progenitor malignant cells exhibited an inverse correlation with glycolysis levels. Trajectory analysis indicated delayed myeloid cell differentiation in NR group, with high ENO1-expressing LSCs at the initial stages of differentiation being preserved post-treatment. Simultaneously, ENO1 and stemness-related genes were upregulated and co-expressed in malignant cells during early differentiation. ENO1 level in our AML cohort was significantly higher than the controls, with higher levels in NR compared to those in complete remission. Knockdown of ENO1 in AML cell line resulted in the activation of LSCs, promoting cell differentiation and apoptosis, and inhibited proliferation. ENO1 inhibitor can impede the proliferation of AML cells. Furthermore, survival analyses associated higher ENO1 expression with poorer outcome in AML patients.

Conclusions: Our findings underscore the critical role of ENO1 as a plausible driver of LSC self-renewal, a potential target for AML target therapy and a biomarker for AML prognosis.

背景:白血病干细胞(LSCs)的静止自我更新和对常规化疗的耐药性是导致急性髓性白血病(AML)复发的主要因素。α-烯醇化酶(ENO1)是一种关键的糖酵解酶,已被证明能调节胚胎干细胞分化,促进自我更新和各种癌症干细胞的恶性表型。在此,我们试图检验ENO1是否以及如何影响急性髓细胞性白血病背景下的LSCs更新和化疗耐药性:我们利用生物信息学和机器学习算法分析了8名复发/难治性AML患者和4名健康对照者骨髓样本的单细胞RNA测序数据。此外,我们还比较了急性髓细胞性白血病组群与37名对照组的ENO1表达水平,并进行了生存分析,以确定ENO1表达与临床结果的相关性。此外,我们还进行了ENO1在AML细胞系中敲除和抑制的功能研究:结果:我们使用机器学习方法对急性髓细胞白血病中的恶性细胞进行建模和推断,发现无应答(NR)组中有更多原始恶性细胞。LSCs和祖先恶性细胞的分化能力与糖酵解水平呈反比。轨迹分析表明,NR 组骨髓细胞分化延迟,治疗后仍保留了分化初期高 ENO1 表达的 LSCs。同时,在早期分化过程中,ENO1和干性相关基因在恶性细胞中上调并共同表达。我们的急性髓细胞群中的ENO1水平明显高于对照组,与完全缓解者相比,NR中的ENO1水平更高。在急性髓性白血病细胞系中敲除ENO1可激活LSCs,促进细胞分化和凋亡,并抑制细胞增殖。ENO1抑制剂可抑制AML细胞的增殖。此外,生存分析显示,ENO1表达越高,急性髓细胞白血病患者的预后越差:我们的研究结果强调了ENO1的关键作用,它是LSC自我更新的合理驱动因素、AML靶向治疗的潜在靶点以及AML预后的生物标志物。
{"title":"Single-cell dissection reveals promotive role of ENO1 in leukemia stem cell self-renewal and chemoresistance in acute myeloid leukemia.","authors":"Yun Tian, Jiafan Guo, Lipeng Mao, Zhixi Chen, Xingwei Zhang, Yangqiu Li, Yikai Zhang, Xianfeng Zha, Oscar Junhong Luo","doi":"10.1186/s13287-024-03969-w","DOIUrl":"10.1186/s13287-024-03969-w","url":null,"abstract":"<p><strong>Background: </strong>Quiescent self-renewal of leukemia stem cells (LSCs) and resistance to conventional chemotherapy are the main factors leading to relapse of acute myeloid leukemia (AML). Alpha-enolase (ENO1), a key glycolytic enzyme, has been shown to regulate embryonic stem cell differentiation and promote self-renewal and malignant phenotypes in various cancer stem cells. Here, we sought to test whether and how ENO1 influences LSCs renewal and chemoresistance within the context of AML.</p><p><strong>Methods: </strong>We analyzed single-cell RNA sequencing data from bone marrow samples of 8 relapsed/refractory AML patients and 4 healthy controls using bioinformatics and machine learning algorithms. In addition, we compared ENO1 expression levels in the AML cohort with those in 37 control subjects and conducted survival analyses to correlate ENO1 expression with clinical outcomes. Furthermore, we performed functional studies involving ENO1 knockdown and inhibition in AML cell line.</p><p><strong>Results: </strong>We used machine learning to model and infer malignant cells in AML, finding more primitive malignant cells in the non-response (NR) group. The differentiation capacity of LSCs and progenitor malignant cells exhibited an inverse correlation with glycolysis levels. Trajectory analysis indicated delayed myeloid cell differentiation in NR group, with high ENO1-expressing LSCs at the initial stages of differentiation being preserved post-treatment. Simultaneously, ENO1 and stemness-related genes were upregulated and co-expressed in malignant cells during early differentiation. ENO1 level in our AML cohort was significantly higher than the controls, with higher levels in NR compared to those in complete remission. Knockdown of ENO1 in AML cell line resulted in the activation of LSCs, promoting cell differentiation and apoptosis, and inhibited proliferation. ENO1 inhibitor can impede the proliferation of AML cells. Furthermore, survival analyses associated higher ENO1 expression with poorer outcome in AML patients.</p><p><strong>Conclusions: </strong>Our findings underscore the critical role of ENO1 as a plausible driver of LSC self-renewal, a potential target for AML target therapy and a biomarker for AML prognosis.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"347"},"PeriodicalIF":7.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11463110/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142393464","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
SMYD1 modulates the proliferation of multipotent cardiac progenitor cells derived from human pluripotent stem cells during myocardial differentiation through GSK3β/β-catenin&ERK signaling. 在心肌分化过程中,SMYD1通过GSK3β/β-catenin&ERK信号转导调节源自人类多能干细胞的多能心脏祖细胞的增殖。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-10-08 DOI: 10.1186/s13287-024-03899-7
Yun Chang, Rui Bai, Yongshuai Zhang, Wen-Jing Lu, Shuhong Ma, Min Zhu, Feng Lan, Youxu Jiang

