npj Regenerative Medicine最新文献

筛选
英文 中文
Role of umbilical cord mesenchymal stromal cells in skin rejuvenation. 脐带间充质基质细胞在皮肤年轻化中的作用。
IF 7.2 1区 医学
npj Regenerative Medicine Pub Date : 2024-05-10 DOI: 10.1038/s41536-024-00363-1
Le Chang, Wei-Wen Fan, He-Ling Yuan, Xin Liu, Qiang Wang, Guang-Ping Ruan, Xing-Hua Pan, Xiang-Qing Zhu
{"title":"Role of umbilical cord mesenchymal stromal cells in skin rejuvenation.","authors":"Le Chang, Wei-Wen Fan, He-Ling Yuan, Xin Liu, Qiang Wang, Guang-Ping Ruan, Xing-Hua Pan, Xiang-Qing Zhu","doi":"10.1038/s41536-024-00363-1","DOIUrl":"10.1038/s41536-024-00363-1","url":null,"abstract":"<p><p>Aging is the main cause of many degenerative diseases. The skin is the largest and the most intuitive organ that reflects the aging of the body. Under the interaction of endogenous and exogenous factors, there are cumulative changes in the structure, function, and appearance of the skin, which are characterized by decreased synthesis of collagen and elastin, increased wrinkles, relaxation, pigmentation, and other aging characteristics. skin aging is inevitable, but it can be delayed. The successful isolation of mesenchymal stromal cells (MSC) in 1991 has greatly promoted the progress of cell therapy in human diseases. The International Society for Cellular Therapy (ISCT) points out that the MSC is a kind of pluripotent progenitor cells that have self-renewal ability (limited) in vitro and the potential for mesenchymal cell differentiation. This review mainly introduces the role of perinatal umbilical cord-derived MSC(UC-MSC) in the field of skin rejuvenation. An in-depth and systematic understanding of the mechanism of UC-MSCs against skin aging is of great significance for the early realization of the clinical transformation of UC-MSCs. This paper summarized the characteristics of skin aging and summarized the mechanism of UC-MSCs in skin rejuvenation reported in recent years. In order to provide a reference for further research of UC-MSCs to delay skin aging.</p>","PeriodicalId":54236,"journal":{"name":"npj Regenerative Medicine","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2024-05-10","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11087646/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140905187","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Exploiting in silico modelling to enhance translation of liver cell therapies from bench to bedside. 利用硅学建模促进肝细胞疗法从实验室到临床的转化。
IF 7.2 1区 医学
npj Regenerative Medicine Pub Date : 2024-05-09 DOI: 10.1038/s41536-024-00361-3
Candice Ashmore-Harris, Evangelia Antonopoulou, Simon M Finney, Melissa R Vieira, Matthew G Hennessy, Andreas Muench, Wei-Yu Lu, Victoria L Gadd, Alicia J El Haj, Stuart J Forbes, Sarah L Waters
{"title":"Exploiting in silico modelling to enhance translation of liver cell therapies from bench to bedside.","authors":"Candice Ashmore-Harris, Evangelia Antonopoulou, Simon M Finney, Melissa R Vieira, Matthew G Hennessy, Andreas Muench, Wei-Yu Lu, Victoria L Gadd, Alicia J El Haj, Stuart J Forbes, Sarah L Waters","doi":"10.1038/s41536-024-00361-3","DOIUrl":"10.1038/s41536-024-00361-3","url":null,"abstract":"<p><p>Cell therapies are emerging as promising treatments for a range of liver diseases but translational bottlenecks still remain including: securing and assessing the safe and effective delivery of cells to the disease site; ensuring successful cell engraftment and function; and preventing immunogenic responses. Here we highlight three therapies, each utilising a different cell type, at different stages in their clinical translation journey: transplantation of multipotent mesenchymal stromal/signalling cells, hepatocytes and macrophages. To overcome bottlenecks impeding clinical progression, we advocate for wider use of mechanistic in silico modelling approaches. We discuss how in silico approaches, alongside complementary experimental approaches, can enhance our understanding of the mechanisms underlying successful cell delivery and engraftment. Furthermore, such combined theoretical-experimental approaches can be exploited to develop novel therapies, address safety and efficacy challenges, bridge the gap between in vitro and in vivo model systems, and compensate for the inherent differences between animal model systems and humans. We also highlight how in silico model development can result in fewer and more targeted in vivo experiments, thereby reducing preclinical costs and experimental animal numbers and potentially accelerating translation to the clinic. The development of biologically-accurate in silico models that capture the mechanisms underpinning the behaviour of these complex systems must be reinforced by quantitative methods to assess cell survival post-transplant, and we argue that non-invasive in vivo imaging strategies should be routinely integrated into transplant studies.