Stem Cell Research & Therapy最新文献

筛选
英文 中文
Impact of photobiomodulation on neural embryoid body formation from immortalized adipose-derived stem cells.
IF 7.1 2区 医学
Stem Cell Research & Therapy Pub Date : 2024-12-20 DOI: 10.1186/s13287-024-04088-2
Precious Earldom Mulaudzi, Heidi Abrahamse, Anine Crous
{"title":"Impact of photobiomodulation on neural embryoid body formation from immortalized adipose-derived stem cells.","authors":"Precious Earldom Mulaudzi, Heidi Abrahamse, Anine Crous","doi":"10.1186/s13287-024-04088-2","DOIUrl":"10.1186/s13287-024-04088-2","url":null,"abstract":"<p><strong>Background: </strong>Embryoid bodies (EBs) are three-dimensional (3D) multicellular cell aggregates that are derived from stem cell and play a pivotal role in regenerative medicine. They recapitulate many crucial aspects of the early stages of embryonic development and is the first step in the generation of various types of stem cells, including neuronal stem cells. Current methodologies for differentiating stem cells into neural embryoid bodies (NEBs) in vitro have advanced significantly, but they still have limitations which necessitate improvement. Photobiomodulation (PBM) a low powered light therapy is a non-invasive technique shown to promote stem cell proliferation and differentiation.</p><p><strong>Methods: </strong>This in vitro study elucidated the effects of photobiomodulation (PBM) on the differentiation of immortalized adipose-derived stem cells (iADSCs) into NEBs within a 3D cell culture environment. The study utilized PBM at wavelengths of 825 nm, 525 nm, and a combination of both, with fluences of 5 and 10 J/cm<sup>2</sup>. Morphology, viability, metabolic activity, and differentiation following PBM treatment was analysed.</p><p><strong>Results: </strong>The results revealed that the effects of photobiomodulation (PBM) are dose dependent. PBM, at 825 nm with a fluence of 10 J/cm<sup>2</sup>, significantly enhanced the size of neural embryoid bodies (NEBs), improved cell viability and proliferation, and reduced lactate dehydrogenase (LDH) levels, indicating minimal cell damage. Interestingly, the stem cell marker CD 44 was upregulated at 5 J/cm<sup>2</sup> in all treatment groups at 24 and 96 hpi, CD105 increased with 825 nm at 10 J/cm<sup>2</sup> at 24 hpi, which may be attributed to a heterogeneous cell population within the NEBs. Pax6 expression showed transient activation. Nestin was upregulated at 825 nm with 10 J/cm<sup>2</sup> at 96 hpi, suggesting a promotion of neural precursor populations. GFAP an intermediate filament protein was upregulated at 825 nm at 10 J/cm2 at both 24 and 96 hpi. SOX2, a pluripotency marker, was expressed at 5 J/cm<sup>2</sup> across all wavelengths. Neu N a neuronal nuclei marker was expressed at 5 J/cm<sup>2</sup> in all treatments at 24 hpi and over time the expression was observed in all treatment groups at 10 J/cm<sup>2</sup>.</p><p><strong>Conclusion: </strong>In conclusion, the application of PBM at 825 nm with a fluence of 10 J/cm<sup>2</sup> during the differentiation of iADSCs into NEBs resulted in optimal differentiation. Notably, the neuronal marker Nestin was significantly upregulated, highlighting the potential of the PBM approach for enhancing neuronal differentiation its promising applications in regenerative medicine.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"489"},"PeriodicalIF":7.1,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662703/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142872594","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
Human neural stem cells directly programmed from peripheral blood show functional integration into the adult mouse brain.
