首页 > 最新文献

Inflammation and regeneration最新文献

英文 中文
The oral-gut axis: a missing piece in the IBD puzzle. 口腔肠道轴:IBD难题中缺失的一块。
Pub Date : 2023-11-06 DOI: 10.1186/s41232-023-00304-3
Sho Kitamoto, Nobuhiko Kamada

Inflammatory bowel disease (IBD) is a multifactorial intractable intestinal disease. Focusing on only one facet of the pathogenesis of IBD is insufficient to fully capture the complexity of the disease, and results in limited advance in clinical management. Therefore, it is critical to dissect the interactions amongst the multifarious contributors to the pathogenesis to comprehensively understand its pathology and subsequently improve clinical outcomes. In this context, the systemic interactions between organs, particularly the oral-gut axis mediated by host immune cells and resident microorganisms, have garnered significant attention in IBD research. More specifically, periodontal disease such as periodontitis has been implicated in augmenting intestinal inflammation beyond the confines of the oral cavity. There is mounting evidence suggesting that potentially harmful oral resident bacteria, termed pathobionts, and pro-inflammatory immune cells from the oral mucosa can migrate to the gastrointestinal tract, thereby potentiating intestinal inflammation. This article aims to provide a holistic overview of the causal relationship between periodontal disease and intestinal inflammation. Furthermore, we will discuss potential determinants that facilitate the translocation of oral pathobionts into the gut, a key event underpinning the oral-gut axis. Unraveling the complex dynamics of microbiota and immunity in the oral-gut continuum will lead to a better understanding of the pathophysiology inherent in both oral and intestinal diseases and the development of prospective therapeutic strategies.

炎症性肠病(IBD)是一种多因素的难治性肠病。仅仅关注IBD发病机制的一个方面不足以充分了解疾病的复杂性,并且在临床管理方面进展有限。因此,剖析发病机制的多种因素之间的相互作用,以全面了解其病理学并随后改善临床结果,这一点至关重要。在这种情况下,器官之间的系统性相互作用,特别是由宿主免疫细胞和常驻微生物介导的口腔肠道轴,在IBD研究中引起了极大的关注。更具体地说,牙周炎等牙周病与口腔以外的肠道炎症有关。越来越多的证据表明,口腔黏膜中潜在有害的口腔常驻细菌(称为致病菌)和促炎免疫细胞可以迁移到胃肠道,从而增强肠道炎症。本文旨在全面概述牙周病和肠道炎症之间的因果关系。此外,我们将讨论促进口腔病理生物转移到肠道的潜在决定因素,这是支撑口腔肠道轴的关键事件。解开口腔肠道连续体中微生物群和免疫的复杂动力学,将有助于更好地了解口腔和肠道疾病固有的病理生理学,并制定前瞻性的治疗策略。
{"title":"The oral-gut axis: a missing piece in the IBD puzzle.","authors":"Sho Kitamoto, Nobuhiko Kamada","doi":"10.1186/s41232-023-00304-3","DOIUrl":"10.1186/s41232-023-00304-3","url":null,"abstract":"<p><p>Inflammatory bowel disease (IBD) is a multifactorial intractable intestinal disease. Focusing on only one facet of the pathogenesis of IBD is insufficient to fully capture the complexity of the disease, and results in limited advance in clinical management. Therefore, it is critical to dissect the interactions amongst the multifarious contributors to the pathogenesis to comprehensively understand its pathology and subsequently improve clinical outcomes. In this context, the systemic interactions between organs, particularly the oral-gut axis mediated by host immune cells and resident microorganisms, have garnered significant attention in IBD research. More specifically, periodontal disease such as periodontitis has been implicated in augmenting intestinal inflammation beyond the confines of the oral cavity. There is mounting evidence suggesting that potentially harmful oral resident bacteria, termed pathobionts, and pro-inflammatory immune cells from the oral mucosa can migrate to the gastrointestinal tract, thereby potentiating intestinal inflammation. This article aims to provide a holistic overview of the causal relationship between periodontal disease and intestinal inflammation. Furthermore, we will discuss potential determinants that facilitate the translocation of oral pathobionts into the gut, a key event underpinning the oral-gut axis. Unraveling the complex dynamics of microbiota and immunity in the oral-gut continuum will lead to a better understanding of the pathophysiology inherent in both oral and intestinal diseases and the development of prospective therapeutic strategies.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"43 1","pages":"54"},"PeriodicalIF":0.0,"publicationDate":"2023-11-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10626704/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71490446","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Preadipocytes in human granulation tissue: role in wound healing and response to macrophage polarization. 人类肉芽组织中的前脂肪细胞:在伤口愈合和巨噬细胞极化反应中的作用。
Pub Date : 2023-10-31 DOI: 10.1186/s41232-023-00302-5
Tina Rauchenwald, Florian Handle, Catherine E Connolly, Antonia Degen, Christof Seifarth, Martin Hermann, Christoph H Tripp, Doris Wilflingseder, Susanne Lobenwein, Dragana Savic, Leo Pölzl, Evi M Morandi, Dolores Wolfram, Ira-Ida Skvortsova, Patrizia Stoitzner, Johannes Haybaeck, Marko Konschake, Gerhard Pierer, Christian Ploner

Background: Chronic non-healing wounds pose a global health challenge. Under optimized conditions, skin wounds heal by the formation of scar tissue. However, deregulated cell activation leads to persistent inflammation and the formation of granulation tissue, a type of premature scar tissue without epithelialization. Regenerative cells from the wound periphery contribute to the healing process, but little is known about their cellular fate in an inflammatory, macrophage-dominated wound microenvironment.

