首页 > 最新文献

Stem Cell Research & Therapy最新文献

英文 中文
Cell-based regenerative and rejuvenation strategies for treating neurodegenerative diseases.
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-04-06 DOI: 10.1186/s13287-025-04285-7
Sixiu Deng, Huangfan Xie, Bingqing Xie

Neurodegenerative diseases including Alzheimer's and Parkinson's disease are age-related disorders which severely impact quality of life and impose significant societal burdens. Cellular senescence is a critical factor in these disorders, contributing to their onset and progression by promoting permanent cell cycle arrest and reducing cellular function, affecting various types of cells in brain. Recent advancements in regenerative medicine have highlighted "R3" strategies-rejuvenation, regeneration, and replacement-as promising therapeutic approaches for neurodegeneration. This review aims to critically analyze the role of cellular senescence in neurodegenerative diseases and organizes therapeutic approaches within the R3 regenerative medicine paradigm. Specifically, we examine stem cell therapy, direct lineage reprogramming, and partial reprogramming in the context of R3, emphasizing how these interventions mitigate cellular senescence and counteracting aging-related neurodegeneration. Ultimately, this review seeks to provide insights into the complex interplay between cellular senescence and neurodegeneration while highlighting the promise of cell-based regenerative strategies to address these debilitating conditions.

{"title":"Cell-based regenerative and rejuvenation strategies for treating neurodegenerative diseases.","authors":"Sixiu Deng, Huangfan Xie, Bingqing Xie","doi":"10.1186/s13287-025-04285-7","DOIUrl":"10.1186/s13287-025-04285-7","url":null,"abstract":"<p><p>Neurodegenerative diseases including Alzheimer's and Parkinson's disease are age-related disorders which severely impact quality of life and impose significant societal burdens. Cellular senescence is a critical factor in these disorders, contributing to their onset and progression by promoting permanent cell cycle arrest and reducing cellular function, affecting various types of cells in brain. Recent advancements in regenerative medicine have highlighted \"R3\" strategies-rejuvenation, regeneration, and replacement-as promising therapeutic approaches for neurodegeneration. This review aims to critically analyze the role of cellular senescence in neurodegenerative diseases and organizes therapeutic approaches within the R3 regenerative medicine paradigm. Specifically, we examine stem cell therapy, direct lineage reprogramming, and partial reprogramming in the context of R3, emphasizing how these interventions mitigate cellular senescence and counteracting aging-related neurodegeneration. Ultimately, this review seeks to provide insights into the complex interplay between cellular senescence and neurodegeneration while highlighting the promise of cell-based regenerative strategies to address these debilitating conditions.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"167"},"PeriodicalIF":7.1,"publicationDate":"2025-04-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11974143/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143796332","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
Effect of immunosuppression on hESC-derived retina organoids in vitro and in vivo.
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-04-05 DOI: 10.1186/s13287-025-04271-z
Robert Sims, Bin Lin, Yuntian Xue, Raghda Fouda, Bryce T McLelland, Gabriel Nistor, Hans S Keirstead, Andrew W Browne, Magdalene J Seiler

Background: Photoreceptor (PR) enriched retinal organoid (RO) sheets (human embryonic stem cell [hESC]-derived ROs) resulted in restoration of visual acuity in immunocompromised retinal degenerate (RD) animal models after transplantation. Further assessment of their clinical potential requires evaluation in immunocompetent RD disease models with effective immune suppression. We characterized safety and efficacy profiles of both donor tissues and prospective immunosuppressive treatments in vitro; and in vivo in immunocompetent RD rats (strain SD-foxn1 Tg(S334ter)3Lav).

Methods: Retinal identity of ROs was validated by histology, flow cytometry and gene expression profiling, and their immunogenicity to sensitized human immune cells was measured by mixed lymphocyte reactions (MLR). We measured the effect of RO exposure for 1-4 weeks to therapeutic concentrations of our immunosuppressant drugs of choice on gene expression and metabolic function using quantitative PCR (qPCR) and functional and structural fluorescence lifetime imaging (FLIM), respectively. Immunocompetent RD graft recipients were immunosuppressed by implanted tacrolimus (TAC) pellets and mycophenolate mofetil (MMF) in food. In vivo, LCMS aided assessments of drug pharmacodynamics. Flow cytometry immunophenotyping and assay of post-surgery cytokines were used to assess and monitor drug efficacy. Retinal transplants were imaged in situ using optical coherence tomography (OCT) at defined time points post-surgery. Visual function was assessed by optokinetic tests (OKT) and superior colliculus electrophysiology recording. At study endpoints, immune cell infiltration and donor photoreceptor engraftment into host retinal architecture was evaluated by immunohistochemistry.

Results: Immunosuppressive drugs have no negative effects on RO development and metabolism in vitro; and low alloreactivity of ROs determined by MLR may be predictive to that of human graft recipients. In vivo, minimum effective dosing ranges of TAC and MMF were determined. We characterized the mechanisms and critical immune populations implicated in rejection; and subsequently demonstrated their effective suppression in our xenograft RD model. OKT measured significant visual improvement after RO transplantation. Transplants developed most retinal cell types including photoreceptors; and integrated with the host retina. However, immunosuppression induced higher sensitivity to ketamine anesthesia.

Conclusions: This study proves the concept that immunosuppression is likely tolerable in retinal transplantation and human stem cell therapy for retinal degeneration patients.