IF 7.1 2区 医学
Stem Cell Research & Therapy Pub Date : 2024-12-20 DOI: 10.1186/s13287-024-04110-7
Lea Jessica Berg, Chung Ku Lee, Hideaki Matsumura, Anke Leinhaas, Rachel Konang, Ali H Shaib, Pedro Royero, Julia Schlee, Chao Sheng, Heinz Beck, Martin Karl Schwarz, Nils Brose, Jeong Seop Rhee, Oliver Brüstle
{"title":"Human neural stem cells directly programmed from peripheral blood show functional integration into the adult mouse brain.","authors":"Lea Jessica Berg, Chung Ku Lee, Hideaki Matsumura, Anke Leinhaas, Rachel Konang, Ali H Shaib, Pedro Royero, Julia Schlee, Chao Sheng, Heinz Beck, Martin Karl Schwarz, Nils Brose, Jeong Seop Rhee, Oliver Brüstle","doi":"10.1186/s13287-024-04110-7","DOIUrl":"10.1186/s13287-024-04110-7","url":null,"abstract":"<p><p>Transplantation of induced pluripotent stem cell-derived neural cells represents a promising strategy for treating neurodegenerative diseases. However, reprogramming of somatic cells and their subsequent neural differentiation is complex and time-consuming, thereby impeding autologous applications. Recently, direct transcription factor-based conversion of blood cells into induced neural stem cells (iNSCs) has emerged as a potential alternative. However, little is known about the functionality of iNSC-derived neurons upon in vivo transplantation. Here, we grafted human iNSCs derived from adult peripheral blood by temporary overexpression of the transcription factors SOX2 and cMYC into the hippocampus or striatum of adult unlesioned immunodeficient Rag2<sup>tm1Fwa</sup>Il2rg<sup>tm1Wjl</sup> mice of both sexes. Engrafted cells gave rise to stable transplants composed of mature neurons displaying extensive neurite outgrowth and dendritic spine formation. Functional analyses of acute slices using patch clamp recordings revealed that already after 12 weeks of in vivo maturation, most of iNSC-derived cells possess unique properties exclusive to neurons and exhibit voltage-dependent ion channel currents as well as action potential firing. Moreover, the formation of spontaneous inhibitory and excitatory postsynaptic currents, along with Rabies virus-based retrograde monosynaptic tracing data, strongly supports the structural and functional integration of graft-derived neurons. Taken together, our data demonstrate that iNSCs directly derived from peripheral blood cells have the inherent capacity to achieve full functional maturation in vivo, qualifying them as an alternative potential donor source for restorative applications and deserving further investigation.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"488"},"PeriodicalIF":7.1,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662720/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142872654","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
Matrix-free human lung organoids derived from induced pluripotent stem cells to model lung injury.
IF 7.1 2区 医学
Stem Cell Research & Therapy Pub Date : 2024-12-18 DOI: 10.1186/s13287-024-04106-3
Bettina Budeus, Chiara Kroepel, Lisa Marie Stasch, Diana Klein
{"title":"Matrix-free human lung organoids derived from induced pluripotent stem cells to model lung injury.","authors":"Bettina Budeus, Chiara Kroepel, Lisa Marie Stasch, Diana Klein","doi":"10.1186/s13287-024-04106-3","DOIUrl":"10.1186/s13287-024-04106-3","url":null,"abstract":"<p><strong>Background: </strong>Organoids, as near-physiological 3D culture systems, offer new opportunities to study the pathogenesis of various organs in mimicking the cellular complexity and functionality of human organs.</p><p><strong>Method: </strong>Here we used a quite simple and very practicable method to successfully generate induced pluripotent stem cell (iPSC)-derived human lung organoids (LuOrg) in a matrix-free manner as an alternative to the widely used preclinical mouse models in order to investigate normal lung damage in detail and as close as possible to the patient. We performed detailed morphological and molecular analyses, including bulk and single cell RNA sequencing, of generated lung organoids and evaluated the quality and robustness of our model as a potential in vitro platform for lung diseases, namely radiation-induced lung injury.</p><p><strong>Results: </strong>A matrix-free method for differentiation of iPSCs can be used to obtain lung organoids that morphologically reflect the target tissue of the human lung very well, especially with regard to the cellular composition. The different cellular fates were investigated following the genotoxic stress induced by radiation and revealed further insights in the radiation-sensitivity of the different lung cells. Finally, we provide cellular gene sets found to be induced in the different lung organoid cellular subsets after irradiation, which could be used as additional RT response and particularly senescence gene sets in future studies.</p><p><strong>Conclusion: </strong>By establishing these free-floating LuOrgs for the investigation of cancer therapeutic approaches as a new and patient-oriented in vitro platform particularly in experimental radiooncology, not only a reduction in the number of experimental animals, but also an adequately and meaningfully replacement of corresponding animal experiments can be achieved.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"468"},"PeriodicalIF":7.1,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11657174/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142854759","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
Multiomics analyses reveal adipose-derived stem cells inhibit the inflammatory response of M1-like macrophages through secreting lactate.
