首页 > 最新文献

Stem Cell Research & Therapy最新文献

英文 中文
Human umbilical mesenchymal stem cells ameliorate atrophic gastritis in aging mice by participating in mitochondrial autophagy through Ndufs8 signaling. 人脐带间充质干细胞通过Ndufs8信号参与线粒体自噬,改善衰老小鼠萎缩性胃炎。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-12-20 DOI: 10.1186/s13287-024-04094-4
Qiang Rui, Chuyu Li, Yiqi Rui, Chuanzhuo Zhang, Cunbing Xia, Qing Wang, Yuanyuan Liu, Peng Wang

Background: Chronic atrophic gastritis (CAG) is a chronic disease of the gastric mucosa characterized by a reduction or an absolute disappearance of the original gastric glands, possibly replaced by pseudopyloric fibrosis, intestinal metaplasia, or fibrosis. CAG develops progressively into intestinal epithelial metaplasia, dysplasia, and ultimately, gastric cancer. Epidemiological statistics have revealed a positive correlation between the incidence of CAG and age. Mesenchymal stem cells (MSCs) are a type of adult stem cells derived from mesoderm, with strong tissue repair capabilities. Therefore, the restoration of the gastric mucosa may serve as an efficacious strategy to ameliorate CAG and avert gastric cancer. However, the mechanisms by which MSCs inhibit the relentless progression of aging atrophic gastritis remain to be elucidated. This study endeavored to assess a novel approach utilizing MSCs to treat CAG and forestall carcinogenics.

Methods: In this study, we selected mice with atrophic gastritis from naturally aging mice and administered human umbilical cord-derived mesenchymal stem cells (hUMSCs) via tail vein injection to evaluate the therapeutic effects of hUMSCs on age-related chronic atrophic gastritis. Initially, we employed methods such as ELISA, immunohistochemical analysis, and TUNEL assays to detect changes in the mice post-hUMSC injection. Proteomic and bioinformatics analyses were conducted to identify differentially expressed proteins, focusing on NADH: ubiquinone oxidoreductase core subunit S8 (Ndufs8). Co-culturing hUMSCs with Ndufs8 knockout gastric mucosal epithelial cells (GMECs), we utilized flow cytometry, Western blotting, real-time quantitative PCR, and immunofluorescence to investigate the mechanisms of action of hUMSCs.

Results: We observed that hUMSCs are capable of migrating to and repairing damaged gastric mucosa. Initially, hUMSCs significantly enhanced the secretion of gastric proteins PG-1 and G17, while concurrently reducing inflammatory cytokines. Furthermore, hUMSCs mitigated gastric fibrosis and apoptosis in mucosal cells. Proteomic and bioinformatic analyses revealed alterations in the protein network involved in mitochondrial autophagy, with Ndufs8 playing a pivotal role. Upon knocking out Ndufs8 in GMECs, we noted mitochondrial damage and reduced autophagy, leading to an aged phenotype in GMECs. Co-culturing Ndufs8-knockout GMECs with hUMSCs demonstrated that hUMSCs could ameliorate mitochondrial dysfunction and restore the cell cycle in GMECs.