Methods: We examined CD45-/CD31-/CD34+ preadipocytes and CD68+ macrophages in human granulation tissue from pressure ulcers (n=6) using immunofluorescence, immunohistochemistry, and flow cytometry. In vitro, we studied macrophage-preadipocyte interactions using primary human adipose-derived stem cells (ASCs) exposed to conditioned medium harvested from IFNG/LPS (M1)- or IL4/IL13 (M2)-activated macrophages. Macrophages were derived from THP1 cells or CD14+ monocytes. In addition to confocal microscopy and flow cytometry, ASCs were analyzed for metabolic (OXPHOS, glycolysis), morphological (cytoskeleton), and mitochondrial (ATP production, membrane potential) changes. Angiogenic properties of ASCs were determined by HUVEC-based angiogenesis assay. Protein and mRNA levels were assessed by immunoblotting and quantitative RT-PCR.

Results: CD45-/CD31-/CD34+ preadipocytes were observed with a prevalence of up to 1.5% of total viable cells in human granulation tissue. Immunofluorescence staining suggested a spatial proximity of these cells to CD68+ macrophages in vivo. In vitro, ASCs exposed to M1, but not to M2 macrophage secretome showed a pro-fibrotic response characterized by stress fiber formation, elevated alpha smooth muscle actin (SMA), and increased expression of integrins ITGA5 and ITGAV. Macrophage-secreted IL1B and TGFB1 mediated this response via the PI3K/AKT and p38-MAPK pathways. In addition, ASCs exposed to M1-inflammatory stress demonstrated reduced migration, switched to a glycolysis-dominated metabolism with reduced ATP production, and increased levels of inflammatory cytokines such as IL1B, IL8, and MCP1. Notably, M1 but not M2 macrophages enhanced the angiogenic potential of ASCs.

Conclusion: Preadipocyte fate in wound tissue is influenced by macrophage polarization. Pro-inflammatory M1 macrophages induce a pro-fibrotic response in ASCs through IL1B and TGFB1 signaling, while anti-inflammatory M2 macrophages have limited effects. These findings shed light on cellular interactions in chronic wounds and provide important information for the potential therapeutic use of ASCs in human wound healing.

