Gu Jianing, Su Zhanyu, Wang Yini, Chen Yuexi, Cui Zekai, Li Shengguo, Ding Chengcheng, Sheng Wang, Li Kangjun, Tang Shibo, Chen Jiansu
{"title":"The influence of femtosecond laser intrastromal lenticules on the characteristics and maturity in tissue-engineered stem cell-derived retinal pigment epithelium sheets.","authors":"Gu Jianing, Su Zhanyu, Wang Yini, Chen Yuexi, Cui Zekai, Li Shengguo, Ding Chengcheng, Sheng Wang, Li Kangjun, Tang Shibo, Chen Jiansu","doi":"10.1186/s13287-025-04463-7","DOIUrl":"10.1186/s13287-025-04463-7","url":null,"abstract":"<p><strong>Background: </strong>Recent advances in clinical trials have involved the transplantation of induced retinal pigment epithelium (iRPE) cells from stem cells in creating a functional monolayer that mimics the characteristics of natural adult RPE cells. One method of achieving this goal is through the use of tissue engineering. In this research, decellularised femtosecond laser intrastromal lenticules (dfLEN) were employed as a scaffold for cultivating a bioengineered iRPE monolayer sheet.</p><p><strong>Methods: </strong>iRPE cells were obtained by differentiating induced pluripotent stem cells (iPSC). These cells were then seeded on decellularized FLI-lenticules (dfLEN). The functionality, characterization, and oxidative stress of iRPE cultured on dfLEN were compared with those cultured on plates (TCP) using various assays such as immunofluorescence (IF), Edu, CCK8, ELISA, DFCH-DA, and JC-1. Additionally, RNA-seq assays and electron microscope (SEM and TEM) were used to test the iRPE characteristic on engineered dfLEN. Finally, we evaluated the biocompatibility of iRPE-dfLEN sheets by transplanting them into the subretinal space of New Zealand white rabbits.</p><p><strong>Results: </strong>The iRPE cells cultured on dfLEN exhibited morphology and physiology similar to that of native RPE tissue. The dfLEN not only increased the resistance capacity of iRPE cells but also improved their functional properties compared to TCP. In addition, our results indicate that dfLEN enhances the expression of genes associated with cilium assembly, resulting in notable improvements in ciliogenesis in iRPE cells. Finally, the dfLEN-iRPE sheets demonstrated favorable biocompatibility and some viability when transplanted into the subretinal space of rabbits for a period of 14 days.</p><p><strong>Conclusions: </strong>We determine that engineered RPE sheets using dfLEN scaffolds enhance RPE characteristics and functions, and suggest that dfLEN scaffolds promote cilium process maturation and polarization of iPSC-derived epithelial cells. Such a strategy for constructing iRPE sheets holds significant potential for advancing RPE cell therapy, disease models, and drug screening platforms.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"316"},"PeriodicalIF":7.1,"publicationDate":"2025-06-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12181919/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144337095","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}
{"title":"The role and mechanism of mesenchymal stem cells in immunomodulation of type 1 diabetes mellitus and its complications: recent research progress and challenges: a review.","authors":"Chengran Wang, Yimeng Wu, Jinlan Jiang","doi":"10.1186/s13287-025-04431-1","DOIUrl":"10.1186/s13287-025-04431-1","url":null,"abstract":"<p><strong>Background: </strong>Mesenchymal stem cells are of great interest because of their multipotency, immune modulation capacity, and tissue and vascular regeneration effects. They are used in treating type 1 diabetes mellitus, helping improve the pancreatic environment and insulin secretion. Type 1 diabetes mellitus predominantly affects children and adolescents, with early onset and a prolonged course that can lead to multiorgan complications and related disorders. Studies using mesenchymal stem cells to treat type 1 diabetes mellitus have yielded promising results. This review discusses the common animal models of type 1 diabetes mellitus, mesenchymal stem cell immunotherapy mechanisms, and combined diabetes treatments. Its purpose is to summarize the current evidence on mesenchymal stem cell use in type 1 diabetes, providing insights for further research directions.</p><p><strong>Main findings: </strong>Current studies show that mesenchymal stem cells play an active role in the treatment of type 1 diabetes; however, clinical trials remain rare, necessitating more basic and preclinical research to identify optimal treatments.</p><p><strong>Conclusions: </strong>Mesenchymal stem cells can treat type 1 diabetes through a variety of immune mechanisms and also play a positive role in the treatment of type 1 diabetes complications. At the same time, it can be combined with other therapies to play a better therapeutic role.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"308"},"PeriodicalIF":7.1,"publicationDate":"2025-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12175325/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144317868","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}