Background: The histone-lysine N-methyltransferase SMYD1, which is specific to striated muscle, plays a crucial role in regulating early heart development. Its deficiency has been linked to the occurrence of congenital heart disease. Nevertheless, the precise mechanism by which SMYD1 deficiency contributes to congenital heart disease remains unclear.

Methods: We established a SMYD1 knockout pluripotent stem cell line and a doxycycline-inducible SMYD1 expression pluripotent stem cell line to investigate the functions of SMYD1 utilizing an in vitro-directed myocardial differentiation model.

Results: Cardiomyocytes lacking SMYD1 displayed drastically diminished differentiation efficiency, concomitant with heightened proliferation capacity of cardiac progenitor cells during the early cardiac differentiation stage. These cellular phenotypes were confirmed through experiments inducing the re-expression of SMYD1. Transcriptome sequencing and small molecule inhibitor intervention suggested that the GSK3β/β-catenin&ERK signaling pathway was involved in the proliferation of cardiac progenitor cells. Chromatin immunoprecipitation demonstrated that SMYD1 acted as a transcriptional activator of GSK3β through histone H3 lysine 4 trimethylation. Additionally, dual-luciferase analyses indicated that SMYD1 could interact with the promoter region of GSK3β, thereby augmenting its transcriptional activity. Moreover, administering insulin and Insulin-like growth factor 1 can enhance the efficacy of myocardial differentiation in SMYD1 knockout cells.

Conclusions: Our research indicated that the participation of SMYD1 in the GSK3β/β-catenin&ERK signaling cascade modulated the proliferation of cardiac progenitor cells during myocardial differentiation. This process was partly reliant on the transcription of GSK3β. Our research provided a novel insight into the genetic modification effect of SMYD1 during early myocardial differentiation. The findings were essential to the molecular mechanism and potential interventions for congenital heart disease.