背景:慢性不愈合的伤口对全球健康构成挑战。在优化的条件下,皮肤伤口通过形成疤痕组织而愈合。然而,失调的细胞活化会导致持续的炎症和肉芽组织的形成,肉芽组织是一种没有上皮化的过早瘢痕组织。伤口周围的再生细胞有助于愈合过程,但对它们在炎症、巨噬细胞主导的伤口微环境中的细胞命运知之甚少。方法:采用免疫荧光、免疫组织化学和流式细胞术检测6例压疮肉芽组织中CD45-/CD31-/CD34+前脂肪细胞和CD68+巨噬细胞。在体外,我们使用暴露于从IFNG/LPS(M1)或IL4/IL13(M2)激活的巨噬细胞获得的条件培养基的原代人脂肪衍生干细胞(ASCs)研究了巨噬细胞-前脂肪细胞的相互作用。巨噬细胞来源于THP1细胞或CD14+单核细胞。除了共聚焦显微镜和流式细胞术外,还分析了ASCs的代谢(OXPHOS、糖酵解)、形态(细胞骨架)和线粒体(ATP产生、膜电位)变化。通过基于HUVEC的血管生成测定测定ASCs的血管生成特性。通过免疫印迹和定量RT-PCR评估蛋白质和mRNA水平。结果:CD45-/CD31-/CD34+前脂肪细胞在人肉芽组织中的存在率高达总活细胞的1.5%。免疫荧光染色显示这些细胞与体内CD68+巨噬细胞在空间上接近。在体外,暴露于M1而非M2巨噬细胞分泌组的ASCs表现出促纤维化反应,其特征是应激纤维形成、α-平滑肌肌动蛋白(SMA)升高以及整合素ITGA5和ITGAV的表达增加。巨噬细胞分泌的IL1B和TGFB1通过PI3K/AKT和p38MAPK途径介导这种反应。此外,暴露于M1炎症应激的ASCs表现出迁移减少,转变为糖酵解主导的代谢,ATP产生减少,炎性细胞因子如IL1B、IL8和MCP1水平增加。值得注意的是,M1而不是M2巨噬细胞增强了ASCs的血管生成潜力。结论:创伤组织中前脂肪细胞的命运受巨噬细胞极化的影响。促炎性M1巨噬细胞通过IL1B和TGFB1信号在ASCs中诱导促纤维化反应,而抗炎性M2巨噬细胞的作用有限。这些发现揭示了慢性伤口中的细胞相互作用,并为ASCs在人类伤口愈合中的潜在治疗用途提供了重要信息。
{"title":"Preadipocytes in human granulation tissue: role in wound healing and response to macrophage polarization.","authors":"Tina Rauchenwald,&nbsp;Florian Handle,&nbsp;Catherine E Connolly,&nbsp;Antonia Degen,&nbsp;Christof Seifarth,&nbsp;Martin Hermann,&nbsp;Christoph H Tripp,&nbsp;Doris Wilflingseder,&nbsp;Susanne Lobenwein,&nbsp;Dragana Savic,&nbsp;Leo Pölzl,&nbsp;Evi M Morandi,&nbsp;Dolores Wolfram,&nbsp;Ira-Ida Skvortsova,&nbsp;Patrizia Stoitzner,&nbsp;Johannes Haybaeck,&nbsp;Marko Konschake,&nbsp;Gerhard Pierer,&nbsp;Christian Ploner","doi":"10.1186/s41232-023-00302-5","DOIUrl":"10.1186/s41232-023-00302-5","url":null,"abstract":"<p><strong>Background: </strong>Chronic non-healing wounds pose a global health challenge. Under optimized conditions, skin wounds heal by the formation of scar tissue. However, deregulated cell activation leads to persistent inflammation and the formation of granulation tissue, a type of premature scar tissue without epithelialization. Regenerative cells from the wound periphery contribute to the healing process, but little is known about their cellular fate in an inflammatory, macrophage-dominated wound microenvironment.</p><p><strong>Methods: </strong>We examined CD45<sup>-</sup>/CD31<sup>-</sup>/CD34<sup>+</sup> preadipocytes and CD68<sup>+</sup> macrophages in human granulation tissue from pressure ulcers (n=6) using immunofluorescence, immunohistochemistry, and flow cytometry. In vitro, we studied macrophage-preadipocyte interactions using primary human adipose-derived stem cells (ASCs) exposed to conditioned medium harvested from IFNG/LPS (M1)- or IL4/IL13 (M2)-activated macrophages. Macrophages were derived from THP1 cells or CD14<sup>+</sup> monocytes. In addition to confocal microscopy and flow cytometry, ASCs were analyzed for metabolic (OXPHOS, glycolysis), morphological (cytoskeleton), and mitochondrial (ATP production, membrane potential) changes. Angiogenic properties of ASCs were determined by HUVEC-based angiogenesis assay. Protein and mRNA levels were assessed by immunoblotting and quantitative RT-PCR.</p><p><strong>Results: </strong>CD45<sup>-</sup>/CD31<sup>-</sup>/CD34<sup>+</sup> preadipocytes were observed with a prevalence of up to 1.5% of total viable cells in human granulation tissue. Immunofluorescence staining suggested a spatial proximity of these cells to CD68<sup>+</sup> macrophages in vivo. In vitro, ASCs exposed to M1, but not to M2 macrophage secretome showed a pro-fibrotic response characterized by stress fiber formation, elevated alpha smooth muscle actin (SMA), and increased expression of integrins ITGA5 and ITGAV. Macrophage-secreted IL1B and TGFB1 mediated this response via the PI3K/AKT and p38-MAPK pathways. In addition, ASCs exposed to M1-inflammatory stress demonstrated reduced migration, switched to a glycolysis-dominated metabolism with reduced ATP production, and increased levels of inflammatory cytokines such as IL1B, IL8, and MCP1. Notably, M1 but not M2 macrophages enhanced the angiogenic potential of ASCs.</p><p><strong>Conclusion: </strong>Preadipocyte fate in wound tissue is influenced by macrophage polarization. Pro-inflammatory M1 macrophages induce a pro-fibrotic response in ASCs through IL1B and TGFB1 signaling, while anti-inflammatory M2 macrophages have limited effects. These findings shed light on cellular interactions in chronic wounds and provide important information for the potential therapeutic use of ASCs in human wound healing.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"43 1","pages":"53"},"PeriodicalIF":0.0,"publicationDate":"2023-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10617061/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71416293","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CXCL10 deficiency limits macrophage infiltration, preserves lung matrix, and enables lung growth in bronchopulmonary dysplasia. CXCL10缺乏限制巨噬细胞浸润,保留肺基质,促进支气管肺发育不良患者肺生长。
Pub Date : 2023-10-24 DOI: 10.1186/s41232-023-00301-6
Dharmesh V Hirani, Florian Thielen, Siavash Mansouri, Soula Danopoulos, Christina Vohlen, Pinar Haznedar-Karakaya, Jasmine Mohr, Rebecca Wilke, Jaco Selle, Thomas Grosch, Ivana Mizik, Margarete Odenthal, Cristina M Alvira, Celien Kuiper-Makris, Gloria S Pryhuber, Christian Pallasch, S van Koningsbruggen-Rietschel, Denise Al-Alam, Werner Seeger, Rajkumar Savai, Jörg Dötsch, Miguel A Alejandre Alcazar