</p>","PeriodicalId":54236,"journal":{"name":"npj Regenerative Medicine","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2024-05-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11081951/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140900219","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Structural, angiogenic, and immune responses influencing myocardial regeneration: a glimpse into the crucible 影响心肌再生的结构、血管生成和免疫反应:熔炉一瞥
IF 7.2 1区 医学
npj Regenerative Medicine Pub Date : 2024-04-30 DOI: 10.1038/s41536-024-00357-z
Basil M. Baccouche, Stefan Elde, Hanjay Wang, Y. Joseph Woo
{"title":"Structural, angiogenic, and immune responses influencing myocardial regeneration: a glimpse into the crucible","authors":"Basil M. Baccouche, Stefan Elde, Hanjay Wang, Y. Joseph Woo","doi":"10.1038/s41536-024-00357-z","DOIUrl":"https://doi.org/10.1038/s41536-024-00357-z","url":null,"abstract":"<p>Complete cardiac regeneration remains an elusive therapeutic goal. Although much attention has been focused on cardiomyocyte proliferation, especially in neonatal mammals, recent investigations have unearthed mechanisms by which non-cardiomyocytes, such as endothelial cells, fibroblasts, macrophages, and other immune cells, play critical roles in modulating the regenerative capacity of the injured heart. The degree to which each of these cell types influence cardiac regeneration, however, remains incompletely understood. This review highlights the roles of these non-cardiomyocytes and their respective contributions to cardiac regeneration, with emphasis on natural heart regeneration after cardiac injury during the neonatal period.</p>","PeriodicalId":54236,"journal":{"name":"npj Regenerative Medicine","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2024-04-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140837530","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Epigenetic mechanisms regulate sex differences in cardiac reparative functions of bone marrow progenitor cells 表观遗传机制调控骨髓祖细胞心脏修复功能的性别差异
IF 7.2 1区 医学
npj Regenerative Medicine Pub Date : 2024-04-29 DOI: 10.1038/s41536-024-00362-2
Charan Thej, Rajika Roy, Zhongjian Cheng, Venkata Naga Srikanth Garikipati, May M. Truongcao, Darukeshwara Joladarashi, Vandana Mallaredy, Maria Cimini, Carolina Gonzalez, Ajit Magadum, Jayashri Ghosh, Cindy Benedict, Walter J. Koch, Raj Kishore
{"title":"Epigenetic mechanisms regulate sex differences in cardiac reparative functions of bone marrow progenitor cells","authors":"Charan Thej, Rajika Roy, Zhongjian Cheng, Venkata Naga Srikanth Garikipati, May M. Truongcao, Darukeshwara Joladarashi, Vandana Mallaredy, Maria Cimini, Carolina Gonzalez, Ajit Magadum, Jayashri Ghosh, Cindy Benedict, Walter J. Koch, Raj Kishore","doi":"10.1038/s41536-024-00362-2","DOIUrl":"https://doi.org/10.1038/s41536-024-00362-2","url":null,"abstract":"<p>Historically, a lower incidence of cardiovascular diseases (CVD) and related deaths in women as compared with men of the same age has been attributed to female sex hormones, particularly estrogen and its receptors. Autologous bone marrow stem cell (BMSC) clinical trials for cardiac cell therapy overwhelmingly included male patients. However, meta-analysis data from these trials suggest a better functional outcome in postmenopausal women as compared with aged-matched men. Mechanisms governing sex-specific cardiac reparative activity in BMSCs, with and without the influence of sex hormones, remain unexplored. To discover these mechanisms, Male (M), female (F), and ovariectomized female (OVX) mice-derived EPCs were subjected to a series of