背景:慢性萎缩性胃炎(CAG)是一种胃粘膜慢性疾病,其特征是原有胃腺减少或完全消失,可能被假门纤维化、肠化生或纤维化所取代。CAG逐渐发展为肠上皮化、不典型增生,并最终发展为胃癌。流行病学统计显示CAG的发病率与年龄呈正相关。间充质干细胞(Mesenchymal stem cells, MSCs)是一种来源于中胚层的成体干细胞,具有很强的组织修复能力。因此,胃粘膜的修复可能是改善CAG和预防胃癌的有效策略。然而,MSCs抑制萎缩性胃炎无情进展的机制仍有待阐明。本研究试图评估一种利用间充质干细胞治疗CAG和预防致癌物质的新方法。方法:本研究从自然衰老小鼠中选取萎缩性胃炎小鼠,通过尾静脉注射人脐带间充质干细胞(hUMSCs),观察其对年龄相关性慢性萎缩性胃炎的治疗作用。最初,我们采用ELISA、免疫组织化学分析和TUNEL等方法检测小鼠注射humsc后的变化。通过蛋白质组学和生物信息学分析鉴定差异表达蛋白,重点关注NADH:泛醌氧化还原酶核心亚基S8 (Ndufs8)。我们将hUMSCs与Ndufs8基因敲除的胃粘膜上皮细胞(gmec)共培养,利用流式细胞术、Western blotting、实时定量PCR和免疫荧光技术研究hUMSCs的作用机制。结果:我们观察到hUMSCs能够迁移并修复受损的胃黏膜。最初,hUMSCs显著增强胃蛋白PG-1和G17的分泌,同时降低炎症因子。此外,hUMSCs减轻了胃粘膜细胞的纤维化和凋亡。蛋白质组学和生物信息学分析揭示了参与线粒体自噬的蛋白质网络的改变,其中Ndufs8起着关键作用。在敲除gmec中的Ndufs8后,我们注意到线粒体损伤和自噬减少,导致gmec中的衰老表型。ndufs8敲除的gmec与hUMSCs共培养表明,hUMSCs可以改善gmec的线粒体功能障碍,恢复细胞周期。
{"title":"Human umbilical mesenchymal stem cells ameliorate atrophic gastritis in aging mice by participating in mitochondrial autophagy through Ndufs8 signaling.","authors":"Qiang Rui, Chuyu Li, Yiqi Rui, Chuanzhuo Zhang, Cunbing Xia, Qing Wang, Yuanyuan Liu, Peng Wang","doi":"10.1186/s13287-024-04094-4","DOIUrl":"10.1186/s13287-024-04094-4","url":null,"abstract":"<p><strong>Background: </strong>Chronic atrophic gastritis (CAG) is a chronic disease of the gastric mucosa characterized by a reduction or an absolute disappearance of the original gastric glands, possibly replaced by pseudopyloric fibrosis, intestinal metaplasia, or fibrosis. CAG develops progressively into intestinal epithelial metaplasia, dysplasia, and ultimately, gastric cancer. Epidemiological statistics have revealed a positive correlation between the incidence of CAG and age. Mesenchymal stem cells (MSCs) are a type of adult stem cells derived from mesoderm, with strong tissue repair capabilities. Therefore, the restoration of the gastric mucosa may serve as an efficacious strategy to ameliorate CAG and avert gastric cancer. However, the mechanisms by which MSCs inhibit the relentless progression of aging atrophic gastritis remain to be elucidated. This study endeavored to assess a novel approach utilizing MSCs to treat CAG and forestall carcinogenics.</p><p><strong>Methods: </strong>In this study, we selected mice with atrophic gastritis from naturally aging mice and administered human umbilical cord-derived mesenchymal stem cells (hUMSCs) via tail vein injection to evaluate the therapeutic effects of hUMSCs on age-related chronic atrophic gastritis. Initially, we employed methods such as ELISA, immunohistochemical analysis, and TUNEL assays to detect changes in the mice post-hUMSC injection. Proteomic and bioinformatics analyses were conducted to identify differentially expressed proteins, focusing on NADH: ubiquinone oxidoreductase core subunit S8 (Ndufs8). Co-culturing hUMSCs with Ndufs8 knockout gastric mucosal epithelial cells (GMECs), we utilized flow cytometry, Western blotting, real-time quantitative PCR, and immunofluorescence to investigate the mechanisms of action of hUMSCs.</p><p><strong>Results: </strong>We observed that hUMSCs are capable of migrating to and repairing damaged gastric mucosa. Initially, hUMSCs significantly enhanced the secretion of gastric proteins PG-1 and G17, while concurrently reducing inflammatory cytokines. Furthermore, hUMSCs mitigated gastric fibrosis and apoptosis in mucosal cells. Proteomic and bioinformatic analyses revealed alterations in the protein network involved in mitochondrial autophagy, with Ndufs8 playing a pivotal role. Upon knocking out Ndufs8 in GMECs, we noted mitochondrial damage and reduced autophagy, leading to an aged phenotype in GMECs. Co-culturing Ndufs8-knockout GMECs with hUMSCs demonstrated that hUMSCs could ameliorate mitochondrial dysfunction and restore the cell cycle in GMECs.</p>","PeriodicalId":21876,"journal":{"name":"Stem Cell Research & Therapy","volume":"15 1","pages":"491"},"PeriodicalIF":7.1,"publicationDate":"2024-12-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11662822/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142872587","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
Impact of photobiomodulation on neural embryoid body formation from immortalized adipose-derived stem cells. 光生物调节对永生化脂肪干细胞形成神经胚状体的影响。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-12-20 DOI: 10.1186/s13287-024-04088-2
Precious Earldom Mulaudzi, Heidi Abrahamse, Anine Crous

Background: 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.