背景:富含光感受器(PR)的视网膜类器官(RO)片(人类胚胎干细胞[hESC]衍生的ROs)在免疫受损的视网膜变性(RD)动物模型中移植后可恢复视力。要进一步评估它们的临床潜力,需要在具有有效免疫抑制的免疫功能健全的RD疾病模型中进行评估。我们对供体组织和前瞻性免疫抑制治疗在体外和免疫功能正常的 RD 大鼠(品系 SD-foxn1 Tg(S334ter)3Lav)体内的安全性和有效性进行了鉴定:方法:通过组织学、流式细胞术和基因表达谱分析验证 RO 的视网膜特性,并通过混合淋巴细胞反应 (MLR) 测定它们对致敏人类免疫细胞的免疫原性。我们使用定量 PCR(qPCR)和功能与结构荧光寿命成像(FLIM)分别测量了 RO 暴露于治疗浓度的免疫抑制剂 1-4 周对基因表达和代谢功能的影响。通过植入他克莫司(TAC)颗粒和食物中的霉酚酸酯(MMF)对免疫功能正常的 RD 移植受体进行免疫抑制。在体内,LCMS 可帮助评估药物的药效学。流式细胞术免疫分型和手术后细胞因子检测用于评估和监测药物疗效。在手术后的规定时间点,使用光学相干断层扫描(OCT)对视网膜移植进行原位成像。视功能通过光动能测试(OKT)和上丘电生理记录进行评估。在研究终点,免疫组化对免疫细胞浸润和供体感光细胞移植到宿主视网膜结构进行评估:结果:免疫抑制药物在体外对RO的发育和代谢没有负面影响;通过MLR测定的RO的低异体反应性可能与人类移植受体的异体反应性具有预测性。在体内,确定了TAC和MMF的最小有效剂量范围。我们确定了与排斥反应有关的机制和关键免疫群体,并随后在异种移植 RD 模型中证明了它们的有效抑制作用。OKT测得RO移植后视力明显改善。移植体发育了包括光感受器在内的大多数视网膜细胞类型,并与宿主视网膜融为一体。然而,免疫抑制诱导了对氯胺酮麻醉更高的敏感性:这项研究证明了一个概念,即视网膜移植和人类干细胞疗法治疗视网膜变性患者时,免疫抑制是可以耐受的。
{"title":"Effect of immunosuppression on hESC-derived retina organoids in vitro and in vivo.","authors":"Robert Sims, Bin Lin, Yuntian Xue, Raghda Fouda, Bryce T McLelland, Gabriel Nistor, Hans S Keirstead, Andrew W Browne, Magdalene J Seiler","doi":"10.1186/s13287-025-04271-z","DOIUrl":"10.1186/s13287-025-04271-z","url":null,"abstract":"<p><strong>Background: </strong>Photoreceptor (PR) enriched retinal organoid (RO) sheets (human embryonic stem cell [hESC]-derived ROs) resulted in restoration of visual acuity in immunocompromised retinal degenerate (RD) animal models after transplantation. Further assessment of their clinical potential requires evaluation in immunocompetent RD disease models with effective immune suppression. We characterized safety and efficacy profiles of both donor tissues and prospective immunosuppressive treatments in vitro; and in vivo in immunocompetent RD rats (strain SD-foxn1 Tg(S334ter)3Lav).</p><p><strong>Methods: </strong>Retinal identity of ROs was validated by histology, flow cytometry and gene expression profiling, and their immunogenicity to sensitized human immune cells was measured by mixed lymphocyte reactions (MLR). We measured the effect of RO exposure for 1-4 weeks to therapeutic concentrations of our immunosuppressant drugs of choice on gene expression and metabolic function using quantitative PCR (qPCR) and functional and structural fluorescence lifetime imaging (FLIM), respectively. Immunocompetent RD graft recipients were immunosuppressed by implanted tacrolimus (TAC) pellets and mycophenolate mofetil (MMF) in food. In vivo, LCMS aided assessments of drug pharmacodynamics. Flow cytometry immunophenotyping and assay of post-surgery cytokines were used to assess and monitor drug efficacy. Retinal transplants were imaged in situ using optical coherence tomography (OCT) at defined time points post-surgery. Visual function was assessed by optokinetic tests (OKT) and superior colliculus electrophysiology recording. At study endpoints, immune cell infiltration and donor photoreceptor engraftment into host retinal architecture was evaluated by immunohistochemistry.</p><p><strong>Results: </strong>Immunosuppressive drugs have no negative effects on RO development and metabolism in vitro; and low alloreactivity of ROs determined by MLR may be predictive to that of human graft recipients. In vivo, minimum effective dosing ranges of TAC and MMF were determined. We characterized the mechanisms and critical immune populations implicated in rejection; and subsequently demonstrated their effective suppression in our xenograft RD model. OKT measured significant visual improvement after RO transplantation. Transplants developed most retinal cell types including photoreceptors; and integrated with the host retina. However, immunosuppression induced higher sensitivity to ketamine anesthesia.</p><p><strong>Conclusions: </strong>This study proves the concept that immunosuppression is likely tolerable in retinal transplantation and human stem cell therapy for retinal degeneration patients.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"165"},"PeriodicalIF":7.1,"publicationDate":"2025-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11972460/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143789024","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
Consecutive intrabronchial administration of Wharton's jelly-derived mesenchymal stromal cells in ECMO-supported pediatric patients with end-stage interstitial lung disease: a safety and feasibility study (CIBA method).
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-04-05 DOI: 10.1186/s13287-025-04289-3
Nerea Dominguez-Pinilla, Luis Ignacio González-Granado, Aitor Gonzaga, María López Diaz, Cecilia Castellano Yáñez, Clara Aymerich, Xabier Freire, Olga Ordoñez, Álvaro Gimeno Diaz de Atauri, María Salomé Albi Rodríguez, Elisa Martínez López, Rodrigo Iñiguez, Olga Serrano Garrote, Almudena Castro Frontiñán, Etelvina Andreu, Ana María Gutierrez-Vilchez, Marga Anton-Bonete, Gema Martinez-Navarrete, Nerea Castillo-Flores, Cristina Prat-Vidal, Margarita Blanco, Rocío Morante Valverde, Eduardo Fernandez, Sergi Querol, Luis Manuel Hernández-Blasco, Sylvia Belda-Hofheinz, Bernat Soria

Background: Patients ineligible for lung transplant with end-stage Interstitial Lung Disease (ILD) on Extra-Corporeal Membrane Oxygenation (ECMO) face an appalling prognosis with limited therapeutic options. Due to the beneficial effect of Mesenchymal Stromal Cells (MSC) on inflammatory, immunological and infectious diseases, cell therapy has been proposed as an option, but administration is hampered by the ECMO.