molecular and epigenetic analyses followed by in vivo functional assessments of cardiac repair. F-EPCs and OVX EPCs show a lower inflammatory profile and promote enhanced cardiac reparative activity after intra-cardiac injections in a male mouse model of myocardial infarction (MI). Epigenetic sequencing revealed a marked difference in the occupancy of the gene repressive H3K9me3 mark, particularly at transcription start sites of key angiogenic and proinflammatory genes in M-EPCs compared with F-EPCs and OVX-EPCs. Our study unveiled that functional sex differences in EPCs are, in part, mediated by differential epigenetic regulation of the proinflammatory and anti-angiogenic gene CCL3, orchestrated by the control of H3K9me3 by histone methyltransferase, G9a/Ehmt2. Our research highlights the importance of considering the sex of donor cells for progenitor-based tissue repair.</p>","PeriodicalId":54236,"journal":{"name":"npj Regenerative Medicine","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2024-04-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140837495","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The adult environment promotes the transcriptional maturation of human iPSC-derived muscle grafts 成人环境促进人类 iPSC 衍生肌肉移植物的转录成熟
IF 7.2 1区 医学
npj Regenerative Medicine Pub Date : 2024-04-04 DOI: 10.1038/s41536-024-00360-4
Sarah B. Crist, Karim Azzag, James Kiley, Ilsa Coleman, Alessandro Magli, Rita C. R. Perlingeiro
{"title":"The adult environment promotes the transcriptional maturation of human iPSC-derived muscle grafts","authors":"Sarah B. Crist, Karim Azzag, James Kiley, Ilsa Coleman, Alessandro Magli, Rita C. R. Perlingeiro","doi":"10.1038/s41536-024-00360-4","DOIUrl":"https://doi.org/10.1038/s41536-024-00360-4","url":null,"abstract":"<p>Pluripotent stem cell (PSC)-based cell therapy is an attractive option for the treatment of multiple human disorders, including muscular dystrophies. While in vitro differentiating PSCs can generate large numbers of human lineage-specific tissue, multiple studies evidenced that these cell populations mostly display embryonic/fetal features. We previously demonstrated that transplantation of PSC-derived myogenic progenitors provides long-term engraftment and functional improvement in several dystrophic mouse models, but it remained unknown whether donor-derived myofibers mature to match adult tissue. Here, we transplanted iPAX7 myogenic progenitors into muscles of non-dystrophic and dystrophic mice and compared the transcriptional landscape of human grafts with respective in vitro-differentiated iPAX7 myotubes as well as human skeletal muscle biospecimens. Pairing bulk RNA sequencing with computational deconvolution of human reads, we were able to pinpoint key myogenic changes that occur during the in vitro–to–in vivo transition, confirm developmental maturity, and consequently evaluate their applicability for cell-based therapies.</p>","PeriodicalId":54236,"journal":{"name":"npj Regenerative Medicine","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2024-04-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140579619","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A two-way street – cellular metabolism and myofibroblast contraction 双行道--细胞新陈代谢与肌成纤维细胞收缩
IF 7.2 1区 医学
npj Regenerative Medicine Pub Date : 2024-04-03 DOI: 10.1038/s41536-024-00359-x
Anne Noom, Birgit Sawitzki, Petra Knaus, Georg N. Duda
{"title":"A two-way street – cellular metabolism and myofibroblast contraction","authors":"Anne Noom, Birgit Sawitzki, Petra Knaus, Georg N. Duda","doi":"10.1038/s41536-024-00359-x","DOIUrl":"https://doi.org/10.1038/s41536-024-00359-x","url":null,"abstract":"<p>Tissue fibrosis is characterised by the high-energy consumption associated with myofibroblast contraction. Although myofibroblast contraction relies on ATP production, the role of cellular metabolism in myofibroblast contraction has not yet been elucidated. Studies have so far only focused on myofibroblast contraction regulators, such as integrin receptors, TGF-β and their shared transcription factor YAP/TAZ, in a fibroblast-myofibroblast transition setting. Additionally, the influence of the regulators on metabolism and vice versa have been described in