Methods: 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/cm2. Morphology, viability, metabolic activity, and differentiation following PBM treatment was analysed.

Results: The results revealed that the effects of photobiomodulation (PBM) are dose dependent. PBM, at 825 nm with a fluence of 10 J/cm2, 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/cm2 in all treatment groups at 24 and 96 hpi, CD105 increased with 825 nm at 10 J/cm2 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/cm2 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/cm2 across all wavelengths. Neu N a neuronal nuclei marker was expressed at 5 J/cm2 in all treatments at 24 hpi and over time the expression was observed in all treatment groups at 10 J/cm2.

Conclusion: In conclusion, the application of PBM at 825 nm with a fluence of 10 J/cm2 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.

背景:胚状体(EBs)是来源于干细胞的三维(3D)多细胞聚集体,在再生医学中起着关键作用。它们概括了胚胎发育早期阶段的许多关键方面,是产生各种类型干细胞(包括神经干细胞)的第一步。目前体外将干细胞分化为神经胚状体(neb)的方法取得了显著进展,但仍有局限性,需要改进。光生物调节(PBM)是一种低功率光疗法,是一种促进干细胞增殖和分化的非侵入性技术。方法:本体外研究阐明了光生物调节(PBM)在三维细胞培养环境中对永生化脂肪来源干细胞(iADSCs)向neb分化的影响。该研究利用了波长为825 nm、525 nm以及两者结合的PBM,影响分别为5和10 J/cm2。分析了PBM处理后的形态学、活力、代谢活性和分化。结果:光生物调节(PBM)的作用具有剂量依赖性。在825 nm, 10 J/cm2的作用下,PBM显著增加了神经胚状体(neb)的大小,提高了细胞活力和增殖,降低了乳酸脱氢酶(LDH)水平,表明细胞损伤最小。有趣的是,干细胞标记物cd44在24和96 hpi时以5 J/cm2的速度上调,CD105在24 hpi时以10 J/cm2的速度增加825 nm,这可能归因于neb内的异质细胞群体。Pax6表达呈瞬时激活。Nestin在825 nm和96 hpi下以10 J/cm2的速度上调,表明其促进了神经前体种群。GFAP是一种中间丝蛋白,在24和96 hpi下,在825 nm、10 J/cm2处上调。多能性标记SOX2在所有波长下均以5 J/cm2的速度表达。Neu N a神经元细胞核标记物在24 hpi时均以5 J/cm2的速度表达,随着时间的推移,所有处理组均以10 J/cm2的速度表达。结论:综上所述,在825 nm施加10 J/cm2的PBM诱导下,iADSCs向neb分化效果最佳。值得注意的是,神经元标记物Nestin显著上调,突出了PBM方法在增强神经元分化方面的潜力及其在再生医学中的应用前景。
{"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. 直接从外周血中提取的人类神经干细胞显示出与成年小鼠大脑的功能整合。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING 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

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 Rag2tm1FwaIl2rgtm1Wjl 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.