Methods: Cryopreserved Wharton-jelly derived MSC (WJ-MSC) were conveniently diluted and directly applied consecutively on each lobule (5,1 ml = 107 cells) at a continuous slow rate infused over one hour via flexible bronchoscopy (Consecutive IntraBronchial Administration method, CIBA method).

Results: Intrabronchial administration of MSC to a patient on ECMO was well tolerated by the patient even though it did not reverse the patient's ILD. This manuscript presents preliminary evidence from ongoing clinical trials program on Cell Therapy of Inflammatory, Immune and Infectious Diseases and, to our knowledge, is the first report of intrabronchial administration of MSC in a paediatric ECMO patient with ILD. Even more, MSC administered by this method do not reach the systemic circulation and do get blocked on ECMO membrane.

Conclusions: Direct intrabronchial administration of MSC in a patient on ECMO is feasible and safe, and may be a new avenue to be assayed in ECMO patients with inflammatory, immunological and infectious diseases of the lung.

背景:不符合肺移植条件的终末期间质性肺病(ILD)患者在接受体外膜肺氧合(ECMO)治疗后,预后极差,但治疗方案有限。由于间充质干细胞(MSC)对炎症、免疫和感染性疾病的有益作用,细胞疗法已被提出作为一种选择,但其应用受到 ECMO 的阻碍:方法:将冷冻保存的沃顿果冻提取的间充质干细胞(WJ-MSC)稀释后,通过柔性支气管镜(连续支气管内给药法,CIBA 法),以连续缓慢的速度在一小时内将其直接应用于每个小叶(5.1 毫升 = 107 个细胞):结果:为一名接受 ECMO 治疗的患者进行间充质干细胞支气管内给药,尽管未能逆转患者的 ILD,但患者的耐受性良好。据我们所知,这是第一份在患有 ILD 的儿科 ECMO 患者中进行间充质干细胞支气管内给药的报告。此外,通过这种方法给药的间充质干细胞不会进入全身循环,也不会在 ECMO 膜上受阻:在 ECMO 患者体内直接经支气管给药间充质干细胞是可行且安全的,这可能是对患有肺部炎症、免疫学和感染性疾病的 ECMO 患者进行检测的新途径。
{"title":"Consecutive intrabronchial administration of Wharton's jelly-derived mesenchymal stromal cells in ECMO-supported pediatric patients with end-stage interstitial lung disease: a safety and feasibility study (CIBA method).","authors":"Nerea Dominguez-Pinilla, Luis Ignacio González-Granado, Aitor Gonzaga, María López Diaz, Cecilia Castellano Yáñez, Clara Aymerich, Xabier Freire, Olga Ordoñez, Álvaro Gimeno Diaz de Atauri, María Salomé Albi Rodríguez, Elisa Martínez López, Rodrigo Iñiguez, Olga Serrano Garrote, Almudena Castro Frontiñán, Etelvina Andreu, Ana María Gutierrez-Vilchez, Marga Anton-Bonete, Gema Martinez-Navarrete, Nerea Castillo-Flores, Cristina Prat-Vidal, Margarita Blanco, Rocío Morante Valverde, Eduardo Fernandez, Sergi Querol, Luis Manuel Hernández-Blasco, Sylvia Belda-Hofheinz, Bernat Soria","doi":"10.1186/s13287-025-04289-3","DOIUrl":"10.1186/s13287-025-04289-3","url":null,"abstract":"<p><strong>Background: </strong>Patients ineligible for lung transplant with end-stage Interstitial Lung Disease (ILD) on Extra-Corporeal Membrane Oxygenation (ECMO) face an appalling prognosis with limited therapeutic options. Due to the beneficial effect of Mesenchymal Stromal Cells (MSC) on inflammatory, immunological and infectious diseases, cell therapy has been proposed as an option, but administration is hampered by the ECMO.</p><p><strong>Methods: </strong>Cryopreserved Wharton-jelly derived MSC (WJ-MSC) were conveniently diluted and directly applied consecutively on each lobule (5,1 ml = 10<sup>7</sup> cells) at a continuous slow rate infused over one hour via flexible bronchoscopy (Consecutive IntraBronchial Administration method, CIBA method).</p><p><strong>Results: </strong>Intrabronchial administration of MSC to a patient on ECMO was well tolerated by the patient even though it did not reverse the patient's ILD. This manuscript presents preliminary evidence from ongoing clinical trials program on Cell Therapy of Inflammatory, Immune and Infectious Diseases and, to our knowledge, is the first report of intrabronchial administration of MSC in a paediatric ECMO patient with ILD. Even more, MSC administered by this method do not reach the systemic circulation and do get blocked on ECMO membrane.</p><p><strong>Conclusions: </strong>Direct intrabronchial administration of MSC in a patient on ECMO is feasible and safe, and may be a new avenue to be assayed in ECMO patients with inflammatory, immunological and infectious diseases of the lung.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"164"},"PeriodicalIF":7.1,"publicationDate":"2025-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11972491/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143789087","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
Interactions among Merlin, Arkadia, and SKOR2 mediate NF2-associated human Schwann cell proliferation.
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-04-05 DOI: 10.1186/s13287-025-04281-x
Pei-Ciao Tang, Seyoung Um, Anderson B Mayfield, Olena R Bracho, Christian Del Castillo, Christine T Dinh, Derek M Dykxhoorn, Xue Zhong Liu

Background: NF2-related Schwannomatosis (previously referred to as Neurofibromatosis Type 2, or NF2) is a genetic-associated disease resulting from mutations in the gene, NF2. NF2 encodes the Merlin protein, which acts as a tumor suppressor. Bilateral vestibular schwannoma (VS) is a hallmark of NF2. Although the exactly molecular mechanism mediating NF2-driven schwannomatosis is not fully understood, it is known that defective Merlin protein functionality leads to abnormal cell proliferation.

Methods: Herein, we utilized a human induced pluripotent stem cell (hiPSC)-based Schwann cell (SC) model to investigate the role of Merlin in human SCs. SCs were derived from hiPSCs carrying a NF2 mutation (c.191 T > C; p. L64P), its isogenic wild-type control cell line, and a NF2 patient-derived hiPSC line. Phenotypes were determined via immunocytochemistry and various bioassays. Different proteins interacting with Merlin in wild-type and NF2 mutation SCs were identified using co-immunoprecipitation followed by mass spectrometry.

Results: SC derived from NF2L64P hiPSCs showed significantly higher proliferation and abnormal morphology compared to NF2WT SCs. Phenotypes that could be restored by wildtype NF2 overexpression. Interactome profiling of Merlin (NF2) in SCs derived from NF2WT- and NF2L64P- hiPCSs identified differential protein binding levels. Among identified proteins, we validated the interaction among Merlin, an E3 ubiquitin ligase (Arkadia), and a SKI family co-repressor (SKOR2). This complex plays a significant role for this interaction in SC proliferation. Our findings were further validated by SCs derived from the patient-derived hiPSCs carrying a deletion in the chromosome 22 which spans the NF2 gene.

Conclusions: Our results presented a hiPSC-derived SC system for SC-related disease modeling and established a new model in which Merlin interacts with Arkadia and SKOR2. This interaction is required for the proper cell proliferation in human SCs.

背景:NF2相关许旺瘤病(以前称为神经纤维瘤病2型,或NF2)是一种由NF2基因突变引起的遗传相关性疾病。NF2 编码 Merlin 蛋白,它是一种肿瘤抑制因子。双侧前庭分裂瘤(VS)是 NF2 的特征。方法:在此,我们利用基于人诱导多能干细胞(hiPSC)的许旺细胞(SC)模型来研究 Merlin 在人 SC 中的作用。SC来源于携带NF2突变(c.191 T > C; p. L64P)的hiPSC、其同源野生型对照细胞系和NF2患者来源的hiPSC系。表型是通过免疫细胞化学和各种生物测定确定的。通过共免疫沉淀和质谱分析,确定了野生型和 NF2 突变 SC 中与 Merlin 相互作用的不同蛋白质:结果:与NF2WT SCs相比,NF2L64P hiPSCs衍生的SC显示出明显更高的增殖和异常形态。野生型 NF2 过表达可恢复这些表型。在源自 NF2WT- 和 NF2L64P- hiPCSs 的 SCs 中,Merlin(NF2)的相互作用组图谱确定了不同的蛋白结合水平。在确定的蛋白质中,我们验证了 Merlin、E3 泛素连接酶(Arkadia)和 SKI 家族共抑制因子(SKOR2)之间的相互作用。这一复合体在SC增殖过程中发挥了重要作用。我们的研究结果得到了来自患者的hiPSCs的SCs的进一步验证,这些hiPSCs携带有NF2基因的22号染色体缺失:我们的研究结果提出了一种用于SC相关疾病建模的hiPSC衍生SC系统,并建立了一种新的模型,在该模型中,Merlin与Arkadia和SKOR2相互作用。这种相互作用是人类 SC 细胞正常增殖所必需的。
{"title":"Interactions among Merlin, Arkadia, and SKOR2 mediate NF2-associated human Schwann cell proliferation.","authors":"Pei-Ciao Tang, Seyoung Um, Anderson B Mayfield, Olena R Bracho, Christian Del Castillo, Christine T Dinh, Derek M Dykxhoorn, Xue Zhong Liu","doi":"10.1186/s13287-025-04281-x","DOIUrl":"10.1186/s13287-025-04281-x","url":null,"abstract":"<p><strong>Background: </strong>NF2-related Schwannomatosis (previously referred to as Neurofibromatosis Type 2, or NF2) is a genetic-associated disease resulting from mutations in the gene, NF2. NF2 encodes the Merlin protein, which acts as a tumor suppressor. Bilateral vestibular schwannoma (VS) is a hallmark of NF2. Although the exactly molecular mechanism mediating NF2-driven schwannomatosis is not fully understood, it is known that defective Merlin protein functionality leads to abnormal cell proliferation.</p><p><strong>Methods: </strong>Herein, we utilized a human induced pluripotent stem cell (hiPSC)-based Schwann cell (SC) model to investigate the role of Merlin in human SCs. SCs were derived from hiPSCs carrying a NF2 mutation (c.191 T > C; p. L64P), its isogenic wild-type control cell line, and a NF2 patient-derived hiPSC line. Phenotypes were determined via immunocytochemistry and various bioassays. Different proteins interacting with Merlin in wild-type and NF2 mutation SCs were identified using co-immunoprecipitation followed by mass spectrometry.</p><p><strong>Results: </strong>SC derived from NF2<sup>L64P</sup> hiPSCs showed significantly higher proliferation and abnormal morphology compared to NF2<sup>WT</sup> SCs. Phenotypes that could be restored by wildtype NF2 overexpression. Interactome profiling of Merlin (NF2) in SCs derived from NF2<sup>WT</sup>- and NF2<sup>L64P</sup>- hiPCSs identified differential protein binding levels. Among identified proteins, we validated the interaction among Merlin, an E3 ubiquitin ligase (Arkadia), and a SKI family co-repressor (SKOR2). This complex plays a significant role for this interaction in SC proliferation. Our findings were further validated by SCs derived from the patient-derived hiPSCs carrying a deletion in the chromosome 22 which spans the NF2 gene.</p><p><strong>Conclusions: </strong>Our results presented a hiPSC-derived SC system for SC-related disease modeling and established a new model in which Merlin interacts with Arkadia and SKOR2. This interaction is required for the proper cell proliferation in human SCs.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"163"},"PeriodicalIF":7.1,"publicationDate":"2025-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11972516/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143789025","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
Determination of the miRNA profile of extracellular vesicles from equine mesenchymal stem cells after different treatments.
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-04-05 DOI: 10.1186/s13287-025-04287-5
Michele C Klymiuk, Julia Speer, Isabelle De Marco, Mohamed I Elashry, Manuela Heimann, Sabine Wenisch, Stefan Arnhold

Background: Osteoarthritis (OA) is a common and incurable disease in humans and animals. To gain a better understanding of the pathogenesis and identify potential treatments, miRNAs will be extracted and analysed from extracellular vesicles (EVs) of equine adipose derived mesenchymal stem cells (AdMSCs).

Methods: For this purpose we cultivated and pretreated AdMSCs under different conditions: interleukin 1β, shock wave, chondrogenic differentiation, chondrogenic differentiation under hypoxia, or after senescence. After treatment, EVs were harvested from the cell culture supernatants. Next-generation sequencing (NGS) was used to sequence the miRNAs from the EVs.

Results: A total of 89 miRNAs whose expression was significantly altered compared with that of an untreated negative control were identified. On average, 53 miRNAs were upregulated and 6 miRNAs were downregulated. Among others, the miRNAs eca-miR-101, eca-miR-143, eca-miR-145, eca-miR-146a, eca-miR-27a, eca-miR-29b, eca-miR-93, eca-miR-98, and eca-miR-221 were significantly increased after the stimulations, which, as known anti-inflammatory miRNAs, could be candidates for therapeutic use in the treatment of OA.

Conclusion: These results lay the foundation for further research into the significance and efficacy of these miRNAs so that this knowledge can be improved in further experiments and, ideally, translated into therapeutic use.

{"title":"Determination of the miRNA profile of extracellular vesicles from equine mesenchymal stem cells after different treatments.","authors":"Michele C Klymiuk, Julia Speer, Isabelle De Marco, Mohamed I Elashry, Manuela Heimann, Sabine Wenisch, Stefan Arnhold","doi":"10.1186/s13287-025-04287-5","DOIUrl":"10.1186/s13287-025-04287-5","url":null,"abstract":"<p><strong>Background: </strong>Osteoarthritis (OA) is a common and incurable disease in humans and animals. To gain a better understanding of the pathogenesis and identify potential treatments, miRNAs will be extracted and analysed from extracellular vesicles (EVs) of equine adipose derived mesenchymal stem cells (AdMSCs).</p><p><strong>Methods: </strong>For this purpose we cultivated and pretreated AdMSCs under different conditions: interleukin 1β, shock wave, chondrogenic differentiation, chondrogenic differentiation under hypoxia, or after senescence. After treatment, EVs were harvested from the cell culture supernatants. Next-generation sequencing (NGS) was used to sequence the miRNAs from the EVs.</p><p><strong>Results: </strong>A total of 89 miRNAs whose expression was significantly altered compared with that of an untreated negative control were identified. On average, 53 miRNAs were upregulated and 6 miRNAs were downregulated. Among others, the miRNAs eca-miR-101, eca-miR-143, eca-miR-145, eca-miR-146a, eca-miR-27a, eca-miR-29b, eca-miR-93, eca-miR-98, and eca-miR-221 were significantly increased after the stimulations, which, as known anti-inflammatory miRNAs, could be candidates for therapeutic use in the treatment of OA.</p><p><strong>Conclusion: </strong>These results lay the foundation for further research into the significance and efficacy of these miRNAs so that this knowledge can be improved in further experiments and, ideally, translated into therapeutic use.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"162"},"PeriodicalIF":7.1,"publicationDate":"2025-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11972531/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143789022","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
Differential efficacy of olfactory neurospheres from deviated nasal septum and chronic rhinosinusitis patients in regenerating olfactory epithelium.
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-04-05 DOI: 10.1186/s13287-025-04270-0
Rong-San Jiang, Chiang-Wen Lee, Yu-Hsuan Lin, Jing-Jie Wang, Jia-Bin Liao, Kuo-Ti Peng, Yao-Chang Chiang, Pei-Ling Chi

Background: Olfactory epithelial stem cells hold significant potential for treating olfactory dysfunction by facilitating tissue maintenance and repair. Understanding the inherent qualities of these stem cells is crucial for optimizing their therapeutic efficacy.

Methods: Olfactory epithelial samples were collected from patients with deviated nasal septum (DNS) and chronic rhinosinusitis (CRS). These were cultured to form olfactory neurospheres (ONS), which were then analyzed for neural stem cell markers, neurotrophic factor production, and their ability to differentiate into olfactory sensory neurons (OSNs). The regenerative efficacy of these ONS was tested in a methimazole-induced hyposmic mouse model, with the effects on cellular senescence, apoptosis, and proliferation in the olfactory epithelium assessed.

Results: Both DNS- and CRS-derived ONS exhibited neural stem cell characteristics. DNS-ONS displayed superior self-renewal capacity and higher neurotrophic factor production compared to CRS-ONS, which showed impaired OSN maturation and lower neurotrophic factor levels. In vivo, DNS-ONS were more effective in restoring olfaction, as evidenced by reduced cellular senescence, decreased apoptosis, and increased cell proliferation in the OE of methimazole-induced hyposmic mice.

Conclusions: These findings highlight the importance of selecting the appropriate ONS source for therapeutic applications, with DNS-ONS showing greater promise for olfactory epithelium repair and olfactory function restoration.