IF 7.1 2区 医学
Stem Cell Research & Therapy Pub Date : 2024-12-18 DOI: 10.1186/s13287-024-04072-w
Tetsuhiro Horie, Hiroaki Hirata, Takuya Sakamoto, Hironori Kitajima, Atsushi Fuku, Yuka Nakamura, Yumi Sunatani, Ikuhiro Tanida, Hiroshi Sunami, Yoshiyuki Tachi, Yasuhito Ishigaki, Naoki Yamamoto, Yusuke Shimizu, Toru Ichiseki, Ayumi Kaneuji, Kuniyoshi Iwabuchi, Satoshi Osawa, Norio Kawahara
{"title":"Multiomics analyses reveal adipose-derived stem cells inhibit the inflammatory response of M1-like macrophages through secreting lactate.","authors":"Tetsuhiro Horie, Hiroaki Hirata, Takuya Sakamoto, Hironori Kitajima, Atsushi Fuku, Yuka Nakamura, Yumi Sunatani, Ikuhiro Tanida, Hiroshi Sunami, Yoshiyuki Tachi, Yasuhito Ishigaki, Naoki Yamamoto, Yusuke Shimizu, Toru Ichiseki, Ayumi Kaneuji, Kuniyoshi Iwabuchi, Satoshi Osawa, Norio Kawahara","doi":"10.1186/s13287-024-04072-w","DOIUrl":"10.1186/s13287-024-04072-w","url":null,"abstract":"<p><strong>Background: </strong>Adipose-derived stem cells (ADSCs) are widely used in the field of regenerative medicine because of their various functions, including anti-inflammatory effects. ADSCs are considered to exert their anti-inflammatory effects by secreting anti-inflammatory cytokines and extracellular vesicles. Although recent studies have reported that metabolites have a variety of physiological activities, whether those secreted by ADSCs have anti-inflammatory properties remains unclear. Here, we performed multiomics analyses to examine the effect of ADSC-derived metabolites on M1-like macrophages, which play an important role in inflammatory responses.</p><p><strong>Methods: </strong>The concentration of metabolites in the culture supernatant of ADSCs was quantified using capillary electrophoresis time-of-flight mass spectrometry. To evaluate their effects on inflammatory responses, M1-like macrophages were exposed to the conditioned ADSC medium or their metabolites, and RNA sequencing was used to detect gene expression changes. Immunoblotting was performed to examine how the metabolite suppresses inflammatory processes. To clarify the contribution of the metabolite in the conditioned medium to its anti-inflammatory effects, metabolite uptake was pharmacologically inhibited, and gene expression and the tumor necrosis factor-α concentration were measured by quantitative PCR and enzyme-linked immunosorbent assay, respectively.</p><p><strong>Results: </strong>Metabolomic analysis showed large amounts of lactate in the culture supernatant. The conditioned medium and lactate significantly suppressed or increased the pro-inflammatory and anti-inflammatory gene expressions. However, sequencing and immunoblotting analysis revealed that lactate did not induce polarization from M1- to M2-like macrophages. Based on a recent report that the immunosuppressive effect of lactate depends on epigenetic reprogramming, histone acetylation was investigated, and H3K27ac expression was upregulated. In addition, 7ACC2, which specifically inhibits the monocarboxylate transporter 1, significantly inhibited the anti-inflammatory effect of the conditioned ADSC medium on M1-like macrophages.</p><p><strong>Conclusions: </strong>Our results showed that ADSCs suppress pro-inflammatory effects of M1-like macrophages by secreting lactate. This study adds to our understanding of the importance of metabolites and is also expected to elucidate new mechanisms of ADSC treatments.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"485"},"PeriodicalIF":7.1,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11657992/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855220","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
FAM96B negatively regulates FOSL1 to modulate the osteogenic differentiation and regeneration of periodontal ligament stem cells via ferroptosis.