诱导多能干细胞来源的神经细胞移植是治疗神经退行性疾病的一种很有前途的策略。然而,体细胞的重编程及其随后的神经分化是复杂和耗时的,因此阻碍了自体应用。最近,基于直接转录因子的血细胞转化为诱导神经干细胞(iNSCs)已成为一种潜在的替代方法。然而,关于insc来源的神经元在体内移植后的功能知之甚少。在这里,我们通过暂时过表达转录因子SOX2和cMYC,将来自成人外周血的人iNSCs移植到未受损的成年免疫缺陷小鼠Rag2tm1FwaIl2rgtm1Wjl的海马或纹状体中。移植细胞产生稳定的移植物,由成熟神经元组成,显示广泛的神经突生长和树突棘形成。使用膜片钳记录急性切片的功能分析显示,在体内成熟12周后,大多数insc来源的细胞具有神经元独有的特性,并表现出电压依赖性离子通道电流和动作电位放电。此外,自发抑制性和兴奋性突触后电流的形成,以及基于狂犬病毒的逆行单突触追踪数据,有力地支持了移植物来源神经元的结构和功能整合。综上所述,我们的数据表明,直接来源于外周血细胞的iNSCs具有在体内实现完全功能成熟的内在能力,使其成为修复应用的替代潜在供体来源,值得进一步研究。
{"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. 由诱导多能干细胞衍生的无基质人肺类器官模拟肺损伤。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-12-18 DOI: 10.1186/s13287-024-04106-3
Bettina Budeus, Chiara Kroepel, Lisa Marie Stasch, Diana Klein

Background: 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.

Method: 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.

Results: 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.

Conclusion: 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.

背景:类器官作为一种接近生理的三维培养系统,为研究各种器官的发病机制、模拟人体器官的细胞复杂性和功能提供了新的机会。方法:我们采用一种简单易行的方法,成功地以无基质的方式生成诱导多能干细胞(iPSC)衍生的人肺类器官(LuOrg),以替代广泛使用的临床前小鼠模型,以便更详细、更接近患者地研究正常肺损伤。我们对生成的肺类器官进行了详细的形态学和分子分析,包括批量和单细胞RNA测序,并评估了我们的模型作为肺部疾病(即辐射诱导的肺损伤)的潜在体外平台的质量和稳健性。结果:利用无基质诱导多能干细胞分化的方法可以获得在形态学上很好地反映人肺靶组织的肺类器官,特别是在细胞组成方面。在辐射诱导的基因毒性应激后,研究了不同的细胞命运,并进一步揭示了不同肺细胞的辐射敏感性。最后,我们提供了辐照后在不同肺类器官细胞亚群中被诱导的细胞基因集,这些基因集可以在未来的研究中用作额外的RT反应,特别是衰老基因集。结论:通过建立这些自由浮动的LuOrgs作为一种新的、以患者为导向的癌症治疗方法研究的体外平台,特别是在实验放射肿瘤学中,不仅可以减少实验动物的数量,而且可以实现相应动物实验的充分和有意义的替代。
{"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. 多组学分析显示,脂肪源性干细胞通过分泌乳酸来抑制m1样巨噬细胞的炎症反应。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING 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

Background: 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.

Methods: 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.

Results: 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.

Conclusions: 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.

背景:脂肪源性干细胞(ADSCs)因其多种功能(包括抗炎作用)在再生医学领域得到广泛应用。ADSCs被认为通过分泌抗炎细胞因子和细胞外囊泡发挥抗炎作用。虽然最近的研究报道了代谢物具有多种生理活性,但ADSCs分泌的代谢物是否具有抗炎特性尚不清楚。在这里,我们进行了多组学分析,以研究adsc衍生的代谢物对m1样巨噬细胞的影响,m1样巨噬细胞在炎症反应中起重要作用。方法:采用毛细管电泳飞行时间质谱法测定ADSCs培养上清中代谢物的浓度。为了评估其对炎症反应的影响,将m1样巨噬细胞暴露于条件ADSC培养基或其代谢物中,并使用RNA测序检测基因表达变化。采用免疫印迹法检查代谢物如何抑制炎症过程。为了明确条件培养基中代谢物对其抗炎作用的贡献,我们从药理学上抑制代谢物的摄取,并分别用定量PCR和酶联免疫吸附法检测基因表达和肿瘤坏死因子-α浓度。结果:代谢组学分析显示培养上清中有大量乳酸。条件培养基和乳酸显著抑制或增加促炎和抗炎基因的表达。然而,测序和免疫印迹分析显示,乳酸不会诱导巨噬细胞从M1样向m2样极化。基于最近的一篇报道,乳酸的免疫抑制作用依赖于表观遗传重编程,研究了组蛋白乙酰化,并上调了H3K27ac的表达。此外,7ACC2特异性抑制单羧酸转运蛋白1,显著抑制条件ADSC培养基对m1样巨噬细胞的抗炎作用。结论:我们的研究结果表明,ADSCs通过分泌乳酸来抑制m1样巨噬细胞的促炎作用。这项研究增加了我们对代谢物重要性的理解,也有望阐明ADSC治疗的新机制。
{"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. FAM96B负向调控FOSL1,通过铁下垂调节牙周韧带干细胞的成骨分化和再生。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING 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

Background: 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.