{"title":"Differential efficacy of olfactory neurospheres from deviated nasal septum and chronic rhinosinusitis patients in regenerating olfactory epithelium.","authors":"Rong-San Jiang, Chiang-Wen Lee, Yu-Hsuan Lin, Jing-Jie Wang, Jia-Bin Liao, Kuo-Ti Peng, Yao-Chang Chiang, Pei-Ling Chi","doi":"10.1186/s13287-025-04270-0","DOIUrl":"10.1186/s13287-025-04270-0","url":null,"abstract":"<p><strong>Background: </strong>Olfactory epithelial stem cells hold significant potential for treating olfactory dysfunction by facilitating tissue maintenance and repair. Understanding the inherent qualities of these stem cells is crucial for optimizing their therapeutic efficacy.</p><p><strong>Methods: </strong>Olfactory epithelial samples were collected from patients with deviated nasal septum (DNS) and chronic rhinosinusitis (CRS). These were cultured to form olfactory neurospheres (ONS), which were then analyzed for neural stem cell markers, neurotrophic factor production, and their ability to differentiate into olfactory sensory neurons (OSNs). The regenerative efficacy of these ONS was tested in a methimazole-induced hyposmic mouse model, with the effects on cellular senescence, apoptosis, and proliferation in the olfactory epithelium assessed.</p><p><strong>Results: </strong>Both DNS- and CRS-derived ONS exhibited neural stem cell characteristics. DNS-ONS displayed superior self-renewal capacity and higher neurotrophic factor production compared to CRS-ONS, which showed impaired OSN maturation and lower neurotrophic factor levels. In vivo, DNS-ONS were more effective in restoring olfaction, as evidenced by reduced cellular senescence, decreased apoptosis, and increased cell proliferation in the OE of methimazole-induced hyposmic mice.</p><p><strong>Conclusions: </strong>These findings highlight the importance of selecting the appropriate ONS source for therapeutic applications, with DNS-ONS showing greater promise for olfactory epithelium repair and olfactory function restoration.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"166"},"PeriodicalIF":7.1,"publicationDate":"2025-04-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11972463/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143789023","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
Cellular mechanical memory: a potential tool for mesenchymal stem cell-based therapy. 细胞机械记忆:间充质干细胞疗法的潜在工具。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-03-31 DOI: 10.1186/s13287-025-04249-x
Sanjay Kumar Kureel, Rosario Maroto, Kristen Davis, Michael Sheetz

Recent studies have shown that mechanical properties such as extracellular matrix stiffness, fluid flow, weight loading, compression, and stretching can affect cellular functions. Some examples of cell responses to mechanical properties could be the migration of cancer cells from rigid to soft surfaces or the differentiation of fibroblasts into myofibroblasts. Cellular responses to mechanical changes can modify the insertion of proteins in the extracellular matrix (ECM), causing an increase in tissue stiffness with functional consequences. In general, mechanical and physical factors can affect any kind of cell phenotype in culture conditions and in vivo tissues. Cells sense mechanical stimuli by applying force and restructuring their shape and functions in response to the resistance of the stimuli. Furthermore, mechanical triggers can develop a "memory" for altering cellular plasticity and adaptation. This phenomenon is called cellular mechanical memory (CMM), a singular feature of mesenchymal stem cells (MSCs). Controlled targeting of CMM may resolve the scarcity of viable cells needed for cell based therapy (CBT) and implement studies concerning cancer research, fibrosis, and senescence. This review focusses on cells from the mesodermal lineage, such as MSCs, fibroblasts and chondrocytes, and the role of CMM as a potential target for CBT.

{"title":"Cellular mechanical memory: a potential tool for mesenchymal stem cell-based therapy.","authors":"Sanjay Kumar Kureel, Rosario Maroto, Kristen Davis, Michael Sheetz","doi":"10.1186/s13287-025-04249-x","DOIUrl":"10.1186/s13287-025-04249-x","url":null,"abstract":"<p><p>Recent studies have shown that mechanical properties such as extracellular matrix stiffness, fluid flow, weight loading, compression, and stretching can affect cellular functions. Some examples of cell responses to mechanical properties could be the migration of cancer cells from rigid to soft surfaces or the differentiation of fibroblasts into myofibroblasts. Cellular responses to mechanical changes can modify the insertion of proteins in the extracellular matrix (ECM), causing an increase in tissue stiffness with functional consequences. In general, mechanical and physical factors can affect any kind of cell phenotype in culture conditions and in vivo tissues. Cells sense mechanical stimuli by applying force and restructuring their shape and functions in response to the resistance of the stimuli. Furthermore, mechanical triggers can develop a \"memory\" for altering cellular plasticity and adaptation. This phenomenon is called cellular mechanical memory (CMM), a singular feature of mesenchymal stem cells (MSCs). Controlled targeting of CMM may resolve the scarcity of viable cells needed for cell based therapy (CBT) and implement studies concerning cancer research, fibrosis, and senescence. This review focusses on cells from the mesodermal lineage, such as MSCs, fibroblasts and chondrocytes, and the role of CMM as a potential target for CBT.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"159"},"PeriodicalIF":7.1,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11960036/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754513","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
Tremella polysaccharide microneedles loaded with magnetic dental pulp stem cell intracellular vesicles used for androgenic alopecia. 含磁性牙髓干细胞胞内囊泡的 Tremella 多糖微针用于治疗雄激素性脱发。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-03-31 DOI: 10.1186/s13287-025-04219-3
Yonghao Liu, Heng Zhou, Pengcheng Pang, Ye Liu, Jieying Situ, Junnan Wang, Yan He, Qingsong Ye

Background: Mesenchymal stem cell exosomes are the most extensively researched type of Extracellular vesicles (EVs) that offer novel avenues for hair regeneration. However, their use in the field of hair regeneration was limited by their poor production of exosomes. It has been discovered that intracellular vesicles (IVs), which are produced at a higher rate than exosomes, play a comparable biological purpose. As a result, we developed HTMI-MN, a microneedle that uses tremella, a type of Tremella polysaccharide (TPS), and hyaluronic acid (HA) as matrix materials. It has magnetic intracellular vesicles (Mag-IVs), which work in concert to treat androgenic alopecia (AGA) and encourage hair growth.