IF 7.1 2区 医学
Stem Cell Research & Therapy Pub Date : 2024-12-18 DOI: 10.1186/s13287-024-04083-7
Qianyi Qin, Haoqing Yang, Runzhi Guo, Yunfei Zheng, Yiping Huang, Luyuan Jin, Zhipeng Fan, Weiran Li
{"title":"FAM96B negatively regulates FOSL1 to modulate the osteogenic differentiation and regeneration of periodontal ligament stem cells via ferroptosis.","authors":"Qianyi Qin, Haoqing Yang, Runzhi Guo, Yunfei Zheng, Yiping Huang, Luyuan Jin, Zhipeng Fan, Weiran Li","doi":"10.1186/s13287-024-04083-7","DOIUrl":"10.1186/s13287-024-04083-7","url":null,"abstract":"<p><strong>Background: </strong>Periodontal ligament stem cell (PDLSC)-based therapy is one of the methods to assist bone regeneration. Understanding the functional regulation of PDLSCs and the mechanisms involved is a crucial issue in bone regeneration. This study aimed to explore the roles of the family with sequence similarity 96 member B (FAM96B) in the functional regulation of PDLSCs.</p><p><strong>Methods: </strong>To assess the osteogenic differentiation of PDLSCs, the alkaline phosphatase (ALP) activity assay, Alizarin red staining, quantitative calcium analysis, and osteogenic marker detection were conducted. Transplantation PDLSCs under the dorsum of nude mice and into the rat calvarial defects were also performed. Then, FAM96B-overexpressed PDLSCs were used for RNA-sequencing and bioinformatic analysis. To evaluate the ferroptosis of PDLSCs, cytosolic reactive oxygen species (ROS), expression of glutathione peroxidase 4 (GPX4), mitochondrial morphology and functions including the mitochondrial ROS, mitochondria membrane potential, and mitochondrial respiration were detected.</p><p><strong>Results: </strong>The osteogenic indicators ALP activity, level of mineralization, and osteocalcin expression were decreased in PDLSCs by FAM96B, which demonstrated that FAM96B inhibited the osteogenic differentiation of PDLSCs. FAM96B knockdown promoted the new bone formation of PDLSCs subcutaneously transplanted to the dorsum of nude mice. Then, related biological functions were detected by the RNA-sequencing and the ferroptosis was focused. FAM96B enhanced the cytosolic ROS level and inhibited the expression of GPX4 and mitochondrial functions in PDLSCs. Hence, FAM96B promoted the ferroptosis of PDLSCs. Meanwhile, we found that FAM96B inhibition upregulated the target gene FOS like 1, AP-1 transcription factor subunit (FOSL1) expression and FOSL1 promoted the osteogenic differentiation of PDLSCs in vitro. FOSL1 also promoted the new bone formation of PDLSCs transplanted subcutaneously to the dorsum of nude mice and transplanted into rat calvarial defects. Then, the inhibitory effect of FOSL1 on the ferroptosis was confirmed.</p><p><strong>Conclusions: </strong>FAM96B depletion promoted the osteogenic differentiation and suppressed the ferroptosis of PDLSCs. FAM96B negatively regulated the downstream gene FOSL1 and FOSL1 promoted the osteogenic differentiation of PDLSCs via the ferroptosis. Hence, our findings provided a foundation for understanding the FAM96B-FOSL1 axis acting as a target for MSC mediated bone regeneration.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"471"},"PeriodicalIF":7.1,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11656916/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855332","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
Glutamine-αKG axis affects dentin regeneration and regulates osteo/odontogenic differentiation of mesenchymal adult stem cells via IGF2 m6A modification.
IF 7.1 2区 医学
Stem Cell Research & Therapy Pub Date : 2024-12-18 DOI: 10.1186/s13287-024-04092-6
Qinglu Tian, Shiqi Gao, Siying Li, Mian Wan, Xin Zhou, Wei Du, Xuedong Zhou, Liwei Zheng, Yachuan Zhou
{"title":"Glutamine-αKG axis affects dentin regeneration and regulates osteo/odontogenic differentiation of mesenchymal adult stem cells via IGF2 m6A modification.","authors":"Qinglu Tian, Shiqi Gao, Siying Li, Mian Wan, Xin Zhou, Wei Du, Xuedong Zhou, Liwei Zheng, Yachuan Zhou","doi":"10.1186/s13287-024-04092-6","DOIUrl":"10.1186/s13287-024-04092-6","url":null,"abstract":"<p><strong>Background: </strong>Multi-lineage differentiation of mesenchymal adult stem cells (m-ASCs) is crucial for tissue regeneration and accompanied with metabolism reprogramming, among which dental-pulp-derived m-ASCs has obvious advantage of easy accessibility. Stem cell fate determination and differentiation are closely related to metabolism status in cell microenvironment, which could actively interact with epigenetic modification. In recent years, glutamine-α-ketoglutarate (αKG) axis was proved to be related to aging, tumorigenesis, osteogenesis etc., while its role in m-ASCs still lack adequate research evidence.</p><p><strong>Methods: </strong>We employed metabolomic analysis to explore the change pattern of metabolites during dental-pulp-derived m-ASCs differentiation. A murine incisor clipping model was established to investigate the influence of αKG on dental tissue repairment. shRNA technique was used to knockdown the expression of related key enzyme-dehydrogenase 1(GLUD1). RNA-seq, m6A evaluation and MeRIP-qPCR were used to dig into the underlying epigenetic mechanism.</p><p><strong>Results: </strong>Here we found that the glutamine-αKG axis displayed an increased tendency along with the osteo/odontogenic differentiation of dental-pulp-derived m-ASCs, same as expression pattern of GLUD1. Further, the key metabolite αKG was found able to accelerate the repairment of clipped mice incisor and promote dentin formation. Exogenous DM-αKG was proved able to promote osteo/odontogenic differentiation of dental-pulp-derived m-ASCs, while the inhibition of glutamine-derived αKG level via GLUD1 knockdown had the opposite effect. Under the circumstance of GLUD1 knockdown, extracellular matrix (ECM) function and PI3k-Akt signaling pathway was screened out to be widely involved in the process with insulin-like growth factor 2 (IGF2) participation via RNA-seq. Inhibition of glutamine-αKG axis may affect IGF2 translation efficiency via m6A methylation and can be significantly rescued by αKG supplementation.</p><p><strong>Conclusion: </strong>Our findings indicate that glutamine-αKG axis may epigenetically promote osteo/odontogenic differentiation of dental-pulp-derived m-ASCs and dentin regeneration, which provide a new research vision of potential dental tissue repairment therapy method or metabolite-based drug research.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"479"},"PeriodicalIF":7.1,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11657990/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855410","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
Advances in the treatment of liver injury based on mesenchymal stem cell-derived exosomes.
IF 7.1 2区 医学
Stem Cell Research & Therapy Pub Date : 2024-12-18 DOI: 10.1186/s13287-024-04087-3
Changlong Hu, Lin Wang
{"title":"Advances in the treatment of liver injury based on mesenchymal stem cell-derived exosomes.","authors":"Changlong Hu, Lin Wang","doi":"10.1186/s13287-024-04087-3","DOIUrl":"10.1186/s13287-024-04087-3","url":null,"abstract":"<p><p>Mesenchymal stem cells (MSCs) have shown a great potential role in treating liver injury. MSCs can promote liver regeneration by differentiating into hepatocytes, and can also secrete exosomes to participate in the repair of liver injury. Increasing evidence has shown that mesenchymal stem cell-derived exosomes (MSC-EXOs) play an important role in treating liver injury. In this review, the biogenesis and function of exosomes and the characteristics of MSC-EXOs were analyzed based on recent research results. MSC-EXOs are significant in liver injuries such as liver fibrosis, liver failure, hepatocellular carcinoma, oxidative stress, and lipid steatosis, and participate in the process of liver regeneration.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"474"},"PeriodicalIF":7.1,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11657567/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855360","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
Time-efficient strategies in human iPS cell-derived pancreatic progenitor differentiation and cryopreservation: advancing towards practical applications.