this context. However, this has so far not yet been connected to myofibroblast contraction. This review focuses on the known and unknown of how cellular metabolism influences the processes leading to myofibroblast contraction and vice versa. We elucidate the signalling cascades responsible for myofibroblast contraction by looking at FMT regulators, mechanical cues, biochemical signalling, ECM properties and how they can influence and be influenced by cellular metabolism. By reviewing the existing knowledge on the link between cellular metabolism and the regulation of myofibroblast contraction, we aim to pinpoint gaps of knowledge and eventually help identify potential research targets to identify strategies that would allow switching tissue fibrosis towards tissue regeneration.</p>","PeriodicalId":54236,"journal":{"name":"npj Regenerative Medicine","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2024-04-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140579218","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Comparison studies identify mesenchymal stromal cells with potent regenerative activity in osteoarthritis treatment. 对比研究发现间充质基质细胞在骨关节炎治疗中具有强大的再生活性。
IF 7.2 1区 医学
npj Regenerative Medicine Pub Date : 2024-04-01 DOI: 10.1038/s41536-024-00358-y
Hongshang Chu, Shaoyang Zhang, Zhenlin Zhang, Hua Yue, Huijuan Liu, Baojie Li, Feng Yin
{"title":"Comparison studies identify mesenchymal stromal cells with potent regenerative activity in osteoarthritis treatment.","authors":"Hongshang Chu, Shaoyang Zhang, Zhenlin Zhang, Hua Yue, Huijuan Liu, Baojie Li, Feng Yin","doi":"10.1038/s41536-024-00358-y","DOIUrl":"10.1038/s41536-024-00358-y","url":null,"abstract":"<p><p>Osteoarthritis affects 15% of people over 65 years of age. It is characterized by articular cartilage degradation and inflammation, leading to joint pain and disability. Osteoarthritis is incurable and the patients may eventually need joint replacement. An emerging treatment is mesenchymal stromal cells (MSCs), with over two hundred clinical trials being registered. However, the outcomes of these trials have fallen short of the expectation, due to heterogeneity of MSCs and uncertain mechanisms of action. It is generally believed that MSCs exert their function mainly by secreting immunomodulatory and trophic factors. Here we used knee osteoarthritis mouse model to assess the therapeutic effects of MSCs isolated from the white adipose or dermal adipose tissue of Prrx1-Cre; R26<sup>tdTomato</sup> mice and Dermo1-Cre; R26<sup>tdTomato</sup> mice. We found that the Prrx1-lineage MSCs from the white adipose tissues showed the greatest in vitro differentiation potentials among the four MSC groups and single cell profiling showed that the Prrx1-lineage MSCs contained more stem cells than the Dermo1 counterpart. Only the Prrx1-lineage cells isolated from white adipose tissues showed long-term therapeutic effectiveness on early-stage osteoarthritis models. Mechanistically, Prrx1-lineage MSCs differentiated into Col2<sup>+</sup> chondrocytes and replaced the damage cartilage, activated Col1 expressing in resident chondrocytes, and inhibited synovial inflammation. Transcriptome analysis showed that the articular chondrocytes derived from injected MSCs expressed immunomodulatory cytokines, trophic factors, and chondrocyte-specific genes. Our study identified a MSC population genetically marked by Prrx1 that has great multipotentiality and can differentiate into chondrocytes to replace the damaged cartilage.</p>","PeriodicalId":54236,"journal":{"name":"npj Regenerative Medicine","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10984924/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140337650","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Intrinsic and extrinsic actions of human neural progenitors with SUFU inhibition promote tissue repair and functional recovery from severe spinal cord injury. 抑制 SUFU 的人类神经祖细胞的内在和外在作用可促进严重脊髓损伤的组织修复和功能恢复。
IF 7.2 1区 医学
npj Regenerative Medicine Pub Date : 2024-03-22 DOI: 10.1038/s41536-024-00352-4
Yong-Long Chen, Xiang-Lan Feng, Kin-Wai Tam, Chao-Yang Fan, May Pui-Lai Cheung, Yong-Ting Yang, Stanley Wong, Daisy Kwok-Yan Shum, Ying-Shing Chan, Chi-Wai Cheung, Martin Cheung, Jessica Aijia Liu
{"title":"Intrinsic and extrinsic actions of human neural progenitors with SUFU inhibition promote tissue repair and functional recovery from severe spinal cord injury.","authors":"Yong-Long Chen, Xiang-Lan Feng, Kin-Wai Tam, Chao-Yang Fan, May Pui-Lai Cheung, Yong-Ting Yang, Stanley Wong, Daisy Kwok-Yan Shum, Ying-Shing Chan, Chi-Wai Cheung, Martin Cheung, Jessica Aijia Liu","doi":"10.1038/s41536-024-00352-4","DOIUrl":"10.1038/s41536-024-00352-4","url":null,"abstract":"<p><p>Neural progenitor cells (NPCs) derived from human pluripotent stem cells(hPSCs) provide major cell sources for repairing damaged neural circuitry and enabling axonal regeneration after spinal cord injury (SCI). However, the injury niche and inadequate intrinsic factors in the adult spinal cord restrict the therapeutic potential of transplanted NPCs. The Sonic Hedgehog protein (Shh) has crucial roles in neurodevelopment by promoting the formation of motorneurons and oligodendrocytes as well as its recently described neuroprotective features in response to the injury, indicating its essential role in neural homeostasis and tissue repair. In this study, we demonstrate that elevated SHH signaling in hNPCs by inhibiting its negative regulator, SUFU, enhanced cell survival and promoted robust neuronal differentiation with extensive axonal outgrowth, counteracting the harmful effects of the injured niche. Importantly, SUFU inhibition in NPCs exert non-cell autonomous effects on promoting survival and neurogenesis of endogenous cells and modulating the microenvironment by reducing suppressive barriers around lesion sites. The combined beneficial effects of SUFU inhibition in hNPCs resulted in the effective reconstruction of neuronal connectivity with the host and corticospinal regeneration, significantly improving neurobehavioral recovery in recipient animals. These results demonstrate that SUFU inhibition confers hNPCs with potent therapeutic potential to overcome extrinsic and intrinsic barriers in transplantation treatments for SCI.</p>","PeriodicalId":54236,"journal":{"name":"npj Regenerative Medicine","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2024-03-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10959923/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140195087","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Multichannel bridges and NSC synergize to enhance axon regeneration, myelination, synaptic reconnection, and recovery after SCI. 多通道桥和间叶干细胞可协同促进轴突再生、髓鞘化、突触重新连接以及损伤后的恢复。
IF 7.2 1区 医学
npj Regenerative Medicine Pub Date : 2024-03-18 DOI: 10.1038/s41536-024-00356-0
Usha Nekanti, Pooja S Sakthivel, Atena Zahedi, Dana A Creasman, Rebecca A Nishi, Courtney M Dumont, Katja M Piltti, Glenn L Guardamondo, Norbert Hernandez, Xingyuan Chen, Hui Song, Xiaoxiao Lin, Joshua Martinez, Lillian On, Anita Lakatos, Kiran Pawar, Brian T David, Zhiling Guo, Stephanie K Seidlits, Xiangmin Xu, Lonnie D Shea, Brian J Cummings, Aileen J Anderson
{"title":"Multichannel bridges and NSC synergize to enhance axon regeneration, myelination, synaptic reconnection, and recovery after SCI.","authors":"Usha Nekanti, Pooja S Sakthivel, Atena Zahedi, Dana A Creasman, Rebecca A Nishi, Courtney M Dumont, Katja M Piltti, Glenn L Guardamondo, Norbert Hernandez, Xingyuan Chen, Hui Song, Xiaoxiao Lin, Joshua Martinez, Lillian On, Anita Lakatos, Kiran Pawar, Brian T David, Zhiling Guo, Stephanie K Seidlits, Xiangmin Xu, Lonnie D Shea, Brian J Cummings, Aileen J Anderson","doi":"10.1038/s41536-024-00356-0","DOIUrl":"10.1038/s41536-024-00356-0","url":null,"abstract":"<p><p>Regeneration in the injured spinal cord is limited by physical and chemical barriers. Acute implantation of a multichannel poly(lactide-co-glycolide) (PLG) bridge mechanically stabilizes the injury, modulates inflammation, and provides a permissive environment for rapid cellularization and robust axonal regrowth through this otherwise inhibitory milieu. However, without additional intervention, regenerated axons remain largely unmyelinated (<10%), limiting functional repair. While transplanted human neural stem cells (hNSC) myelinate axons after spinal cord injury (SCI), hNSC fate is highly influenced by the SCI inflammatory