Methods: 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.

Results: 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.

Conclusions: 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.

背景:以牙周韧带干细胞(PDLSC)为基础的治疗是辅助骨再生的方法之一。了解PDLSCs的功能调控及其机制是骨再生的关键问题。本研究旨在探讨序列相似96成员B家族(FAM96B)在PDLSCs功能调控中的作用。方法:采用碱性磷酸酶(ALP)活性测定、茜素红染色、定量钙分析、成骨标志物检测等方法评价PDLSCs的成骨分化。将PDLSCs移植到裸鼠背下和大鼠颅骨缺损内。然后,使用fam96b过表达的PDLSCs进行rna测序和生物信息学分析。为了评估PDLSCs的铁下垂,检测细胞质活性氧(ROS)、谷胱甘肽过氧化物酶4 (GPX4)的表达、线粒体形态和功能,包括线粒体ROS、线粒体膜电位和线粒体呼吸。结果:FAM96B降低了PDLSCs的成骨指标ALP活性、矿化水平和骨钙素表达,表明FAM96B抑制了PDLSCs的成骨分化。FAM96B基因敲低可促进裸鼠背皮下移植PDLSCs的新骨形成。然后通过rna测序检测相关生物学功能,重点研究铁下垂。FAM96B可提高PDLSCs的胞浆ROS水平,抑制GPX4的表达和线粒体功能。因此,FAM96B促进了PDLSCs的铁下垂。同时,我们发现FAM96B抑制上调靶基因FOS如1,AP-1转录因子亚单位(FOSL1)的表达,FOSL1促进PDLSCs体外成骨分化。FOSL1对裸鼠背部皮下移植和大鼠颅骨缺损移植的PDLSCs新生骨形成也有促进作用。进而证实FOSL1对铁下垂的抑制作用。结论:FAM96B缺失促进了PDLSCs的成骨分化,抑制了铁下沉。FAM96B负向调控下游基因FOSL1, FOSL1通过铁下垂促进PDLSCs成骨分化。因此,我们的研究结果为理解FAM96B-FOSL1轴作为MSC介导的骨再生靶标提供了基础。
{"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. 谷氨酰胺-αKG轴通过IGF2 m6A修饰影响牙本质再生,调控间充质成体干细胞成骨/牙源性分化。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING 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

Background: 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.

Methods: 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.

Results: 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.

Conclusion: 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.

背景:间充质成体干细胞(mesenchal adult stem cells, m-ASCs)的多系分化对组织再生至关重要,并伴随着代谢重编程,其中牙髓来源的m-ASCs具有明显的易获取优势。干细胞命运的决定和分化与细胞微环境中的代谢状态密切相关,并与表观遗传修饰积极相互作用。近年来,谷氨酰胺-α-酮戊二酸(αKG)轴被证实与衰老、肿瘤发生、成骨等有关,但其在m-ASCs中的作用尚缺乏足够的研究证据。方法:采用代谢组学分析方法,探讨牙髓源性m-ASCs分化过程中代谢物的变化规律。建立小鼠切牙夹断模型,探讨αKG对牙组织修复的影响。利用shRNA技术敲低相关关键酶脱氢酶1(GLUD1)的表达。利用RNA-seq、m6A评价和MeRIP-qPCR技术深入研究其潜在的表观遗传机制。结果:我们发现谷氨酰胺-αKG轴随着牙髓源性m-ASCs的成骨/牙源性分化呈增加趋势,与GLUD1的表达模式一致。关键代谢物αKG能够加速小鼠切牙的修复,促进牙本质的形成。外源性DM-αKG被证明能够促进牙髓源性m-ASCs向骨/牙源性分化,而通过敲低GLUD1抑制谷氨酰胺源性αKG水平则具有相反的作用。在GLUD1敲低的情况下,通过RNA-seq筛选出细胞外基质(extracellular matrix, ECM)功能和PI3k-Akt信号通路广泛参与该过程,胰岛素样生长因子2 (insulin-like growth factor 2, IGF2)参与。抑制谷氨酰胺-αKG轴可能通过m6A甲基化影响IGF2的翻译效率,补充αKG可显著恢复IGF2的翻译效率。结论:谷氨酰胺-αKG轴可能通过表观遗传促进牙髓源性间质干细胞成骨/牙本质分化和牙本质再生,为潜在的牙组织修复治疗方法或基于代谢物的药物研究提供了新的研究视角。
{"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. 基于间充质干细胞衍生外泌体治疗肝损伤的研究进展。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING Pub Date : 2024-12-18 DOI: 10.1186/s13287-024-04087-3
Changlong Hu, Lin Wang