背景:组蛋白-赖氨酸 N-甲基转移酶 SMYD1 是横纹肌的特异性基因,在调节早期心脏发育方面发挥着至关重要的作用。它的缺乏与先天性心脏病的发生有关。然而,SMYD1缺乏导致先天性心脏病的确切机制仍不清楚:方法:我们建立了一个SMYD1基因敲除的多能干细胞系和一个强力霉素诱导的SMYD1表达的多能干细胞系,利用体外引导的心肌分化模型研究SMYD1的功能:结果:在早期心脏分化阶段,缺乏SMYD1的心肌细胞显示出分化效率急剧下降,同时心脏祖细胞的增殖能力增强。通过诱导重新表达 SMYD1 的实验证实了这些细胞表型。转录组测序和小分子抑制剂干预表明,GSK3β/β-catenin&ERK 信号通路参与了心脏祖细胞的增殖。染色质免疫共沉淀表明,SMYD1通过组蛋白H3赖氨酸4三甲基化作用成为GSK3β的转录激活因子。此外,双荧光素酶分析表明,SMYD1 可与 GSK3β 的启动子区域相互作用,从而增强其转录活性。此外,注射胰岛素和胰岛素样生长因子1可增强SMYD1基因敲除细胞的心肌分化效果:我们的研究表明,SMYD1参与GSK3β/β-catenin&ERK信号级联调节了心肌分化过程中心脏祖细胞的增殖。这一过程部分依赖于GSK3β的转录。我们的研究为了解SMYD1在早期心肌分化过程中的遗传修饰作用提供了新的视角。这些发现对先天性心脏病的分子机制和潜在干预措施至关重要。
{"title":"SMYD1 modulates the proliferation of multipotent cardiac progenitor cells derived from human pluripotent stem cells during myocardial differentiation through GSK3β/β-catenin&ERK signaling.","authors":"Yun Chang, Rui Bai, Yongshuai Zhang, Wen-Jing Lu, Shuhong Ma, Min Zhu, Feng Lan, Youxu Jiang","doi":"10.1186/s13287-024-03899-7","DOIUrl":"10.1186/s13287-024-03899-7","url":null,"abstract":"<p><strong>Background: </strong>The histone-lysine N-methyltransferase SMYD1, which is specific to striated muscle, plays a crucial role in regulating early heart development. Its deficiency has been linked to the occurrence of congenital heart disease. Nevertheless, the precise mechanism by which SMYD1 deficiency contributes to congenital heart disease remains unclear.</p><p><strong>Methods: </strong>We established a SMYD1 knockout pluripotent stem cell line and a doxycycline-inducible SMYD1 expression pluripotent stem cell line to investigate the functions of SMYD1 utilizing an in vitro-directed myocardial differentiation model.</p><p><strong>Results: </strong>Cardiomyocytes lacking SMYD1 displayed drastically diminished differentiation efficiency, concomitant with heightened proliferation capacity of cardiac progenitor cells during the early cardiac differentiation stage. These cellular phenotypes were confirmed through experiments inducing the re-expression of SMYD1. Transcriptome sequencing and small molecule inhibitor intervention suggested that the GSK3β/β-catenin&ERK signaling pathway was involved in the proliferation of cardiac progenitor cells. Chromatin immunoprecipitation demonstrated that SMYD1 acted as a transcriptional activator of GSK3β through histone H3 lysine 4 trimethylation. Additionally, dual-luciferase analyses indicated that SMYD1 could interact with the promoter region of GSK3β, thereby augmenting its transcriptional activity. Moreover, administering insulin and Insulin-like growth factor 1 can enhance the efficacy of myocardial differentiation in SMYD1 knockout cells.</p><p><strong>Conclusions: </strong>Our research indicated that the participation of SMYD1 in the GSK3β/β-catenin&ERK signaling cascade modulated the proliferation of cardiac progenitor cells during myocardial differentiation. This process was partly reliant on the transcription of GSK3β. Our research provided a novel insight into the genetic modification effect of SMYD1 during early myocardial differentiation. The findings were essential to the molecular mechanism and potential interventions for congenital heart disease.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"350"},"PeriodicalIF":7.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11462858/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142393466","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
Vasoactive intestinal peptide promotes secretory differentiation and mitigates radiation-induced intestinal injury. 血管活性肠肽促进分泌分化,减轻辐射引起的肠道损伤。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-10-08 DOI: 10.1186/s13287-024-03958-z
Tatiana Agibalova, Anneke Hempel, H Carlo Maurer, Mohab Ragab, Anastasia Ermolova, Jessica Wieland, Caroline Waldherr Ávila de Melo, Fabian Heindl, Maximilian Giller, Julius Clemens Fischer, Markus Tschurtschenthaler, Birgit Kohnke-Ertel, Rupert Öllinger, Katja Steiger, Ihsan Ekin Demir, Dieter Saur, Michael Quante, Roland M Schmid, Moritz Middelhoff

Background: Vasoactive intestinal peptide (VIP) is a neuronal peptide with prominent distribution along the enteric nervous system. While effects of VIP on intestinal motility, mucosal vasodilation, secretion, and mucosal immune cell function are well-studied, the direct impact of VIP on intestinal epithelial cell turnover and differentiation remains less understood. Intestinal stem and progenitor cells are essential for the maintenance of intestinal homeostasis and regeneration, and their functions can be modulated by factors of the stem cell niche, including neuronal mediators. Here, we investigated the role of VIP in regulating intestinal epithelial homeostasis and regeneration following irradiation-induced injury.