microenvironment, also limiting functional repair. Accordingly, we investigated the combination of PLG scaffold bridges with hNSC to improve histological and functional outcome after SCI. In vitro, hNSC culture on a PLG scaffold increased oligodendroglial lineage selection after inflammatory challenge. In vivo, acute PLG bridge implantation followed by chronic hNSC transplantation demonstrated a robust capacity of donor human cells to migrate into PLG bridge channels along regenerating axons and integrate into the host spinal cord as myelinating oligodendrocytes and synaptically integrated neurons. Axons that regenerated through the PLG bridge formed synaptic circuits that connected the ipsilateral forelimb muscle to contralateral motor cortex. hNSC transplantation significantly enhanced the total number of regenerating and myelinated axons identified within the PLG bridge. Finally, the combination of acute bridge implantation and hNSC transplantation exhibited robust improvement in locomotor recovery. These data identify a successful strategy to enhance neurorepair through a temporally layered approach using acute bridge implantation and chronic cell transplantation to spare tissue, promote regeneration, and maximize the function of new axonal connections.</p>","PeriodicalId":54236,"journal":{"name":"npj Regenerative Medicine","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2024-03-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10948859/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140159473","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
Denervation alters the secretome of myofibers and thereby affects muscle stem cell lineage progression and functionality. 去神经支配会改变肌纤维的分泌组,从而影响肌肉干细胞系的发展和功能。
IF 7.2 1区 医学
npj Regenerative Medicine Pub Date : 2024-03-01 DOI: 10.1038/s41536-024-00353-3
Henriette Henze, Sören S Hüttner, Philipp Koch, Svenja C Schüler, Marco Groth, Björn von Eyss, Julia von Maltzahn
{"title":"Denervation alters the secretome of myofibers and thereby affects muscle stem cell lineage progression and functionality.","authors":"Henriette Henze, Sören S Hüttner, Philipp Koch, Svenja C Schüler, Marco Groth, Björn von Eyss, Julia von Maltzahn","doi":"10.1038/s41536-024-00353-3","DOIUrl":"10.1038/s41536-024-00353-3","url":null,"abstract":"<p><p>Skeletal muscle function crucially depends on innervation while repair of skeletal muscle relies on resident muscle stem cells (MuSCs). However, it is poorly understood how innervation affects MuSC properties and thereby regeneration of skeletal muscle. Here, we report that loss of innervation causes precocious activation of MuSCs concomitant with the expression of markers of myogenic differentiation. This aberrant activation of MuSCs after loss of innervation is accompanied by profound alterations on the mRNA and protein level. Combination of muscle injury with loss of innervation results in impaired regeneration of skeletal muscle including shifts in myogenic populations concomitant with delayed maturation of regenerating myofibers. We further demonstrate that loss of innervation leads to alterations in myofibers and their secretome, which then affect MuSC behavior. In particular, we identify an increased secretion of Osteopontin and transforming growth factor beta 1 (Tgfb1) by myofibers isolated from mice which had undergone sciatic nerve transection. The altered secretome results in the upregulation of early activating transcription factors, such as Junb, and their target genes in MuSCs. However, the combination of different secreted factors from myofibers after loss of innervation is required to cause the alterations observed in MuSCs after loss of innervation. These data demonstrate that loss of innervation first affects myofibers causing alterations in their secretome which then affect MuSCs underscoring the importance of proper innervation for MuSC functionality and regeneration of skeletal muscle.</p>","PeriodicalId":54236,"journal":{"name":"npj Regenerative Medicine","volume":null,"pages":null},"PeriodicalIF":7.2,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10904387/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139998297","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信