Jie Chen, Yiwei Zhao, Chongmai Zeng, Guoli Tian, Zhicai Feng, Yang Cao
{"title":"Mechanical tension-induced Dalrd3 elevation enhances osteogenic differentiation of bone suture stem cells by upregulating Id3 translation.","authors":"Jie Chen, Yiwei Zhao, Chongmai Zeng, Guoli Tian, Zhicai Feng, Yang Cao","doi":"10.1186/s13287-025-04380-9","DOIUrl":"10.1186/s13287-025-04380-9","url":null,"abstract":"<p><strong>Background: </strong>Craniomaxillofacial sutures play a critical role in craniomaxillofacial development through continuous bone reconstruction and regeneration, processes modulated by mechanical tension. Bone suture stem cells (SuSCs) are central to these functions. Distraction osteogenesis, which promotes craniomaxillofacial suture growth, is a common therapeutic approach for craniofacial deformities. However, the underlying mechanisms by which mechanical forces drive suture and bone remodeling remain poorly understood, posing significant clinical challenges.</p><p><strong>Methods: </strong>To investigate these mechanisms, we established a rapid maxillary expansion (RME) model in mice to widen the midpalatal suture. Single-cell RNA sequencing (scRNA-seq) was employed to identify subsets of SuSCs responsive to mechanical tension and analyze their differentiation potential under varying conditions. Further functional studies were conducted to explore the role of DALR anticodon binding domain containing 3 (Dalrd3) and its associated tRNA 3-methylcytosine (m3C) modification in SuSCs under mechanical tension.</p><p><strong>Results: </strong>Our study identified a subset of SuSCs with multidirectional differentiation potential that shifted from a chondrogenic to an osteogenic trajectory in response to mechanical tension. Mechanical tension also upregulated Dalrd3 expression and its associated tRNA m3C modification in activated SuSCs. Knockdown of Dalrd3 in SuSCs significantly impaired osteogenic differentiation, proliferation, migratory capacity, and translational activity within the bone morphogenetic protein (BMP) signaling pathway. Furthermore, Dalrd3 knockdown suppressed the translational activity of inhibitor of DNA binding 3 (Id3), a key BMP-induced mediator of osteoblastogenesis. Restoring Id3 expression in Dalrd3-deficient SuSCs rescued their osteogenic, proliferative, and migratory functions.</p><p><strong>Conclusions: </strong>These findings reveal a translational regulatory mechanism in SuSCs activated by mechanical tension and underscore the pivotal role of Dalrd3 in suture remodeling and bone formation. The insights provided by this study have the potential to guide targeted therapeutic strategies for optimizing distraction osteogenesis and other treatments for craniofacial deformities.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"309"},"PeriodicalIF":7.1,"publicationDate":"2025-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12175324/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144317867","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}
{"title":"Mechanical harvesting of cell sheets: an efficient approach for bone and cartilage tissue engineering.","authors":"Fatemeh Leisi Mehrabani, Yalda Alibeigian, Mohamadreza Baghaban Eslaminejad, Samaneh Hosseini","doi":"10.1186/s13287-025-04411-5","DOIUrl":"10.1186/s13287-025-04411-5","url":null,"abstract":"<p><p>Cell sheet engineering (CSE) has demonstrated significant promise for the advancement and application of tissue-engineered constructs in the fields of tissue engineering and regenerative medicine. In this technology, cells are cultured to form a monolayer, which is then detached from the culture surface as a complete sheet. This process preserves cell interactions, maintains cellular phenotypes and functions, and retains the integrity of the cell-extracellular matrix (ECM). A main characteristic of the cell sheet is