Methods: Jejunal organoids were derived from male and female C57Bl6/J, Lgr5-EGFP-IRES-CreERT2 or Lgr5-EGFP-IRES-CreERT2/R26R-LSL-TdTomato mice and treated with VIP prior to analysis. Injury conditions were induced by exposing organoids to 6 Gy of irradiation (IR). To investigate protective effects of VIP in vivo, mice received 12 Gy of abdominal IR followed by intraperitoneal injections of VIP.

Results: We observed that VIP promotes epithelial differentiation towards a secretory phenotype predominantly via the p38 MAPK pathway. Moreover, VIP prominently modulated epithelial proliferation as well as the number and proliferative activity of Lgr5-EGFP+ progenitor cells under homeostatic conditions. In the context of acute irradiation injury in vitro, we observed that IR injury renders Lgr5-EGFP+ progenitor cells more susceptible to VIP-induced modulations, which coincided with the strong promotion of epithelial regeneration by VIP. Finally, the observed effects translate into an in vivo model of abdominal irradiation, where VIP showed to prominently mitigate radiation-induced injury.

Conclusions: VIP prominently governs intestinal homeostasis by regulating epithelial progenitor cell proliferation and differentiation and promotes intestinal regeneration following acute irradiation injury.

背景:血管活性肠肽(VIP)是一种神经肽,主要分布于肠道神经系统。VIP对肠道蠕动、粘膜血管扩张、分泌和粘膜免疫细胞功能的影响已得到充分研究,但VIP对肠上皮细胞更替和分化的直接影响仍鲜为人知。肠道干细胞和祖细胞对维持肠道平衡和再生至关重要,其功能可受干细胞龛因子(包括神经元介质)的调节。在此,我们研究了VIP在辐照损伤后调节肠上皮稳态和再生中的作用:方法:从雌雄 C57Bl6/J、Lgr5-EGFP-IRES-CreERT2 或 Lgr5-EGFP-IRES-CreERT2/R26R-LSL-TdTomato 小鼠中提取空肠器官组织,分析前用 VIP 处理。将器官组织暴露于 6 Gy 的辐照 (IR) 中诱发损伤。为了研究 VIP 在体内的保护作用,小鼠腹部接受了 12 Gy 的红外线照射,然后腹腔注射 VIP:结果:我们观察到,VIP 主要通过 p38 MAPK 通路促进上皮细胞向分泌表型分化。此外,VIP 还能显著调节上皮细胞的增殖,以及平衡状态下 Lgr5-EGFP+ 祖细胞的数量和增殖活性。在体外急性辐照损伤的情况下,我们观察到红外损伤使 Lgr5-EGFP+ 祖细胞更易受 VIP 诱导的调节作用的影响,这与 VIP 强力促进上皮再生的作用不谋而合。最后,观察到的效应转化到腹部照射的体内模型中,VIP 显示出显著减轻辐射诱导的损伤:结论:VIP 通过调节上皮祖细胞的增殖和分化,显著调节肠道稳态,并促进急性辐照损伤后的肠道再生。
{"title":"Vasoactive intestinal peptide promotes secretory differentiation and mitigates radiation-induced intestinal injury.","authors":"Tatiana Agibalova, Anneke Hempel, H Carlo Maurer, Mohab Ragab, Anastasia Ermolova, Jessica Wieland, Caroline Waldherr Ávila de Melo, Fabian Heindl, Maximilian Giller, Julius Clemens Fischer, Markus Tschurtschenthaler, Birgit Kohnke-Ertel, Rupert Öllinger, Katja Steiger, Ihsan Ekin Demir, Dieter Saur, Michael Quante, Roland M Schmid, Moritz Middelhoff","doi":"10.1186/s13287-024-03958-z","DOIUrl":"10.1186/s13287-024-03958-z","url":null,"abstract":"<p><strong>Background: </strong>Vasoactive intestinal peptide (VIP) is a neuronal peptide with prominent distribution along the enteric nervous system. While effects of VIP on intestinal motility, mucosal vasodilation, secretion, and mucosal immune cell function are well-studied, the direct impact of VIP on intestinal epithelial cell turnover and differentiation remains less understood. Intestinal stem and progenitor cells are essential for the maintenance of intestinal homeostasis and regeneration, and their functions can be modulated by factors of the stem cell niche, including neuronal mediators. Here, we investigated the role of VIP in regulating intestinal epithelial homeostasis and regeneration following irradiation-induced injury.</p><p><strong>Methods: </strong>Jejunal organoids were derived from male and female C57Bl6/J, Lgr5-EGFP-IRES-CreER<sup>T2</sup> or Lgr5-EGFP-IRES-CreER<sup>T2</sup>/R26R-LSL-TdTomato mice and treated with VIP prior to analysis. Injury conditions were induced by exposing organoids to 6 Gy of irradiation (IR). To investigate protective effects of VIP in vivo, mice received 12 Gy of abdominal IR followed by intraperitoneal injections of VIP.</p><p><strong>Results: </strong>We observed that VIP promotes epithelial differentiation towards a secretory phenotype predominantly via the p38 MAPK pathway. Moreover, VIP prominently modulated epithelial proliferation as well as the number and proliferative activity of Lgr5-EGFP<sup>+</sup> progenitor cells under homeostatic conditions. In the context of acute irradiation injury in vitro, we observed that IR injury renders Lgr5-EGFP<sup>+</sup> progenitor cells more susceptible to VIP-induced modulations, which coincided with the strong promotion of epithelial regeneration by VIP. Finally, the observed effects translate into an in vivo model of abdominal irradiation, where VIP showed to prominently mitigate radiation-induced injury.</p><p><strong>Conclusions: </strong>VIP prominently governs intestinal homeostasis by regulating epithelial progenitor cell proliferation and differentiation and promotes intestinal regeneration following acute irradiation injury.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"348"},"PeriodicalIF":7.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11462795/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142393468","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
Genetically engineered human induced pluripotent stem cells for the production of brain-targeting extracellular vesicles. 用于生产脑靶向细胞外囊泡的基因工程人类诱导多能干细胞。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-10-08 DOI: 10.1186/s13287-024-03955-2
Fan Tang, Tao Dong, Chengqian Zhou, Leon Deng, Hans B Liu, Wenshen Wang, Guanshu Liu, Mingyao Ying, Pan P Li

Background: Extracellular vesicles (EVs) are cell-secreted membrane vesicles that have become a promising, natural nanoparticle system for delivering either naturally carried or exogenously loaded therapeutic molecules. Among reported cell sources for EV manufacture, human induced pluripotent stem cells (hiPSCs) offer numerous advantages. However, hiPSC-EVs only have a moderate ability for brain delivery. Herein, we sought to develop a stable hiPSC line for producing EVs with substantially enhanced brain targeting by genetic engineering to overexpress rabies viral glycoprotein (RVG) peptide fused to the N terminus of lysosomal associated membrane protein 2B (RVG-Lamp2B) which has been shown capable of boosting the brain delivery of EVs via the nicotinic acetylcholine receptor.

Methods: An RVG-Lamp2B-HA expression cassette was knocked into the AAVS1 safe harbor locus of a control hiPSC line using the CRISPR/Cas9-assisted homologous recombination. Western blot was used to detect the expression of RVG-Lamp2B-HA in RVG-edited hiPSCs as well as EVs derived from RVG-edited hiPSCs. Uptake of EVs by SH-SY5Y cells in the presence of various endocytic inhibitors was analyzed using flow cytometry. Biodistribution and brain delivery of intravenously injected control and RVG-modified EVs in wild-type mice were examined using ex vivo fluorescent imaging.

Results: Here we report that an RVG-Lamp2B-HA expression cassette was knocked into the AAVS1 safe harbor locus of a control hiPSC line using the CRISPR/Cas9-assisted homologous recombination. The RVG-edited iPSCs have normal karyotype, express pluripotency markers, and have differentiation potential. Expression of RVG-Lamp2B-HA was detected in total cell extracts as well as EVs derived from RVG-edited (vs. control) hiPSCs. The RVG-modified EVs enter neuronal cells via distinct endocytic pathways, compared with control EVs. The biodistribution study confirmed that EVs derived from RVG-edited hiPSCs possess higher brain delivery efficiency.

Conclusion: Taken together, we have established stable, genetically engineered hiPSCs for producing EVs with RVG expression, offering the improved ability for brain-targeted drug delivery.