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.

间充质干细胞(MSCs)在肝损伤治疗中显示出巨大的潜在作用。MSCs可以通过分化为肝细胞促进肝脏再生,也可以分泌外泌体参与肝损伤的修复。越来越多的证据表明,间充质干细胞来源的外泌体(MSC-EXOs)在治疗肝损伤中起着重要作用。本文结合近年来的研究成果,对外泌体的生物学发生、功能以及msc - exo的特点进行了综述。msc - exo在肝纤维化、肝衰竭、肝细胞癌、氧化应激、脂质脂肪变性等肝损伤中具有重要作用,并参与肝脏再生过程。
{"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. 人类iPS细胞衍生的胰腺祖细胞分化和冷冻保存的时间效率策略:向实际应用迈进。
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING 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

Background: 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.

Methods: 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.

Results: 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.

Conclusions: 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.

Trial registration: 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).

背景:患者特异性诱导多能干细胞(iPS)的分化有助于研究个体对药物的敏感性。然而,分化方案是耗时的,并不是所有的组织都被研究过。目前关于iPS细胞胰腺外分泌分化的研究较少,对iPS细胞的培养和低温保存也知之甚少。方法:将2例儿童克罗恩病患者的iPS细胞分化为胰腺祖细胞和外分泌细胞,调整并缩短了胚胎干细胞分化方案。我们分别用qPCR和免疫荧光分析了分化过程中关键基因和关键蛋白的表达。我们探索了将已分化的细胞保存在培养中并冷冻解冻的可能性,以缩短分化所需的时间。流式细胞术分析未分化细胞和已分化细胞的细胞周期。结果:胰腺关键分化基因PDX1和胰淀粉酶mRNA水平分析表明,iPS细胞成功分化为表达PDX1的胰腺外分泌细胞(单向方差分析p)。结论:本研究优化了患者特异性胰腺分化细胞的生成,为建立患者特异性胰腺细胞系库奠定了基础,可用于量身定制的药理学分析。试验注册:该研究由母婴健康研究所IRCCS Burlo Garofolo伦理委员会批准,批准号1556(内部ID RC 44/22)。
{"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
Exosome-based therapies for inflammatory disorders: a review of recent advances. 基于外泌体的炎症性疾病治疗:最新进展综述
IF 7.1 2区 医学 Q1 CELL & TISSUE ENGINEERING 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

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.

外泌体是细胞分泌的小细胞外囊泡,在不同的生物医学领域已成为细胞间通讯和治疗干预的焦点介质。炎症性疾病,包括炎症性肠病、急性肝损伤、肺损伤、神经炎症和心肌梗死,是复杂的疾病,需要创新的治疗方法。本文综述了基于外泌体治疗炎症性疾病的最新进展,强调了它们作为诊断生物标志物和治疗药物的潜力。在炎性疾病的临床前模型中,外泌体在减少炎症、促进组织修复和改善功能结果方面显示出了希望。然而,需要进一步的研究来克服与外泌体分离、表征和递送相关的挑战,并充分了解它们的作用机制。目前外泌体研究的局限性和未来的研究方向强调需要加强分离技术和更深入的机制见解,以充分利用外泌体在临床应用中的治疗潜力。尽管存在这些挑战,但基于外泌体的疗法在治疗炎症性疾病方面具有巨大的潜力,并可能为个性化药物治疗提供新的范例。
{"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
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