its ability to retain the native ECM components secreted by cells. When the ECM is preserved in cell sheets, cells are surrounded by a much more biologically appropriate environment to increase their regenerative potential, thereby offering more native conditions for cell growth and differentiation. CSE has shown promising results in a wide range of applications, including bone and cartilage. The cell sheets can be directly transplanted to the target site, where they integrate with the host tissue and enhance regeneration. The main challenge in CSE is how to detach an intact cell sheet without disturbing the ECM and cell‒cell connections. There are various methods for removing cell sheets that lead to the harvesting of intact cell sheets. Among the various methods for harvesting cell sheets, temperature-responsive systems and mechanical peeling are the most common. Mechanical harvesting, in particular, is a simple, cost-effective, accessible method that is widely used in research, especially in the scope of bone and cartilage tissue engineering. This article aims to review the application of cell sheets in bone and cartilage tissue engineering, with a focus on practical and cost-effective mechanical harvesting methods.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"310"},"PeriodicalIF":7.1,"publicationDate":"2025-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12175405/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144317866","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}
Rong Hu, Jin Zhao, Kuan Chen Lai, Shikun Wang, Jianqing Zheng, Christopher Stoddard, Laijun Lai
{"title":"CHD7 regulates definitive endodermal and mesodermal development from human embryonic stem cells.","authors":"Rong Hu, Jin Zhao, Kuan Chen Lai, Shikun Wang, Jianqing Zheng, Christopher Stoddard, Laijun Lai","doi":"10.1186/s13287-025-04437-9","DOIUrl":"10.1186/s13287-025-04437-9","url":null,"abstract":"<p><strong>Background: </strong>CHD7 encodes an ATP-dependent chromodomain helicase DNA binding protein; mutations in this gene lead to multiple developmental disorders, including CHARGE (Coloboma, Heart defects, Atresia of the choanae, Retardation of growth and development, Genital hypoplasia, and Ear anomalies) syndrome. How the mutations cause multiple defects remains largely unclear. Embryonic definitive endoderm (DE) generates the epithelial compartment of vital organs such as the thymus, liver, pancreas, and intestine.</p><p><strong>Methods: </strong>In this study, we used the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 technique to delete the CHD7 gene in human embryonic stem cells (hESCs) to generate CHD7 homozygous mutant (CHD7<sup>-/-</sup>), heterozygous mutant (CHD7<sup>+/-</sup>), and control wild-type (CHD7<sup>+/+</sup>) cells. We then investigated the ability of the hESCs to develop into DE and the other two germ layers, mesoderm and ectoderm in vitro. We also compared global gene expression and chromatin accessibility among the hESC-DE cells by RNA sequencing (RNA-seq) and the assay for transposase-accessible chromatin with sequencing (ATAC-seq).</p><p><strong>Results: </strong>We found that deletion of CHD7 led to reduced capacity to develop into DE and mesoderm in a dose-dependent manner. Loss of CHD7 led to significant changes in the expression and chromatin accessibility of genes associated with several pathways. We identified 40 genes that were highly down-regulated in both the expression and chromatin accessibility in CHD7 deleted hESC-DE cells.</p><p><strong>Conclusions: </strong>CHD7 is critical for DE and mesodermal development from hESCs. Our results provide new insights into the mechanisms by which CHD7 mutations cause multiple congenital anomalies.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"311"},"PeriodicalIF":7.1,"publicationDate":"2025-06-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12175391/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144317865","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}
Yu-Lu Zhou, Shingo Ogura, Hao Ma, Rong-Bin Liang, Shao-Yihan Fang, Yue-Ming Wang, Yan Wo, Wen-Jin Wang, De-Wu Liu
{"title":"Adipose-derived stem cells extracellular vesicles enhance diabetic wound healing via CCN2/PI3K/AKT pathway: therapeutic potential and mechanistic insights.","authors":"Yu-Lu Zhou, Shingo Ogura, Hao Ma, Rong-Bin Liang, Shao-Yihan Fang, Yue-Ming Wang, Yan Wo, Wen-Jin Wang, De-Wu Liu","doi":"10.1186/s13287-025-04354-x","DOIUrl":"10.1186/s13287-025-04354-x","url":null,"abstract":"<p><strong>Background: </strong>Adipose-derived stem cells extracellular vesicles (ADSCs-EVs) hold significant promise in tissue repair and regeneration. While they have been reported to enhance diabetic wound healing, the precise mechanisms remain unclear.</p><p><strong>Methods: </strong>ADSCs-EVs were isolated via ultracentrifugation and characterized through transmission electron microscopy, Western blot, and nanoparticle tracking analysis. Their effects on human umbilical vein endothelial cells (HUVECs) and RAW 264.7 macrophages were assessed in vitro, focusing on cell proliferation, migration, tube formation, and macrophage polarization. A diabetic rat wound model was used to evaluate their therapeutic impact on wound healing and angiogenesis, with histological and immunofluorescence analyses. mRNA sequencing identified Cellular communication network factor 2(CCN2) as a key upregulated gene, leading to further exploration of its role in ADSCs-EVs-mediated angiogenesis and wound healing via the PI3K/AKT pathway. Gene silencing (si-CCN2) and pharmacological inhibition (LY294002) were employed both in vitro and in vivo.</p><p><strong>Results: </strong>ADSCs-EVs were successfully isolated and characterized. In vitro, ADSCs-EVs promoted HUVEC proliferation, migration, and tube formation, and facilitated macrophage polarization to the M2 phenotype. In vivo studies using a diabetic rat wound model confirmed the pro-healing effects of ADSCs-EVs, including enhanced angiogenesis, granulation tissue formation, and accelerated wound closure. mRNA sequencing revealed that CCN2 expression was significantly upregulated in diabetic wound tissues treated with ADSCs-EVs. Further experiments showed that inhibiting CCN2 expression (si-CCN2) or blocking the PI3K/AKT pathway (LY294002) partially suppressed HUVEC proliferation, migration, tube formation, and angiogenesis, and counteracted the pro-healing effects of ADSCs-EVs.</p><p><strong>Conclusions: </strong>ADSCs-EVs promote diabetic wound healing through the CCN2/PI3K/AKT pathway, offering a promising therapeutic target for diabetic wound repair.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"304"},"PeriodicalIF":7.1,"publicationDate":"2025-06-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12168405/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144302774","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}
Xiangying Ou, Juan Yang, Guihua Du, Linfeng Yang, Rong Zhong, Fei Long, Haocong Huang, Huihong Zeng, Lijian Shao
{"title":"Histone hyperacetylation disrupts spermatogonial stem cells homeostasis and impairs spermiogenesis.","authors":"Xiangying Ou, Juan Yang, Guihua Du, Linfeng Yang, Rong Zhong, Fei Long, Haocong Huang, Huihong Zeng, Lijian Shao","doi":"10.1186/s13287-025-04385-4","DOIUrl":"10.1186/s13287-025-04385-4","url":null,"abstract":"<p><strong>Background: </strong>It is well-known that epigenetic regulation is involved in the negative effects of environmental physical, chemical and biological factors exposures on organs. To investigate the effects and mechanisms of histone hyperacetylation caused by environmental stress on spermatogenesis, we used the histone deacetylase inhibitor Panobinostat (PANO) to establish the hyperacetylation models in mice.</p><p><strong>Methods: </strong>To investigate the effects of PANO on testicular function of male reproductive system, we conducted the evaluation of sperm quality parameters in mice. The morphological changes of testes were observed through hematoxylin and eosin (H&E) staining on paraffin sections and quantified by stereological measurements after treatment to explore the cause of infertility and figure out if PANO has any effect on the reproductive system of male mice. Immunofluorescence, immunohistochemistry and immunoblotting were utilized to elucidate the impact and mechanisms of PANO treatment on spermatogenesis. RNA-seq analysis was performed on mouse testes to elucidate the underlying mechanisms of PANO.</p><p><strong>Results: </strong>The rates of sperm survival and movement were reduced while malformation rate of sperm increased in the 34.4-day PANO group. Gene Set Enrichment Analysis (GSEA) supported the significant role of PANO in modulating cilium movement, sperm axoneme assembly and flagellated sperm motility. Numbers of MVH<sup>+</sup> cells were decreased while the numbers of SCP3<sup>+</sup> cells were significantly increased after PANO treatment compared to those in the control (CTL) group. The protein levels of PLZF in PANO-treated testes were dramatically reduced along with the increased distribution changes of SOX9, F4/80 protein. Further data demonstrated that PANO impeded spermiogenesis at the stage XI in the 34.4-day PANO group and enhanced the transcriptome levels of histone variants H2bc4 and H1f2.