背景:细胞外囊泡(EVs)是细胞分泌的膜囊泡,已成为一种很有前景的天然纳米颗粒系统,可用于递送天然携带或外源负载的治疗分子。在已报道的制造 EV 的细胞来源中,人类诱导多能干细胞(hiPSC)具有众多优势。然而,hiPSC-EV 在脑部递送方面的能力一般。在此,我们试图开发一种稳定的hiPSC细胞系,通过基因工程过表达与溶酶体相关膜蛋白2B(RVG-Lamp2B)N末端融合的狂犬病病毒糖蛋白(RVG)多肽来生产EVs,从而大大增强EVs的脑靶向性:方法:利用 CRISPR/Cas9 辅助同源重组技术将 RVG-Lamp2B-HA 表达盒敲入对照 hiPSC 株系的 AAVS1 安全港基因座。用 Western blot 检测 RVG 编辑的 hiPSCs 中 RVG-Lamp2B-HA 的表达,以及从 RVG 编辑的 hiPSCs 中提取的 EVs。使用流式细胞术分析了SH-SY5Y细胞在各种内细胞抑制剂存在下对EVs的吸收。利用体外荧光成像技术检测了野生型小鼠静脉注射对照组和RVG修饰的EVs的生物分布和脑输送情况:结果:我们在此报告利用CRISPR/Cas9辅助同源重组技术将RVG-Lamp2B-HA表达盒敲入对照hiPSC系的AAVS1安全港基因座。经 RVG 编辑的 iPSC 具有正常核型、表达多能性标记和分化潜能。在总细胞提取物以及从 RVG 编辑(与对照组相比)的 hiPSCs 提取的 EVs 中检测到了 RVG-Lamp2B-HA 的表达。与对照EVs相比,RVG修饰的EVs通过不同的内细胞途径进入神经细胞。生物分布研究证实,RVG修饰过的hiPSCs衍生的EVs具有更高的脑输送效率:综上所述,我们建立了稳定的基因工程 hiPSCs,用于生产具有 RVG 表达的 EVs,提高了脑靶向给药能力。
{"title":"Genetically engineered human induced pluripotent stem cells for the production of brain-targeting extracellular vesicles.","authors":"Fan Tang, Tao Dong, Chengqian Zhou, Leon Deng, Hans B Liu, Wenshen Wang, Guanshu Liu, Mingyao Ying, Pan P Li","doi":"10.1186/s13287-024-03955-2","DOIUrl":"10.1186/s13287-024-03955-2","url":null,"abstract":"<p><strong>Background: </strong>Extracellular vesicles (EVs) are cell-secreted membrane vesicles that have become a promising, natural nanoparticle system for delivering either naturally carried or exogenously loaded therapeutic molecules. Among reported cell sources for EV manufacture, human induced pluripotent stem cells (hiPSCs) offer numerous advantages. However, hiPSC-EVs only have a moderate ability for brain delivery. Herein, we sought to develop a stable hiPSC line for producing EVs with substantially enhanced brain targeting by genetic engineering to overexpress rabies viral glycoprotein (RVG) peptide fused to the N terminus of lysosomal associated membrane protein 2B (RVG-Lamp2B) which has been shown capable of boosting the brain delivery of EVs via the nicotinic acetylcholine receptor.</p><p><strong>Methods: </strong>An RVG-Lamp2B-HA expression cassette was knocked into the AAVS1 safe harbor locus of a control hiPSC line using the CRISPR/Cas9-assisted homologous recombination. Western blot was used to detect the expression of RVG-Lamp2B-HA in RVG-edited hiPSCs as well as EVs derived from RVG-edited hiPSCs. Uptake of EVs by SH-SY5Y cells in the presence of various endocytic inhibitors was analyzed using flow cytometry. Biodistribution and brain delivery of intravenously injected control and RVG-modified EVs in wild-type mice were examined using ex vivo fluorescent imaging.</p><p><strong>Results: </strong>Here we report that an RVG-Lamp2B-HA expression cassette was knocked into the AAVS1 safe harbor locus of a control hiPSC line using the CRISPR/Cas9-assisted homologous recombination. The RVG-edited iPSCs have normal karyotype, express pluripotency markers, and have differentiation potential. Expression of RVG-Lamp2B-HA was detected in total cell extracts as well as EVs derived from RVG-edited (vs. control) hiPSCs. The RVG-modified EVs enter neuronal cells via distinct endocytic pathways, compared with control EVs. The biodistribution study confirmed that EVs derived from RVG-edited hiPSCs possess higher brain delivery efficiency.</p><p><strong>Conclusion: </strong>Taken together, we have established stable, genetically engineered hiPSCs for producing EVs with RVG expression, offering the improved ability for brain-targeted drug delivery.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"345"},"PeriodicalIF":7.1,"publicationDate":"2024-10-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11462716/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142393458","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
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: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