IF 7.1 2区 医学
Stem Cell Research & Therapy Pub Date : 2024-12-18 DOI: 10.1186/s13287-024-04068-6
Elena Genova, Paola Rispoli, Yue Fengming, Johkura Kohei, Matteo Bramuzzo, Roberta Bulla, Marianna Lucafò, Rosalba Monica Ferraro, Giuliana Decorti, Gabriele Stocco
{"title":"Time-efficient strategies in human iPS cell-derived pancreatic progenitor differentiation and cryopreservation: advancing towards practical applications.","authors":"Elena Genova, Paola Rispoli, Yue Fengming, Johkura Kohei, Matteo Bramuzzo, Roberta Bulla, Marianna Lucafò, Rosalba Monica Ferraro, Giuliana Decorti, Gabriele Stocco","doi":"10.1186/s13287-024-04068-6","DOIUrl":"10.1186/s13287-024-04068-6","url":null,"abstract":"<p><strong>Background: </strong>Differentiation of patient-specific induced pluripotent stem cells (iPS) helps researchers to study the individual sensibility to drugs. However, differentiation protocols are time-consuming, and not all tissues have been studied. Few works are available regarding pancreatic exocrine differentiation of iPS cells, and little is known on culturing and cryopreserving these cells.</p><p><strong>Methods: </strong>We differentiated the iPS cells of two pediatric Crohn's disease patients into pancreatic progenitors and exocrine cells, adapting and shortening a protocol for differentiating embryonic stem cells. We analyzed the expression of key genes and proteins of the differentiation process by qPCR and immunofluorescence, respectively. We explored the possibility of keeping differentiated cells in culture and freezing and thawing them to shorten the time needed for the differentiation. We analyzed the cell cycle of undifferentiated and differentiated cells by flow cytometry.</p><p><strong>Results: </strong>The analysis of mRNA levels of key pancreatic differentiation genes PDX1 and pancreatic amylase indicate that iPS cells were successfully differentiated into pancreatic exocrine cells with expression of PDX1 (one way ANOVA p < 0.0001), and the two isoforms of amylase (one way ANOVA p < 0.05) significantly higher in exocrine cells in comparison to iPS cells. Differentiation efficiency was also confirmed by immunofluorescence analysis of PDX1 and amylase. We confirmed the possibility of shortening the time necessary for obtaining pancreatic cells without losing differentiation efficiency. Pancreatic progenitors and exocrine cells were maintained in culture and cryopreserved. Interestingly, the stemness marker OCT4 resulted significantly lower after subculturing (OCT4 p < 0.001; one-way ANOVA) and after freezing and thawing procedures (p < 0.05, one-way ANOVA) suggesting a reduction of undifferentiated stem cells leading to a purer population of pancreatic progenitor cells. Also, the stemness marker NANOG resulted lower after passaging, corroborating this result.</p><p><strong>Conclusions: </strong>In this work, we optimized the generation of patient-specific pancreatic differentiated cells and laid the foundation for creating a bank of patient-specific pancreatic lines exploitable for tailored pharmacological assays.</p><p><strong>Trial registration: </strong>The study was approved by the Ethical Committee of the Institute of Maternal and Child Health IRCCS Burlo Garofolo, with approval number 1556 (internal ID RC 44/22).</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"483"},"PeriodicalIF":7.1,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11658428/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855189","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
Microfluidic chip systems for characterizing glucose-responsive insulin-secreting cells equipped with FailSafe kill-switch.
IF 7.1 2区 医学
Stem Cell Research & Therapy Pub Date : 2024-12-18 DOI: 10.1186/s13287-024-04059-7
Mohammad Izadifar, Mohammad Massumi, Kacey J Prentice, Tatiana Oussenko, Biao Li, Judith Elbaz, Mira Puri, Michael B Wheeler, Andras Nagy
{"title":"Microfluidic chip systems for characterizing glucose-responsive insulin-secreting cells equipped with FailSafe kill-switch.","authors":"Mohammad Izadifar, Mohammad Massumi, Kacey J Prentice, Tatiana Oussenko, Biao Li, Judith Elbaz, Mira Puri, Michael B Wheeler, Andras Nagy","doi":"10.1186/s13287-024-04059-7","DOIUrl":"10.1186/s13287-024-04059-7","url":null,"abstract":"<p><strong>Background: </strong>Pluripotent cell-derived islet replacement therapy offers promise for treating Type 1 diabetes (T1D), but concerns about uncontrolled cell proliferation and tumorigenicity present significant safety challenges. To address the safety concern, this study aims to establish a proof-of-concept for a glucose-responsive, insulin-secreting cell line integrated with a built-in FailSafe kill-switch.