Methods: After characterization of the Mag-IVs, we evaluated the effect on angiogenesis by scratch assay, angiogenesis assay, Western Blot and ELISA assay. In addition, we tested the protective effect of Human hair papillary cells (HHDPCs) by CCK-8 method, Western Blot and flow cytometry. Finally, the effects of tremella polysaccharide on M1/M2 polarization of macrophages were detected by fluorescence staining, Western Blot and flow cytometry. AGA model was established in vivo by DHT, and treatment was given by microneedle injection.

Results: Our study found that Mag-IVs have greater power to promote angiogenesis and protect HHDPCs from apoptosis compared to other vesicles. Besides, tremella polysaccharide can make the transformation of macrophages to anti-inflammatory phenotype. Taken together, in vivo experiments showed that hair regeneration was faster in HTMI-MN-treated mice.

Conclusion: These results indicate that Mag-IVs and tremella polysaccharide can synergistically improve the hair microenvironment, which has a promising future for AGA treatment.

{"title":"Tremella polysaccharide microneedles loaded with magnetic dental pulp stem cell intracellular vesicles used for androgenic alopecia.","authors":"Yonghao Liu, Heng Zhou, Pengcheng Pang, Ye Liu, Jieying Situ, Junnan Wang, Yan He, Qingsong Ye","doi":"10.1186/s13287-025-04219-3","DOIUrl":"10.1186/s13287-025-04219-3","url":null,"abstract":"<p><strong>Background: </strong>Mesenchymal stem cell exosomes are the most extensively researched type of Extracellular vesicles (EVs) that offer novel avenues for hair regeneration. However, their use in the field of hair regeneration was limited by their poor production of exosomes. It has been discovered that intracellular vesicles (IVs), which are produced at a higher rate than exosomes, play a comparable biological purpose. As a result, we developed HTMI-MN, a microneedle that uses tremella, a type of Tremella polysaccharide (TPS), and hyaluronic acid (HA) as matrix materials. It has magnetic intracellular vesicles (Mag-IVs), which work in concert to treat androgenic alopecia (AGA) and encourage hair growth.</p><p><strong>Methods: </strong>After characterization of the Mag-IVs, we evaluated the effect on angiogenesis by scratch assay, angiogenesis assay, Western Blot and ELISA assay. In addition, we tested the protective effect of Human hair papillary cells (HHDPCs) by CCK-8 method, Western Blot and flow cytometry. Finally, the effects of tremella polysaccharide on M1/M2 polarization of macrophages were detected by fluorescence staining, Western Blot and flow cytometry. AGA model was established in vivo by DHT, and treatment was given by microneedle injection.</p><p><strong>Results: </strong>Our study found that Mag-IVs have greater power to promote angiogenesis and protect HHDPCs from apoptosis compared to other vesicles. Besides, tremella polysaccharide can make the transformation of macrophages to anti-inflammatory phenotype. Taken together, in vivo experiments showed that hair regeneration was faster in HTMI-MN-treated mice.</p><p><strong>Conclusion: </strong>These results indicate that Mag-IVs and tremella polysaccharide can synergistically improve the hair microenvironment, which has a promising future for AGA treatment.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"161"},"PeriodicalIF":7.1,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11959820/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754514","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
A perfect islet: reviewing recent protocol developments and proposing strategies for stem cell derived functional pancreatic islets.
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-03-31 DOI: 10.1186/s13287-025-04293-7
Sujitha Sali, Leen Azzam, Taraf Jaro, Ahmed Ali Gebril Ali, Ali Mardini, Omar Al-Dajani, Shahryar Khattak, Alexandra E Butler, Juberiya M Azeez, Manjula Nandakumar

The search for an effective cell replacement therapy for diabetes has driven the development of "perfect" pancreatic islets from human pluripotent stem cells (hPSCs). These hPSC-derived pancreatic islet-like β cells can overcome the limitations for disease modelling, drug development and transplantation therapies in diabetes. Nevertheless, challenges remain in generating fully functional and mature β cells from hPSCs. This review underscores the significant efforts made by researchers to optimize various differentiation protocols aimed at enhancing the efficiency and quality of hPSC-derived pancreatic islets and proposes methods for their improvement. By emulating the natural developmental processes of pancreatic embryogenesis, specific growth factors, signaling molecules and culture conditions are employed to guide hPSCs towards the formation of mature β cells capable of secreting insulin in response to glucose. However, the efficiency of these protocols varies greatly among different human embryonic stem cell (hESC) and induced pluripotent stem cell (hiPSC) lines. This variability poses a particular challenge for generating patient-specific β cells. Despite recent advancements, the ultimate goal remains to develop a highly efficient directed differentiation protocol that is applicable across all genetic backgrounds of hPSCs. Although progress has been made, further research is required to optimize the protocols and characterization methods that could ensure the safety and efficacy of hPSC-derived pancreatic islets before they can be utilized in clinical settings.