</p><p><strong>Conclusions: </strong>We have reported that PANO exerts a negative impact on the spermatogonial stem cell pool in mouse testes by disrupting its niche. This disruption leads to a reduction in germ cell numbers and impairs sperm function in mice, ultimately resulting in infertility. Moreover, PANO destabilizes the nucleosomes by increasing the transcriptional levels of H2bc4 and H1f2, affects the histone-to-protamine transition, and arrests spermiogenesis at the elongating spermatid stage. These findings also suggest that H2bc4 and H1f2 may be potential key biomarkers in the testis for diagnosing male infertility associated with aberrant histone hyperacetylation due to the exposure of environmental pollutants.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"305"},"PeriodicalIF":7.1,"publicationDate":"2025-06-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12168342/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144302790","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}
Depti Bellani, Raji Patil, Ashwin Prabhughate, Riya Shahare, Michael Gold, Rinky Kapoor, Debraj Shome
{"title":"Pathophysiological mechanisms of hair follicle regeneration and potential therapeutic strategies.","authors":"Depti Bellani, Raji Patil, Ashwin Prabhughate, Riya Shahare, Michael Gold, Rinky Kapoor, Debraj Shome","doi":"10.1186/s13287-025-04420-4","DOIUrl":"10.1186/s13287-025-04420-4","url":null,"abstract":"<p><p>Androgenetic alopecia (AGA) is a chronic and progressive hair loss disorder marked by follicular miniaturization and a shortened anagen phase. While androgenic and genetic factors contribute to its pathogenesis, increasing evidence highlights the importance of dysregulated molecular signaling in impaired hair follicle (HF) regeneration.This review explores the interconnected signaling pathways that govern HF cycling and regeneration-Wnt/β-catenin, Sonic Hedgehog (Shh), Bone Morphogenetic Protein (BMP), and Notch. Wnt/β-catenin activation initiates anagen by stimulating stem cell proliferation and follicle formation, while Shh supports follicular proliferation and morphogenesis. Notch regulates HF stem cell (HFSC) fate, and BMP enforces quiescence and catagen onset. Crucially, crosstalk between Wnt-BMP and Shh-Notch pathways ensures follicular homeostasis, highlighting the need to view these pathways as an integrated regulatory network.Recent therapeutic innovations focus on modulating these signaling cascades. Small molecules such as valproic acid and CHIR99021 activate Wnt signaling; smoothened agonists target Shh; and Noggin mimetics or BMP-neutralizing antibodies inhibit BMP activity. These approaches have shown promising outcomes in preclinical models, including mouse studies, in vitro HFSC systems, etc. Additionally, emerging gene editing technologies (e.g., CRISPR-Cas9) and stem cell-biomaterial integration offer regenerative strategies that move beyond symptomatic treatments like minoxidil or hair transplantation.Given that AGA is associated with androgen-mediated Wnt suppression and TGF-β activation, targeting these dysregulated networks presents a promising route for long-term management. A deeper understanding of pathway interactions lays the groundwork for precise, durable, and disease-modifying therapies in the evolving landscape of alopecia treatment.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"302"},"PeriodicalIF":7.1,"publicationDate":"2025-06-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12168304/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144302791","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}
Angela Marcianti, Eleonora Spampinato, Sara Nava, Giulia Maria Stella, Paola Perego, Simona Pogliani, Simona Frigerio, Luca Mirra, Paola Gagni, Fabio Moda, Federico Angelo Cazzaniga, Giovanni Luca Beretta, Guido Maronati, Giuseppe Paglia, Angelo Guido Corsico, Catia Traversari, Daniela Lisini