</p><p><strong>Method: </strong>We generated β cell-induced progenitor-like cells (βiPLCs) from primary mouse pancreatic β cells through interrupted reprogramming. Then, we transcriptionally linked our FailSafe (FS) kill-switch, HSV-thymidine kinase (TK), to Cdk1 gene using a CRISPR/Cas9 knock-in strategy, resulting in a FailSafe βiPLC line, designated as FSβiPLCs. Subsequently we evaluated and confirmed the functionality of the drug-inducible kill-switch in FSβiPLCs at different ganciclovir (GCV) concentrations using our PDMS-based transcapillary microfluidic system. Finally, we assessed the functionality of FSβiPLCs by characterizing the dynamics of insulin secretion in response to changes in glucose concentration using our microfluidic perfusion glucose-stimulated insulin secretion (GSIS) assay-on- chip.</p><p><strong>Results: </strong>The βiPLCs exhibited Ins1, Pdx1 and Nkx6.1 expression, and glucose responsive insulin secretion, the essential properties of pancreatic beta cells. The βiPLCs were amenable to genome editing which allowed for the insertion of the kill-switch into the 3'UTR of Cdk1, confirmed by PCR genotyping. Our transcapillary microfluidic system confirmed the functionality of the drug-inducible kill-switch in FSβiPLCs, showing an effective cell ablation of dividing cells from a heterogeneous cell population at different ganciclovir (GCV) concentrations. The Ki67 expression assessment further confirmed that slow- or non-dividing cells in the FSβiPLC population were resistant to GCV. Our perfusion glucose-stimulated insulin secretion (GSIS) assay-on-chip revealed that the resistant non-dividing FSβiPLCs exhibited higher levels of insulin secretion and glucose responsiveness compared to their proliferating counterparts.</p><p><strong>Conclusions: </strong>This study establishes a proof-of-concept for the integration of a FailSafe kill-switch system into a glucose-responsive, insulin-secreting cell line to address the safety concerns in stem cell-derived cell replacement treatment for T1D. The microfluidic systems provided valuable insights into the functionality and safety of these engineered cells, demonstrating the potential of the kill-switch to reduce the risk of tumorigenicity in pluripotent cell-derived insulin-secreting cells.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"486"},"PeriodicalIF":7.1,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11656860/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855077","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
Exosome-based therapies for inflammatory disorders: a review of recent advances.
IF 7.1 2区 医学
Stem Cell Research & Therapy Pub Date : 2024-12-18 DOI: 10.1186/s13287-024-04107-2
Mavra Saleem, Khawar Ali Shahzad, Munazzah Marryum, Shekhar Singh, Quan Zhou, Siting Du, Shuanghu Wang, Chuxiao Shao, Imran Ibrahim Shaikh
{"title":"Exosome-based therapies for inflammatory disorders: a review of recent advances.","authors":"Mavra Saleem, Khawar Ali Shahzad, Munazzah Marryum, Shekhar Singh, Quan Zhou, Siting Du, Shuanghu Wang, Chuxiao Shao, Imran Ibrahim Shaikh","doi":"10.1186/s13287-024-04107-2","DOIUrl":"10.1186/s13287-024-04107-2","url":null,"abstract":"<p><p>Exosomes, small extracellular vesicles secreted by cells, have emerged as focal mediators in intercellular communication and therapeutic interventions across diverse biomedical fields. Inflammatory disorders, including inflammatory bowel disease, acute liver injury, lung injury, neuroinflammation, and myocardial infarction, are complex conditions that require innovative therapeutic approaches. This review summarizes recent advances in exosome-based therapies for inflammatory disorders, highlighting their potential as diagnostic biomarkers and therapeutic agents. Exosomes have shown promise in reducing inflammation, promoting tissue repair, and improving functional outcomes in preclinical models of inflammatory disorders. However, further research is needed to overcome the challenges associated with exosome isolation, characterization, and delivery, as well as to fully understand their mechanisms of action. Current limitations and future directions in exosome research underscore the need for enhanced isolation techniques and deeper mechanistic insights to harness exosomes' full therapeutic potential in clinical applications. Despite these challenges, exosome-based therapies hold great potential for the treatment of inflammatory disorders and may offer a new paradigm for personalized medication.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"477"},"PeriodicalIF":7.1,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11657721/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142855287","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
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