{"title":"A perfect islet: reviewing recent protocol developments and proposing strategies for stem cell derived functional pancreatic islets.","authors":"Sujitha Sali, Leen Azzam, Taraf Jaro, Ahmed Ali Gebril Ali, Ali Mardini, Omar Al-Dajani, Shahryar Khattak, Alexandra E Butler, Juberiya M Azeez, Manjula Nandakumar","doi":"10.1186/s13287-025-04293-7","DOIUrl":"10.1186/s13287-025-04293-7","url":null,"abstract":"<p><p>The search for an effective cell replacement therapy for diabetes has driven the development of \"perfect\" pancreatic islets from human pluripotent stem cells (hPSCs). These hPSC-derived pancreatic islet-like β cells can overcome the limitations for disease modelling, drug development and transplantation therapies in diabetes. Nevertheless, challenges remain in generating fully functional and mature β cells from hPSCs. This review underscores the significant efforts made by researchers to optimize various differentiation protocols aimed at enhancing the efficiency and quality of hPSC-derived pancreatic islets and proposes methods for their improvement. By emulating the natural developmental processes of pancreatic embryogenesis, specific growth factors, signaling molecules and culture conditions are employed to guide hPSCs towards the formation of mature β cells capable of secreting insulin in response to glucose. However, the efficiency of these protocols varies greatly among different human embryonic stem cell (hESC) and induced pluripotent stem cell (hiPSC) lines. This variability poses a particular challenge for generating patient-specific β cells. Despite recent advancements, the ultimate goal remains to develop a highly efficient directed differentiation protocol that is applicable across all genetic backgrounds of hPSCs. Although progress has been made, further research is required to optimize the protocols and characterization methods that could ensure the safety and efficacy of hPSC-derived pancreatic islets before they can be utilized in clinical settings.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"160"},"PeriodicalIF":7.1,"publicationDate":"2025-03-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11959787/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143754491","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 role of the mTOR pathway in breast cancer stem cells (BCSCs): mechanisms and therapeutic potentials.
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2025-03-29 DOI: 10.1186/s13287-025-04218-4
Chen Zhang, Xu Shu, Chuanzheng Yin, Shaobo Hu, Pian Liu

Breast cancer remains the most frequently diagnosed cancer globally, exerting a profound impact on women's health and healthcare systems. Central to its pathogenesis and therapeutic resistance are breast cancer stem cells (BCSCs), which possess unique properties such as self-renewal, differentiation, and resistance to conventional therapies, contributing to tumor initiation, metastasis, and recurrence. This comprehensive review elucidates the pivotal role of the mechanistic target of rapamycin (mTOR) pathway in regulating BCSCs and its implications for breast cancer progression and treatment resistance. We explore the cellular mechanisms by which mTOR influences metastasis, metabolism, autophagy, and ferroptosis in BCSCs, highlighting its contribution to epithelial-to-mesenchymal transition (EMT), metabolic reprogramming, and survival under therapeutic stress. On a molecular level, mTOR interacts with key signaling pathways including PI3K/Akt, Notch, IGF-1R, AMPK, and TGF-β, as well as regulatory proteins and non-coding RNAs, orchestrating a complex network that sustains BCSC properties and mediates chemoresistance and radioresistance. The review further examines various therapeutic strategies targeting the mTOR pathway in BCSCs, encompassing selective PI3K/Akt/mTOR inhibitors, monoclonal antibodies, natural products, and innovative approaches such as nanoparticle-mediated drug delivery. Clinical trials investigating mTOR inhibitors like sirolimus and combination therapies with agents such as everolimus and trastuzumab are discussed, underscoring their potential in eradicating BCSCs and improving patient outcomes. Additionally, natural compounds and repurposed drugs offer promising adjunctive therapies by modulating mTOR activity and targeting BCSC-specific vulnerabilities. In conclusion, targeting the mTOR pathway presents a viable and promising avenue for enhancing breast cancer treatment efficacy by effectively eliminating BCSCs, reducing tumor recurrence, and improving overall patient survival. Continued research and clinical validation of mTOR-targeted therapies are essential to translate these insights into effective clinical interventions, ultimately advancing personalized cancer management and therapeutic outcomes for breast cancer patients.

{"title":"The role of the mTOR pathway in breast cancer stem cells (BCSCs): mechanisms and therapeutic potentials.","authors":"Chen Zhang, Xu Shu, Chuanzheng Yin, Shaobo Hu, Pian Liu","doi":"10.1186/s13287-025-04218-4","DOIUrl":"10.1186/s13287-025-04218-4","url":null,"abstract":"<p><p>Breast cancer remains the most frequently diagnosed cancer globally, exerting a profound impact on women's health and healthcare systems. Central to its pathogenesis and therapeutic resistance are breast cancer stem cells (BCSCs), which possess unique properties such as self-renewal, differentiation, and resistance to conventional therapies, contributing to tumor initiation, metastasis, and recurrence. This comprehensive review elucidates the pivotal role of the mechanistic target of rapamycin (mTOR) pathway in regulating BCSCs and its implications for breast cancer progression and treatment resistance. We explore the cellular mechanisms by which mTOR influences metastasis, metabolism, autophagy, and ferroptosis in BCSCs, highlighting its contribution to epithelial-to-mesenchymal transition (EMT), metabolic reprogramming, and survival under therapeutic stress. On a molecular level, mTOR interacts with key signaling pathways including PI3K/Akt, Notch, IGF-1R, AMPK, and TGF-β, as well as regulatory proteins and non-coding RNAs, orchestrating a complex network that sustains BCSC properties and mediates chemoresistance and radioresistance. The review further examines various therapeutic strategies targeting the mTOR pathway in BCSCs, encompassing selective PI3K/Akt/mTOR inhibitors, monoclonal antibodies, natural products, and innovative approaches such as nanoparticle-mediated drug delivery. Clinical trials investigating mTOR inhibitors like sirolimus and combination therapies with agents such as everolimus and trastuzumab are discussed, underscoring their potential in eradicating BCSCs and improving patient outcomes. Additionally, natural compounds and repurposed drugs offer promising adjunctive therapies by modulating mTOR activity and targeting BCSC-specific vulnerabilities. In conclusion, targeting the mTOR pathway presents a viable and promising avenue for enhancing breast cancer treatment efficacy by effectively eliminating BCSCs, reducing tumor recurrence, and improving overall patient survival. Continued research and clinical validation of mTOR-targeted therapies are essential to translate these insights into effective clinical interventions, ultimately advancing personalized cancer management and therapeutic outcomes for breast cancer patients.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"16 1","pages":"156"},"PeriodicalIF":7.1,"publicationDate":"2025-03-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11954216/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143744034","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