{"title":"Extracellular vesicles isolated from adipose tissue-derived mesenchymal stromal cells as carriers for Paclitaxel delivery.","authors":"Angela Marcianti, Eleonora Spampinato, Sara Nava, Giulia Maria Stella, Paola Perego, Simona Pogliani, Simona Frigerio, Luca Mirra, Paola Gagni, Fabio Moda, Federico Angelo Cazzaniga, Giovanni Luca Beretta, Guido Maronati, Giuseppe Paglia, Angelo Guido Corsico, Catia Traversari, Daniela Lisini","doi":"10.1186/s13287-025-04435-x","DOIUrl":"10.1186/s13287-025-04435-x","url":null,"abstract":"<p><strong>Background: </strong>Mesenchymal Stromal Cells (MSC)-derived Extracellular Vesicles (EV) represent innovative tools for drug delivery systems. However, their clinical use is limited by the lack of standardized good manufacturing practice (GMP)-compliant isolation and conservation protocols. In this study, we developed a GMP-compliant protocol for the preparation of MSC-EVs and investigated the feasibility of producing EVs loaded with paclitaxel (PTX) for clinical application as drug products.</p><p><strong>Methods: </strong>Adipose tissues from 13 donors were used to obtain MSC-EVs via culture supernatant ultracentrifugation. EVs loaded with PTX were manufactured by adding the drug to the culture medium of MSCs before supernatant collection. EV identity was verified in terms of concentration/size, protein content, morphology, and expression of EV surface markers. The anti-proliferative activity, accumulation ability in tumor cells and PTX content, as well as their stability over time, were also evaluated.</p><p><strong>Results: </strong>High numbers of EV/EV-PTX compliant in terms of integrity/identity were obtained and can be successfully stored for up to one year at -80 °C. Cellular studies have shown that EVs are capable of accumulating in tumor cells and, when loaded with PTX, inhibiting the proliferation of a pleural mesothelioma cell line.</p><p><strong>Conclusions: </strong>These results support the potential future clinical use of EVs as carriers for drug delivery to improve cancer treatment strategies.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"307"},"PeriodicalIF":7.1,"publicationDate":"2025-06-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12168270/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144302775","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}
{"title":"Therapeutic potential of isoproterenol in androgenetic alopecia: activation of hair follicle stem cells via the PI3K/AKT/β-Catenin signaling pathway.","authors":"Jiarui Zhang, Jinxing Peng, Zhexiang Fan, Hailin Wang, Lihong Wen, Yong Miao, Yu Chai, Zhiqi Hu, Ruosi Chen","doi":"10.1186/s13287-025-04418-y","DOIUrl":"10.1186/s13287-025-04418-y","url":null,"abstract":"<p><strong>Background: </strong>Androgenetic alopecia (AGA) is characterized by the depletion or dormancy of hair follicle stem cells (HFSCs), leading to hair thinning and miniaturization. Reactivating the dormant HFSCs is a promising therapeutic approach. Adrenergic β2 receptor (ADRB2) activation has been shown to promote hair growth in animal models via the Sonic Hedgehog (SHH) pathway, but its potential for treating clinical AGA patients remains unexamined.</p><p><strong>Methods: </strong>We investigated the role of the PI3K/AKT signaling pathway in AGA pathogenesis, focusing on the hair follicle-sympathetic nerve axis. The ADRB2 agonist, isoproterenol (ISO), was administered to assess its effects on AGA hair follicle organ culture model and HFSC proliferation. The mechanisms underlying these effects were explored by analyzing the PI3K/AKT/β-Catenin pathway.</p><p><strong>Results: </strong>Our results showed abnormal PI3K/AKT pathway expression in AGA hair follicles, with associated defects in the hair follicle-sympathetic nerve axis. ISO treatment accelerated AGA hair follicle growth and promoted the proliferation of HFSC. Mechanistically, ISO facilitated the HFSC activation by modulating the PI3K/AKT/β-Catenin pathway.</p><p><strong>Conclusions: </strong>ISO effectively promotes hair growth in both animal models and AGA patients. ISO stimulating the proliferation of dormant cell population enriched in HFSC. This process was likely mediated by the PI3K/AKT/β-Catenin pathway. These findings provide novel insights into the reactivation of HFSCs and suggest that adrenergic signaling stimulation may be a promising strategy for managing hair loss.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"306"},"PeriodicalIF":7.1,"publicationDate":"2025-06-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12168277/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144302792","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}