Preterm infants with oxygen supplementation are at high risk for bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease. Inflammation with macrophage activation is central to the pathogenesis of BPD. CXCL10, a chemotactic and pro-inflammatory chemokine, is elevated in the lungs of infants evolving BPD and in hyperoxia-based BPD in mice. Here, we tested if CXCL10 deficiency preserves lung growth after neonatal hyperoxia by preventing macrophage activation. To this end, we exposed Cxcl10 knockout (Cxcl10-/-) and wild-type mice to an experimental model of hyperoxia (85% O2)-induced neonatal lung injury and subsequent regeneration. In addition, cultured primary human macrophages and murine macrophages (J744A.1) were treated with CXCL10 and/or CXCR3 antagonist. Our transcriptomic analysis identified CXCL10 as a central hub in the inflammatory network of neonatal mouse lungs after hyperoxia. Quantitative histomorphometric analysis revealed that Cxcl10-/- mice are in part protected from reduced alveolar. These findings were related to the preserved spatial distribution of elastic fibers, reduced collagen deposition, and protection from macrophage recruitment/infiltration to the lungs in Cxcl10-/- mice during acute injury and regeneration. Complimentary, studies with cultured human and murine macrophages showed that hyperoxia induces Cxcl10 expression that in turn triggers M1-like activation and migration of macrophages through CXCR3. Finally, we demonstrated a temporal increase of macrophage-related CXCL10 in the lungs of infants with BPD. In conclusion, our data demonstrate macrophage-derived CXCL10 in experimental and clinical BPD that drives macrophage chemotaxis through CXCR3, causing pro-fibrotic lung remodeling and arrest of alveolarization. Thus, targeting the CXCL10-CXCR3 axis could offer a new therapeutic avenue for BPD.

补充氧气的早产儿患支气管肺发育不良(BPD)的风险很高,这是一种新生儿慢性肺病。巨噬细胞活化的炎症是BPD发病机制的核心。CXCL10是一种趋化性和促炎性趋化因子,在患有BPD的婴儿肺部和小鼠中基于高氧的BPD中升高。在这里,我们测试了CXCL10缺乏是否通过阻止巨噬细胞活化来保护新生儿高氧后的肺部生长。为此,我们将Cxcl10敲除(Cxcl10-/-)和野生型小鼠暴露于高氧(85%O2)诱导的新生儿肺损伤和随后再生的实验模型中。此外,用CXCL10和/或CXCR3拮抗剂处理培养的原代人巨噬细胞和鼠巨噬细胞(J744A.1)。我们的转录组学分析确定CXCL10是高氧后新生小鼠肺部炎症网络的中心枢纽。定量组织形态计量学分析显示,Cxcl10-/-小鼠在一定程度上免受肺泡减少的影响。这些发现与Cxcl10-/-小鼠在急性损伤和再生过程中保留的弹性纤维的空间分布、减少的胶原沉积以及防止巨噬细胞募集/渗透到肺部有关。此外,对培养的人和小鼠巨噬细胞的研究表明,高氧诱导Cxcl10的表达,进而触发巨噬细胞通过CXCR3的M1样激活和迁移。最后,我们证明了患有BPD的婴儿肺部巨噬细胞相关CXCL10的时间增加。总之,我们的数据表明,在实验和临床BPD中,巨噬细胞衍生的CXCL10通过CXCR3驱动巨噬细胞趋化,导致促纤维化肺重塑和肺泡化停滞。因此,靶向CXCL10-CXCR3轴可以为BPD提供一种新的治疗途径。
{"title":"CXCL10 deficiency limits macrophage infiltration, preserves lung matrix, and enables lung growth in bronchopulmonary dysplasia.","authors":"Dharmesh V Hirani, Florian Thielen, Siavash Mansouri, Soula Danopoulos, Christina Vohlen, Pinar Haznedar-Karakaya, Jasmine Mohr, Rebecca Wilke, Jaco Selle, Thomas Grosch, Ivana Mizik, Margarete Odenthal, Cristina M Alvira, Celien Kuiper-Makris, Gloria S Pryhuber, Christian Pallasch, S van Koningsbruggen-Rietschel, Denise Al-Alam, Werner Seeger, Rajkumar Savai, Jörg Dötsch, Miguel A Alejandre Alcazar","doi":"10.1186/s41232-023-00301-6","DOIUrl":"10.1186/s41232-023-00301-6","url":null,"abstract":"<p><p>Preterm infants with oxygen supplementation are at high risk for bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease. Inflammation with macrophage activation is central to the pathogenesis of BPD. CXCL10, a chemotactic and pro-inflammatory chemokine, is elevated in the lungs of infants evolving BPD and in hyperoxia-based BPD in mice. Here, we tested if CXCL10 deficiency preserves lung growth after neonatal hyperoxia by preventing macrophage activation. To this end, we exposed Cxcl10 knockout (Cxcl10<sup>-/-</sup>) and wild-type mice to an experimental model of hyperoxia (85% O<sub>2</sub>)-induced neonatal lung injury and subsequent regeneration. In addition, cultured primary human macrophages and murine macrophages (J744A.1) were treated with CXCL10 and/or CXCR3 antagonist. Our transcriptomic analysis identified CXCL10 as a central hub in the inflammatory network of neonatal mouse lungs after hyperoxia. Quantitative histomorphometric analysis revealed that Cxcl10<sup>-/-</sup> mice are in part protected from reduced alveolar. These findings were related to the preserved spatial distribution of elastic fibers, reduced collagen deposition, and protection from macrophage recruitment/infiltration to the lungs in Cxcl10<sup>-/-</sup> mice during acute injury and regeneration. Complimentary, studies with cultured human and murine macrophages showed that hyperoxia induces Cxcl10 expression that in turn triggers M1-like activation and migration of macrophages through CXCR3. Finally, we demonstrated a temporal increase of macrophage-related CXCL10 in the lungs of infants with BPD. In conclusion, our data demonstrate macrophage-derived CXCL10 in experimental and clinical BPD that drives macrophage chemotaxis through CXCR3, causing pro-fibrotic lung remodeling and arrest of alveolarization. Thus, targeting the CXCL10-CXCR3 axis could offer a new therapeutic avenue for BPD.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"43 1","pages":"52"},"PeriodicalIF":0.0,"publicationDate":"2023-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10594718/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50159611","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Investigation of immune-related diseases using patient-derived induced pluripotent stem cells. 利用患者来源的诱导多能干细胞研究免疫相关疾病。
Pub Date : 2023-10-24 DOI: 10.1186/s41232-023-00303-4
Hirofumi Shoda, Bunki Natsumoto, Keishi Fujio

The precise pathogenesis of immune-related diseases remains unclear, and new effective therapeutic choices are required for the induction of remission or cure in these diseases. Basic research utilizing immune-related disease patient-derived induced pluripotent stem (iPS) cells is expected to be a promising platform for elucidating the pathogenesis of the diseases and for drug discovery. Since autoinflammatory diseases are usually monogenic, genetic mutations affect the cell function and patient-derived iPS cells tend to exhibit disease-specific phenotypes. In particular, iPS cell-derived monocytic cells and macrophages can be used for functional experiments, such as inflammatory cytokine production, and are often employed in research on patients with autoinflammatory diseases.On the other hand, the utilization of disease-specific iPS cells is less successful for research on autoimmune diseases. One reason for this is that autoimmune diseases are usually polygenic, which makes it challenging to determine which factors cause the phenotypes of patient-derived iPS cells are caused by. Another reason is that protocols for differentiating some lymphocytes associated with autoimmunity, such as CD4+T cells or B cells, from iPS cells have not been well established. Nevertheless, several groups have reported studies utilizing autoimmune disease patient-derived iPS cells, including patients with rheumatoid arthritis, systemic lupus erythematosus (SLE), and systemic sclerosis. Particularly, non-hematopoietic cells, such as fibroblasts and cardiomyocytes, differentiated from autoimmune patient-derived iPS cells have shown promising results for further research into the pathogenesis. Recently, our groups established a method for differentiating dendritic cells that produce interferon-alpha, which can be applied as an SLE pathological model. In summary, patient-derived iPS cells can provide a promising platform for pathological research and new drug discovery in the field of immune-related diseases.

免疫相关疾病的确切发病机制尚不清楚,需要新的有效治疗选择来诱导这些疾病的缓解或治愈。利用免疫相关疾病患者来源的诱导多能干细胞(iPS)进行的基础研究有望成为阐明疾病发病机制和药物发现的一个有前途的平台。由于自身炎症性疾病通常是单基因的,基因突变会影响细胞功能,患者来源的iPS细胞往往表现出疾病特异性表型。特别是,iPS细胞衍生的单核细胞和巨噬细胞可用于功能实验,如炎性细胞因子的产生,并经常用于对自身炎症性疾病患者的研究。另一方面,利用疾病特异性iPS细胞研究自身免疫性疾病的成功率较低。其中一个原因是自身免疫性疾病通常是多基因的,这使得确定导致患者来源的iPS细胞表型的因素具有挑战性。另一个原因在于,将一些与自身免疫相关的淋巴细胞(如CD4+T细胞或B细胞)与iPS细胞区分开来的方案尚未建立。然而,一些研究小组已经报道了利用自身免疫性疾病患者来源的iPS细胞的研究,包括类风湿性关节炎、系统性红斑狼疮(SLE)和系统性硬化症患者。特别是,从自身免疫性患者来源的iPS细胞分化而来的非造血细胞,如成纤维细胞和心肌细胞,已显示出对发病机制的进一步研究的有希望的结果。最近,我们的小组建立了一种分化产生干扰素α的树突细胞的方法,该方法可作为SLE的病理模型。总之,患者来源的iPS细胞可以为免疫相关疾病领域的病理研究和新药发现提供一个有前景的平台。
{"title":"Investigation of immune-related diseases using patient-derived induced pluripotent stem cells.","authors":"Hirofumi Shoda, Bunki Natsumoto, Keishi Fujio","doi":"10.1186/s41232-023-00303-4","DOIUrl":"10.1186/s41232-023-00303-4","url":null,"abstract":"<p><p>The precise pathogenesis of immune-related diseases remains unclear, and new effective therapeutic choices are required for the induction of remission or cure in these diseases. Basic research utilizing immune-related disease patient-derived induced pluripotent stem (iPS) cells is expected to be a promising platform for elucidating the pathogenesis of the diseases and for drug discovery. Since autoinflammatory diseases are usually monogenic, genetic mutations affect the cell function and patient-derived iPS cells tend to exhibit disease-specific phenotypes. In particular, iPS cell-derived monocytic cells and macrophages can be used for functional experiments, such as inflammatory cytokine production, and are often employed in research on patients with autoinflammatory diseases.On the other hand, the utilization of disease-specific iPS cells is less successful for research on autoimmune diseases. One reason for this is that autoimmune diseases are usually polygenic, which makes it challenging to determine which factors cause the phenotypes of patient-derived iPS cells are caused by. Another reason is that protocols for differentiating some lymphocytes associated with autoimmunity, such as CD4<sup>+</sup>T cells or B cells, from iPS cells have not been well established. Nevertheless, several groups have reported studies utilizing autoimmune disease patient-derived iPS cells, including patients with rheumatoid arthritis, systemic lupus erythematosus (SLE), and systemic sclerosis. Particularly, non-hematopoietic cells, such as fibroblasts and cardiomyocytes, differentiated from autoimmune patient-derived iPS cells have shown promising results for further research into the pathogenesis. Recently, our groups established a method for differentiating dendritic cells that produce interferon-alpha, which can be applied as an SLE pathological model. In summary, patient-derived iPS cells can provide a promising platform for pathological research and new drug discovery in the field of immune-related diseases.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"43 1","pages":"51"},"PeriodicalIF":0.0,"publicationDate":"2023-10-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10594759/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"50159612","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hepatocyte growth factor pretreatment boosts functional recovery after spinal cord injury through human iPSC-derived neural stem/progenitor cell transplantation. 肝细胞生长因子预处理通过人iPSC衍生的神经干/祖细胞移植促进脊髓损伤后的功能恢复。
Pub Date : 2023-10-16 DOI: 10.1186/s41232-023-00298-y
Yu Suematsu, Narihito Nagoshi, Munehisa Shinozaki, Yoshitaka Kase, Yusuke Saijo, Shogo Hashimoto, Takahiro Shibata, Keita Kajikawa, Yasuhiro Kamata, Masahiro Ozaki, Kaori Yasutake, Tomoko Shindo, Shinsuke Shibata, Morio Matsumoto, Masaya Nakamura, Hideyuki Okano

Background: Human induced pluripotent stem cell-derived neural stem/progenitor cell (hiPSC-NS/PC)-based cell transplantation has emerged as a groundbreaking method for replacing damaged neural cells and stimulating functional recovery, but its efficacy is strongly influenced by the state of the injured spinal microenvironment. This study evaluates the impact of a dual therapeutic intervention utilizing hepatocyte growth factor (HGF) and hiPSC-NS/PC transplantation on motor function restoration following spinal cord injury (SCI).

Methods: Severe contusive SCI was induced in immunocompromised rats, followed by continuous administration of recombinant human HGF protein into the subarachnoid space immediately after SCI for two weeks. Acute-phase histological and RNA sequencing analyses were conducted. Nine days after the injury, hiPSC-NS/PCs were transplanted into the lesion epicenter of the injured spinal cord, and the functional and histological outcomes were determined.

Results: The acute-phase HGF-treated group exhibited vascularization, diverse anti-inflammatory effects, and activation of endogenous neural stem cells after SCI, which collectively contributed to tissue preservation. Following cell transplantation into a favorable environment, the transplanted NS/PCs survived well, facilitating remyelination and neuronal regeneration in host tissues. These comprehensive effects led to substantial enhancements in motor function in the dual-therapy group compared to the single-treatment groups.

Conclusions: We demonstrate that the combined therapeutic approach of HGF preconditioning and hiPSC-NS/PC transplantation enhances locomotor functional recovery post-SCI, highlighting a highly promising therapeutic strategy for acute to subacute SCI.

背景:基于人诱导多能干细胞衍生的神经干/祖细胞(hiPSC NS/PC)的细胞移植已成为一种替代受损神经细胞和刺激功能恢复的开创性方法,但其疗效受到损伤脊髓微环境状态的强烈影响。本研究评估了肝细胞生长因子(HGF)和hiPSC NS/PC移植双重治疗干预对脊髓损伤(SCI)后运动功能恢复的影响,随后在SCI后立即将重组人HGF蛋白连续给予蛛网膜下腔两周。进行急性期组织学和RNA测序分析。损伤后9天,将hiPSC NS/PCs移植到损伤脊髓的病变中心,并确定功能和组织学结果。结果:急性期HGF治疗组在SCI后表现出血管化、多种抗炎作用和内源性神经干细胞的激活,这些共同有助于组织保存。在将细胞移植到有利的环境中后,移植的NS/PCs存活良好,促进了宿主组织中的髓鞘再生和神经元再生。与单一治疗组相比,这些综合作用使双重治疗组的运动功能显著增强。结论:我们证明了HGF预处理和hiPSC NS/PC移植的联合治疗方法可以增强SCI后的运动功能恢复,突出了一种非常有前途的急性至亚急性SCI治疗策略。
{"title":"Hepatocyte growth factor pretreatment boosts functional recovery after spinal cord injury through human iPSC-derived neural stem/progenitor cell transplantation.","authors":"Yu Suematsu, Narihito Nagoshi, Munehisa Shinozaki, Yoshitaka Kase, Yusuke Saijo, Shogo Hashimoto, Takahiro Shibata, Keita Kajikawa, Yasuhiro Kamata, Masahiro Ozaki, Kaori Yasutake, Tomoko Shindo, Shinsuke Shibata, Morio Matsumoto, Masaya Nakamura, Hideyuki Okano","doi":"10.1186/s41232-023-00298-y","DOIUrl":"10.1186/s41232-023-00298-y","url":null,"abstract":"<p><strong>Background: </strong>Human induced pluripotent stem cell-derived neural stem/progenitor cell (hiPSC-NS/PC)-based cell transplantation has emerged as a groundbreaking method for replacing damaged neural cells and stimulating functional recovery, but its efficacy is strongly influenced by the state of the injured spinal microenvironment. This study evaluates the impact of a dual therapeutic intervention utilizing hepatocyte growth factor (HGF) and hiPSC-NS/PC transplantation on motor function restoration following spinal cord injury (SCI).</p><p><strong>Methods: </strong>Severe contusive SCI was induced in immunocompromised rats, followed by continuous administration of recombinant human HGF protein into the subarachnoid space immediately after SCI for two weeks. Acute-phase histological and RNA sequencing analyses were conducted. Nine days after the injury, hiPSC-NS/PCs were transplanted into the lesion epicenter of the injured spinal cord, and the functional and histological outcomes were determined.</p><p><strong>Results: </strong>The acute-phase HGF-treated group exhibited vascularization, diverse anti-inflammatory effects, and activation of endogenous neural stem cells after SCI, which collectively contributed to tissue preservation. Following cell transplantation into a favorable environment, the transplanted NS/PCs survived well, facilitating remyelination and neuronal regeneration in host tissues. These comprehensive effects led to substantial enhancements in motor function in the dual-therapy group compared to the single-treatment groups.</p><p><strong>Conclusions: </strong>We demonstrate that the combined therapeutic approach of HGF preconditioning and hiPSC-NS/PC transplantation enhances locomotor functional recovery post-SCI, highlighting a highly promising therapeutic strategy for acute to subacute SCI.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"43 1","pages":"50"},"PeriodicalIF":0.0,"publicationDate":"2023-10-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10577910/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41242594","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Testosterone upregulates glial cell line-derived neurotrophic factor (GDNF) and promotes neuroinflammation to enhance glioma cell survival and proliferation. 睾酮上调神经胶质细胞源性神经营养因子(GDNF)并促进神经炎症以增强神经胶质瘤细胞的存活和增殖。
Pub Date : 2023-10-13 DOI: 10.1186/s41232-023-00300-7
Kouminin Kanwore, Konimpo Kanwore, Xiaoxiao Guo, Ying Xia, Han Zhou, Lin Zhang, Gabriel Komla Adzika, Adu-Amankwaah Joseph, Ayanlaja Abdulrahman Abiola, Peipei Mu, Piniel Alphayo Kambey, Marie Louis N'dzie Noah, DianShuai Gao

Background: Testosterone contributes to male organism development, such as bone density, muscle development, and fat repartition. Estrogen (derived from testosterone) also contributes to female reproductive system development. Here, we investigated the effect of testosterone on glioma cells and brain neuron inflammation essential for cancer development and progression.

Methods: The human astrocyte and glioma cell lines were treated with 6 ng/ml exogenous testosterone in vitro. We performed cell counting kit-8, transwell, and wound healing assays to determine the effect of testosterone on glioma cell proliferation, migration, and invasion. The glioma cells were injected into the xenograft and treated with 5 µl concentrated testosterone. Transcriptional suppression of glial cell line-derived neurotrophic factor (GDNF) was performed to evaluate brain neuron inflammation and survival. The tumor tissues were assessed by hematoxylin-eosin staining and immunohistochemistry.

Results: Testosterone upregulates GDNF to stimulate proliferation, migration, and invasion of glioma cells. Pathologically, the augmentation of GDNF and cyclophilin A contributed to neuroprotection when treated with testosterone. Our investigation showed that testosterone contributes to brain neuron and astrocyte inflammation through the upregulation of nuclear factor erythroid 2-related factor 2 (NRF2), glial fibrillary acid protein (GFAP), and sirtuin 5 (SIRT5), resulting in pro-inflammatory macrophages recruitments into the neural microenvironment. Mechanically, testosterone treatment regulates GDNF translocation from the glioma cells and astrocyte nuclei to the cytoplasm.

Conclusion: Testosterone upregulates GDNF in glioma cells and astrocytes essential for microglial proliferation, migration, and invasion. Testosterone contributes to brain tumor growth via GDNF and inflammation. The contribution of testosterone, macrophages, and astrocytes, in old neuron rescue, survival, and proliferation. During brain neuron inflammation, the organism activates and stimulates the neuron rescue through the enrichment of the old neuron microenvironment with growth factors such as GDNF, BDNF, SOX1/2, and MAPK secreted by the surrounding neurons and glial cells to maintain the damaged neuron by inflammation alive even if the axon is dead. The immune response also contributes to brain cell survival through the secretion of proinflammatory cytokines, resulting in inflammation maintenance. The rescued old neuron interaction with infiltrated macrophages contributes to angiogenesis to supplement the old neuron with more nutrients leading to metabolism activation and surrounding cell uncontrollable cell growth.

背景:睾酮有助于男性机体的发育,如骨密度、肌肉发育和脂肪重新分配。雌激素(来源于睾酮)也有助于女性生殖系统的发育。在此,我们研究了睾酮对癌症发展和进展所必需的神经胶质瘤细胞和脑神经元炎症的影响。方法:用6ng/ml外源性睾酮对人星形胶质细胞和胶质瘤细胞系进行体外处理。我们进行了细胞计数试剂盒-8、transwell和伤口愈合测定,以确定睾酮对神经胶质瘤细胞增殖、迁移和侵袭的影响。将神经胶质瘤细胞注射到异种移植物中,并用5µl浓缩睾酮处理。对神经胶质细胞源性神经营养因子(GDNF)进行转录抑制,以评估脑神经元炎症和存活率。用苏木精-伊红染色和免疫组织化学方法评价肿瘤组织。结果:睾酮上调GDNF以刺激神经胶质瘤细胞的增殖、迁移和侵袭。病理学上,当用睾酮治疗时,GDNF和亲环素A的增加有助于神经保护。我们的研究表明,睾酮通过上调核因子红系2相关因子2(NRF2)、胶质纤维酸性蛋白(GFAP)和SIRT5(SIRT5),导致促炎巨噬细胞募集到神经微环境中,从而导致脑神经元和星形胶质细胞炎症。从机制上讲,睾酮治疗调节GDNF从神经胶质瘤细胞和星形胶质细胞细胞核向细胞质的移位。结论:睾酮上调胶质瘤细胞和星形胶质细胞的GDNF,这对小胶质细胞的增殖、迁移和侵袭至关重要。睾酮通过GDNF和炎症促进脑肿瘤生长。睾酮、巨噬细胞和星形胶质细胞在老年神经元拯救、存活和增殖中的作用。在脑神经元炎症过程中,生物体通过用周围神经元和神经胶质细胞分泌的生长因子(如GDNF、BDNF、SOX1/2和MAPK)富集旧的神经元微环境来激活和刺激神经元拯救,即使轴突死亡,也能维持炎症损伤的神经元存活。免疫反应还通过分泌促炎细胞因子促进脑细胞存活,从而维持炎症。获救的老神经元与浸润的巨噬细胞的相互作用有助于血管生成,为老神经元补充更多的营养,从而导致代谢激活和周围细胞无法控制的细胞生长。
{"title":"Testosterone upregulates glial cell line-derived neurotrophic factor (GDNF) and promotes neuroinflammation to enhance glioma cell survival and proliferation.","authors":"Kouminin Kanwore, Konimpo Kanwore, Xiaoxiao Guo, Ying Xia, Han Zhou, Lin Zhang, Gabriel Komla Adzika, Adu-Amankwaah Joseph, Ayanlaja Abdulrahman Abiola, Peipei Mu, Piniel Alphayo Kambey, Marie Louis N'dzie Noah, DianShuai Gao","doi":"10.1186/s41232-023-00300-7","DOIUrl":"10.1186/s41232-023-00300-7","url":null,"abstract":"<p><strong>Background: </strong>Testosterone contributes to male organism development, such as bone density, muscle development, and fat repartition. Estrogen (derived from testosterone) also contributes to female reproductive system development. Here, we investigated the effect of testosterone on glioma cells and brain neuron inflammation essential for cancer development and progression.</p><p><strong>Methods: </strong>The human astrocyte and glioma cell lines were treated with 6 ng/ml exogenous testosterone in vitro. We performed cell counting kit-8, transwell, and wound healing assays to determine the effect of testosterone on glioma cell proliferation, migration, and invasion. The glioma cells were injected into the xenograft and treated with 5 µl concentrated testosterone. Transcriptional suppression of glial cell line-derived neurotrophic factor (GDNF) was performed to evaluate brain neuron inflammation and survival. The tumor tissues were assessed by hematoxylin-eosin staining and immunohistochemistry.</p><p><strong>Results: </strong>Testosterone upregulates GDNF to stimulate proliferation, migration, and invasion of glioma cells. Pathologically, the augmentation of GDNF and cyclophilin A contributed to neuroprotection when treated with testosterone. Our investigation showed that testosterone contributes to brain neuron and astrocyte inflammation through the upregulation of nuclear factor erythroid 2-related factor 2 (NRF2), glial fibrillary acid protein (GFAP), and sirtuin 5 (SIRT5), resulting in pro-inflammatory macrophages recruitments into the neural microenvironment. Mechanically, testosterone treatment regulates GDNF translocation from the glioma cells and astrocyte nuclei to the cytoplasm.</p><p><strong>Conclusion: </strong>Testosterone upregulates GDNF in glioma cells and astrocytes essential for microglial proliferation, migration, and invasion. Testosterone contributes to brain tumor growth via GDNF and inflammation. The contribution of testosterone, macrophages, and astrocytes, in old neuron rescue, survival, and proliferation. During brain neuron inflammation, the organism activates and stimulates the neuron rescue through the enrichment of the old neuron microenvironment with growth factors such as GDNF, BDNF, SOX1/2, and MAPK secreted by the surrounding neurons and glial cells to maintain the damaged neuron by inflammation alive even if the axon is dead. The immune response also contributes to brain cell survival through the secretion of proinflammatory cytokines, resulting in inflammation maintenance. The rescued old neuron interaction with infiltrated macrophages contributes to angiogenesis to supplement the old neuron with more nutrients leading to metabolism activation and surrounding cell uncontrollable cell growth.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"43 1","pages":"49"},"PeriodicalIF":0.0,"publicationDate":"2023-10-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10571473/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41224015","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Analysis of distribution, collection, and confirmation of capacity dependency of small extracellular vesicles toward a therapy for liver cirrhosis. 分析细胞外小泡在肝硬化治疗中的分布、收集和容量依赖性。
Pub Date : 2023-10-09 DOI: 10.1186/s41232-023-00299-x
Nobutaka Takeda, Atsunori Tsuchiya, Masaki Mito, Kazuki Natsui, Yui Natusi, Yohei Koseki, Kei Tomiyoshi, Fusako Yamazaki, Yuki Yoshida, Hiroyuki Abe, Masayuki Sano, Taketomo Kido, Yusuke Yoshioka, Junichi Kikuta, Tohru Itoh, Ken Nishimura, