Pub Date : 2024-06-13DOI: 10.1016/j.stem.2024.05.007
Olga Mitrofanova, Mikhail Nikolaev, Quan Xu, Nicolas Broguiere, Irineja Cubela, J. Gray Camp, Michael Bscheider, Matthias P. Lutolf
Organoids and organs-on-a-chip have emerged as powerful tools for modeling human gut physiology and disease in vitro. Although physiologically relevant, these systems often lack the environmental milieu, spatial organization, cell type diversity, and maturity necessary for mimicking human intestinal mucosa. To instead generate models closely resembling in vivo tissue, we herein integrated organoid and organ-on-a-chip technology to develop an advanced human organoid model, called “mini-colons.” By employing an asymmetric stimulation with growth factors, we greatly enhanced tissue longevity and replicated in vivo-like diversity and patterning of proliferative and differentiated cell types. Mini-colons contain abundant mucus-producing goblet cells and, signifying mini-colon maturation, single-cell RNA sequencing reveals emerging mature and functional colonocytes. This methodology is expanded to generate microtissues from the small intestine and incorporate additional microenvironmental components. Finally, our bioengineered organoids provide a precise platform to systematically study human gut physiology and pathology, and a reliable preclinical model for drug safety assessment.
{"title":"Bioengineered human colon organoids with in vivo-like cellular complexity and function","authors":"Olga Mitrofanova, Mikhail Nikolaev, Quan Xu, Nicolas Broguiere, Irineja Cubela, J. Gray Camp, Michael Bscheider, Matthias P. Lutolf","doi":"10.1016/j.stem.2024.05.007","DOIUrl":"https://doi.org/10.1016/j.stem.2024.05.007","url":null,"abstract":"<p>Organoids and organs-on-a-chip have emerged as powerful tools for modeling human gut physiology and disease <em>in vitro</em>. Although physiologically relevant, these systems often lack the environmental milieu, spatial organization, cell type diversity, and maturity necessary for mimicking human intestinal mucosa. To instead generate models closely resembling <em>in vivo</em> tissue, we herein integrated organoid and organ-on-a-chip technology to develop an advanced human organoid model, called “mini-colons.” By employing an asymmetric stimulation with growth factors, we greatly enhanced tissue longevity and replicated <em>in vivo</em>-like diversity and patterning of proliferative and differentiated cell types. Mini-colons contain abundant mucus-producing goblet cells and, signifying mini-colon maturation, single-cell RNA sequencing reveals emerging mature and functional colonocytes. This methodology is expanded to generate microtissues from the small intestine and incorporate additional microenvironmental components. Finally, our bioengineered organoids provide a precise platform to systematically study human gut physiology and pathology, and a reliable preclinical model for drug safety assessment.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":"70 1","pages":""},"PeriodicalIF":23.9,"publicationDate":"2024-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141315832","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}
Pub Date : 2024-06-06DOI: 10.1016/j.stem.2024.05.002
Mehek Ningoo, Miguel Fribourg
In this issue, Yano et al.1 present a method to obtain suppressive regulatory T (Treg) cells from human induced pluripotent stem cells (hiPSCs). This approach has the potential to address the low Treg cell yields of current ex vivo Treg cell expansion and induction protocols, an unmet challenge for autologous Treg cell treatments.
{"title":"From stem cells to regulatory T cells: A tale of plasticity","authors":"Mehek Ningoo, Miguel Fribourg","doi":"10.1016/j.stem.2024.05.002","DOIUrl":"https://doi.org/10.1016/j.stem.2024.05.002","url":null,"abstract":"<p>In this issue<em>,</em> Yano et al.<span><sup>1</sup></span> present a method to obtain suppressive regulatory T (Treg) cells from human induced pluripotent stem cells (hiPSCs). This approach has the potential to address the low Treg cell yields of current <em>ex vivo</em> Treg cell expansion and induction protocols, an unmet challenge for autologous Treg cell treatments.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":"9 1","pages":""},"PeriodicalIF":23.9,"publicationDate":"2024-06-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141265013","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}
Pub Date : 2024-06-06DOI: 10.1016/j.stem.2024.05.001
Christos Karampelias, Heiko Lickert
Getting mature and functional stem cell-derived, insulin-producing β cells is an important step for disease modeling, drug screening, and cell replacement therapy. In this issue, Hua et al.1 used single-cell multiomics analysis coupled with chemical screening to identify a crucial role for ceramides in generating mature stem cell-derived β cells.
{"title":"Greasing the machinery toward maturation of stem cell-derived β cells","authors":"Christos Karampelias, Heiko Lickert","doi":"10.1016/j.stem.2024.05.001","DOIUrl":"https://doi.org/10.1016/j.stem.2024.05.001","url":null,"abstract":"<p>Getting mature and functional stem cell-derived, insulin-producing β cells is an important step for disease modeling, drug screening, and cell replacement therapy. In this issue, Hua et al.<span><sup>1</sup></span> used single-cell multiomics analysis coupled with chemical screening to identify a crucial role for ceramides in generating mature stem cell-derived β cells.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":"34 1","pages":""},"PeriodicalIF":23.9,"publicationDate":"2024-06-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141265056","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}
Pub Date : 2024-06-06DOI: 10.1016/j.stem.2024.05.006
Daniela Paasch, Nico Lachmann
Chimeric antigen receptor (CAR) macrophages have broadened the landscape of anti-cancer immunotherapies to combat solid malignancies. Shah et al. introduce CARs to facilitate a CAR macrophage therapy, which aims to recruit and activate T/natural killer cells, further strengthening the overall immune response to decrease pancreatic cancer burden and metastatic spreading.
嵌合抗原受体(CAR)巨噬细胞拓宽了抗癌免疫疗法的领域,以对抗实体恶性肿瘤。Shah 等人引入了 CAR,以促进 CAR 巨噬细胞疗法,该疗法旨在招募和激活 T/自然杀伤细胞,进一步加强整体免疫反应,以减少胰腺癌的负担和转移扩散。
{"title":"CAR macrophages tuning the immune symphony of anti-cancer therapies","authors":"Daniela Paasch, Nico Lachmann","doi":"10.1016/j.stem.2024.05.006","DOIUrl":"https://doi.org/10.1016/j.stem.2024.05.006","url":null,"abstract":"<p>Chimeric antigen receptor (CAR) macrophages have broadened the landscape of anti-cancer immunotherapies to combat solid malignancies. Shah et al. introduce CARs to facilitate a CAR macrophage therapy, which aims to recruit and activate T/natural killer cells, further strengthening the overall immune response to decrease pancreatic cancer burden and metastatic spreading.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":"34 1","pages":""},"PeriodicalIF":23.9,"publicationDate":"2024-06-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141265097","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}
CD4+ T cells induced from human iPSCs (iCD4+ T cells) offer a therapeutic opportunity for overcoming immune pathologies arising from hematopoietic stem cell transplantation. However, most iCD4+ T cells are conventional helper T cells, which secrete inflammatory cytokines. We induced high-level expression of FOXP3, a master transcription factor of regulatory T cells, in iCD4+ T cells. Human iPSC-derived, FOXP3-induced CD4+ T (iCD4+ Treg-like) cells did not secrete inflammatory cytokines upon activation. Moreover, they showed demethylation of the Treg-specific demethylation region, suggesting successful conversion to immunosuppressive iCD4+ Treg-like cells. We further assessed these iCD4+ Treg-like cells for CAR-mediated immunosuppressive ability. HLA-A2 CAR-transduced iCD4+ Treg-like cells inhibited CD8+ cytotoxic T cell (CTL) division in a mixed lymphocyte reaction assay with A2+ allogeneic CTLs and suppressed xenogeneic graft-versus-host disease (GVHD) in NSG mice treated with A2+ human PBMCs. In most cases, these cells suppressed the xenogeneic GvHD progression as much as natural CD25+CD127− Tregs did.
由人类iPSC诱导的CD4+ T细胞(iCD4+ T细胞)为克服造血干细胞移植引起的免疫病症提供了治疗机会。然而,大多数 iCD4+ T 细胞是传统的辅助性 T 细胞,会分泌炎性细胞因子。我们诱导 iCD4+ T 细胞高水平表达调节性 T 细胞的主转录因子 FOXP3。由 FOXP3 诱导的人类 iPSC 衍生的 CD4+ T(iCD4+ Treg-like)细胞在激活后不会分泌炎性细胞因子。此外,它们显示出 Treg 特异性去甲基化区域的去甲基化,这表明它们成功地转化为免疫抑制性 iCD4+ Treg 样细胞。我们进一步评估了这些 iCD4+ Treg 样细胞的 CAR 介导的免疫抑制能力。HLA-A2 CAR转化的iCD4+ Treg样细胞在与A2+异体CTLs的混合淋巴细胞反应试验中抑制了CD8+细胞毒性T细胞(CTL)分裂,并抑制了用A2+人类PBMCs治疗的NSG小鼠的异种移植物抗宿主病(GVHD)。在大多数情况下,这些细胞对异种移植物抗宿主疾病进展的抑制作用不亚于天然 CD25+CD127- Tregs。
{"title":"Human iPSC-derived CD4+ Treg-like cells engineered with chimeric antigen receptors control GvHD in a xenograft model","authors":"Hisashi Yano, Keiko Koga, Takayuki Sato, Tokuyuki Shinohara, Shoichi Iriguchi, Atsushi Matsuda, Kazuki Nakazono, Maki Shioiri, Yasuyuki Miyake, Yoshiaki Kassai, Hitoshi Kiyoi, Shin Kaneko","doi":"10.1016/j.stem.2024.05.004","DOIUrl":"https://doi.org/10.1016/j.stem.2024.05.004","url":null,"abstract":"<p>CD4<sup>+</sup> T cells induced from human iPSCs (iCD4<sup>+</sup> T cells) offer a therapeutic opportunity for overcoming immune pathologies arising from hematopoietic stem cell transplantation. However, most iCD4<sup>+</sup> T cells are conventional helper T cells, which secrete inflammatory cytokines. We induced high-level expression of FOXP3, a master transcription factor of regulatory T cells, in iCD4<sup>+</sup> T cells. Human iPSC-derived, FOXP3-induced CD4<sup>+</sup> T (iCD4<sup>+</sup> Treg-like) cells did not secrete inflammatory cytokines upon activation. Moreover, they showed demethylation of the Treg-specific demethylation region, suggesting successful conversion to immunosuppressive iCD4<sup>+</sup> Treg-like cells. We further assessed these iCD4<sup>+</sup> Treg-like cells for CAR-mediated immunosuppressive ability. HLA-A2 CAR-transduced iCD4<sup>+</sup> Treg-like cells inhibited CD8<sup>+</sup> cytotoxic T cell (CTL) division in a mixed lymphocyte reaction assay with A2<sup>+</sup> allogeneic CTLs and suppressed xenogeneic graft-versus-host disease (GVHD) in NSG mice treated with A2<sup>+</sup> human PBMCs. In most cases, these cells suppressed the xenogeneic GvHD progression as much as natural CD25<sup>+</sup>CD127<sup>−</sup> Tregs did.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":"34 1","pages":""},"PeriodicalIF":23.9,"publicationDate":"2024-06-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141265107","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}
Pub Date : 2024-06-05DOI: 10.1016/j.stem.2024.05.005
Yongshun Lin, Noriko Sato, Sogun Hong, Kenta Nakamura, Elisa A. Ferrante, Zu Xi Yu, Marcus Y. Chen, Daisy S. Nakamura, Xiulan Yang, Randall R. Clevenger, Timothy J. Hunt, Joni L. Taylor, Kenneth R. Jeffries, Karen J. Keeran, Lauren E. Neidig, Atul Mehta, Robin Schwartzbeck, Shiqin Judy Yu, Conor Kelly, Keron Navarengom, Cynthia E. Dunbar
Cellular therapies with cardiomyocytes produced from induced pluripotent stem cells (iPSC-CMs) offer a potential route to cardiac regeneration as a treatment for chronic ischemic heart disease. Here, we report successful long-term engraftment and in vivo maturation of autologous iPSC-CMs in two rhesus macaques with small, subclinical chronic myocardial infarctions, all without immunosuppression. Longitudinal positron emission tomography imaging using the sodium/iodide symporter (NIS) reporter gene revealed stable grafts for over 6 and 12 months, with no teratoma formation. Histological analyses suggested capability of the transplanted iPSC-CMs to mature and integrate with endogenous myocardium, with no sign of immune cell infiltration or rejection. By contrast, allogeneic iPSC-CMs were rejected within 8 weeks of transplantation. This study provides the longest-term safety and maturation data to date in any large animal model, addresses concerns regarding neoantigen immunoreactivity of autologous iPSC therapies, and suggests that autologous iPSC-CMs would similarly engraft and mature in human hearts.
{"title":"Long-term engraftment and maturation of autologous iPSC-derived cardiomyocytes in two rhesus macaques","authors":"Yongshun Lin, Noriko Sato, Sogun Hong, Kenta Nakamura, Elisa A. Ferrante, Zu Xi Yu, Marcus Y. Chen, Daisy S. Nakamura, Xiulan Yang, Randall R. Clevenger, Timothy J. Hunt, Joni L. Taylor, Kenneth R. Jeffries, Karen J. Keeran, Lauren E. Neidig, Atul Mehta, Robin Schwartzbeck, Shiqin Judy Yu, Conor Kelly, Keron Navarengom, Cynthia E. Dunbar","doi":"10.1016/j.stem.2024.05.005","DOIUrl":"https://doi.org/10.1016/j.stem.2024.05.005","url":null,"abstract":"<p>Cellular therapies with cardiomyocytes produced from induced pluripotent stem cells (iPSC-CMs) offer a potential route to cardiac regeneration as a treatment for chronic ischemic heart disease. Here, we report successful long-term engraftment and <em>in vivo</em> maturation of autologous iPSC-CMs in two rhesus macaques with small, subclinical chronic myocardial infarctions, all without immunosuppression. Longitudinal positron emission tomography imaging using the sodium/iodide symporter (NIS) reporter gene revealed stable grafts for over 6 and 12 months, with no teratoma formation. Histological analyses suggested capability of the transplanted iPSC-CMs to mature and integrate with endogenous myocardium, with no sign of immune cell infiltration or rejection. By contrast, allogeneic iPSC-CMs were rejected within 8 weeks of transplantation. This study provides the longest-term safety and maturation data to date in any large animal model, addresses concerns regarding neoantigen immunoreactivity of autologous iPSC therapies, and suggests that autologous iPSC-CMs would similarly engraft and mature in human hearts.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":"1 1","pages":""},"PeriodicalIF":23.9,"publicationDate":"2024-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141252003","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}
Pub Date : 2024-05-31DOI: 10.1016/j.stem.2024.05.003
Anish Dattani, Elena Corujo-Simon, Arthur Radley, Tiam Heydari, Yasaman Taheriabkenar, Francesca Carlisle, Simeng Lin, Corin Liddle, Jonathan Mill, Peter W. Zandstra, Jennifer Nichols, Ge Guo
The hypoblast is an essential extraembryonic tissue set aside within the inner cell mass in the blastocyst. Research with human embryos is challenging. Thus, stem cell models that reproduce hypoblast differentiation provide valuable alternatives. We show here that human naive pluripotent stem cell (PSC) to hypoblast differentiation proceeds via reversion to a transitional ICM-like state from which the hypoblast emerges in concordance with the trajectory in human blastocysts. We identified a window when fibroblast growth factor (FGF) signaling is critical for hypoblast specification. Revisiting FGF signaling in human embryos revealed that inhibition in the early blastocyst suppresses hypoblast formation. In vitro, the induction of hypoblast is synergistically enhanced by limiting trophectoderm and epiblast fates. This finding revises previous reports and establishes a conservation in lineage specification between mice and humans. Overall, this study demonstrates the utility of human naive PSC-based models in elucidating the mechanistic features of early human embryogenesis.
{"title":"Naive pluripotent stem cell-based models capture FGF-dependent human hypoblast lineage specification","authors":"Anish Dattani, Elena Corujo-Simon, Arthur Radley, Tiam Heydari, Yasaman Taheriabkenar, Francesca Carlisle, Simeng Lin, Corin Liddle, Jonathan Mill, Peter W. Zandstra, Jennifer Nichols, Ge Guo","doi":"10.1016/j.stem.2024.05.003","DOIUrl":"https://doi.org/10.1016/j.stem.2024.05.003","url":null,"abstract":"<p>The hypoblast is an essential extraembryonic tissue set aside within the inner cell mass in the blastocyst. Research with human embryos is challenging. Thus, stem cell models that reproduce hypoblast differentiation provide valuable alternatives. We show here that human naive pluripotent stem cell (PSC) to hypoblast differentiation proceeds via reversion to a transitional ICM-like state from which the hypoblast emerges in concordance with the trajectory in human blastocysts. We identified a window when fibroblast growth factor (FGF) signaling is critical for hypoblast specification. Revisiting FGF signaling in human embryos revealed that inhibition in the early blastocyst suppresses hypoblast formation. <em>In vitro</em>, the induction of hypoblast is synergistically enhanced by limiting trophectoderm and epiblast fates. This finding revises previous reports and establishes a conservation in lineage specification between mice and humans. Overall, this study demonstrates the utility of human naive PSC-based models in elucidating the mechanistic features of early human embryogenesis.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":"41 1","pages":""},"PeriodicalIF":23.9,"publicationDate":"2024-05-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141185424","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}
Aging generally predisposes stem cells to functional decline, impairing tissue homeostasis. Here, we report that hematopoietic stem cells (HSCs) acquire metabolic resilience that promotes cell survival. High-resolution real-time ATP analysis with glucose tracing and metabolic flux analysis revealed that old HSCs reprogram their metabolism to activate the pentose phosphate pathway (PPP), becoming more resistant to oxidative stress and less dependent on glycolytic ATP production at steady state. As a result, old HSCs can survive without glycolysis, adapting to the physiological cytokine environment in bone marrow. Mechanistically, old HSCs enhance mitochondrial complex II metabolism during stress to promote ATP production. Furthermore, increased succinate dehydrogenase assembly factor 1 (SDHAF1) in old HSCs, induced by physiological low-concentration thrombopoietin (TPO) exposure, enables rapid mitochondrial ATP production upon metabolic stress, thereby improving survival. This study provides insight into the acquisition of resilience through metabolic reprogramming in old HSCs and its molecular basis to ameliorate age-related hematopoietic abnormalities.
衰老通常会导致干细胞功能衰退,损害组织稳态。在这里,我们报告了造血干细胞(HSCs)获得了促进细胞存活的代谢恢复能力。通过葡萄糖追踪和代谢通量分析进行的高分辨率实时ATP分析表明,老年造血干细胞重塑了新陈代谢,激活了磷酸戊糖途径(PPP),变得更能抵抗氧化应激,在稳态时对糖酵解ATP生成的依赖性降低。因此,老年造血干细胞可以在没有糖酵解的情况下存活,从而适应骨髓中的细胞因子生理环境。从机制上讲,老年造血干细胞在应激时会增强线粒体复合体 II 的代谢,以促进 ATP 的产生。此外,在生理性低浓度血小板生成素(TPO)暴露的诱导下,老年造血干细胞中琥珀酸脱氢酶组装因子1(SDHAF1)的增加可使线粒体在代谢应激时快速产生ATP,从而提高存活率。这项研究深入探讨了老年造血干细胞通过代谢重编程获得复原力的过程及其分子基础,从而改善与年龄相关的造血异常。
{"title":"SDHAF1 confers metabolic resilience to aging hematopoietic stem cells by promoting mitochondrial ATP production","authors":"Shintaro Watanuki, Hiroshi Kobayashi, Yuki Sugiura, Masamichi Yamamoto, Daiki Karigane, Kohei Shiroshita, Yuriko Sorimachi, Takayuki Morikawa, Shinya Fujita, Kotaro Shide, Miho Haraguchi, Shinpei Tamaki, Takumi Mikawa, Hiroshi Kondoh, Hiroyasu Nakano, Kenta Sumiyama, Go Nagamatsu, Nobuhito Goda, Shinichiro Okamoto, Ayako Nakamura-Ishizu, Keiyo Takubo","doi":"10.1016/j.stem.2024.04.023","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.023","url":null,"abstract":"<p>Aging generally predisposes stem cells to functional decline, impairing tissue homeostasis. Here, we report that hematopoietic stem cells (HSCs) acquire metabolic resilience that promotes cell survival. High-resolution real-time ATP analysis with glucose tracing and metabolic flux analysis revealed that old HSCs reprogram their metabolism to activate the pentose phosphate pathway (PPP), becoming more resistant to oxidative stress and less dependent on glycolytic ATP production at steady state. As a result, old HSCs can survive without glycolysis, adapting to the physiological cytokine environment in bone marrow. Mechanistically, old HSCs enhance mitochondrial complex II metabolism during stress to promote ATP production. Furthermore, increased succinate dehydrogenase assembly factor 1 (SDHAF1) in old HSCs, induced by physiological low-concentration thrombopoietin (TPO) exposure, enables rapid mitochondrial ATP production upon metabolic stress, thereby improving survival. This study provides insight into the acquisition of resilience through metabolic reprogramming in old HSCs and its molecular basis to ameliorate age-related hematopoietic abnormalities.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":"15 1","pages":""},"PeriodicalIF":23.9,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141069209","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}
Pub Date : 2024-05-20DOI: 10.1016/j.stem.2024.04.022
Kun Zhang, Ping Wan, Liren Wang, Zhen Wang, Fangzhi Tan, Jie Li, Xiaolong Ma, Jin Cen, Xiang Yuan, Yang Liu, Zhen Sun, Xi Cheng, Yuanhua Liu, Xuhao Liu, Jiazhi Hu, Guisheng Zhong, Dali Li, Qiang Xia, Lijian Hui
Cell-based ex vivo gene therapy in solid organs, especially the liver, has proven technically challenging. Here, we report a feasible strategy for the clinical application of hepatocyte therapy. We first generated high-quality autologous hepatocytes through the large-scale expansion of patient-derived hepatocytes. Moreover, the proliferating patient-derived hepatocytes, together with the AAV2.7m8 variant identified through screening, enabled CRISPR-Cas9-mediated targeted integration efficiently, achieving functional correction of pathogenic mutations in FAH or OTC. Importantly, these edited hepatocytes repopulated the injured mouse liver at high repopulation levels and underwent maturation, successfully treating mice with tyrosinemia following transplantation. Our study combines ex vivo large-scale cell expansion and gene editing in patient-derived transplantable hepatocytes, which holds potential for treating human liver diseases.
{"title":"Efficient expansion and CRISPR-Cas9-mediated gene correction of patient-derived hepatocytes for treatment of inherited liver diseases","authors":"Kun Zhang, Ping Wan, Liren Wang, Zhen Wang, Fangzhi Tan, Jie Li, Xiaolong Ma, Jin Cen, Xiang Yuan, Yang Liu, Zhen Sun, Xi Cheng, Yuanhua Liu, Xuhao Liu, Jiazhi Hu, Guisheng Zhong, Dali Li, Qiang Xia, Lijian Hui","doi":"10.1016/j.stem.2024.04.022","DOIUrl":"https://doi.org/10.1016/j.stem.2024.04.022","url":null,"abstract":"<p>Cell-based <em>ex vivo</em> gene therapy in solid organs, especially the liver, has proven technically challenging. Here, we report a feasible strategy for the clinical application of hepatocyte therapy. We first generated high-quality autologous hepatocytes through the large-scale expansion of patient-derived hepatocytes. Moreover, the proliferating patient-derived hepatocytes, together with the AAV2.7m8 variant identified through screening, enabled CRISPR-Cas9-mediated targeted integration efficiently, achieving functional correction of pathogenic mutations in FAH or OTC. Importantly, these edited hepatocytes repopulated the injured mouse liver at high repopulation levels and underwent maturation, successfully treating mice with tyrosinemia following transplantation. Our study combines <em>ex vivo</em> large-scale cell expansion and gene editing in patient-derived transplantable hepatocytes, which holds potential for treating human liver diseases.</p>","PeriodicalId":9665,"journal":{"name":"Cell stem cell","volume":"20 1","pages":""},"PeriodicalIF":23.9,"publicationDate":"2024-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141069215","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}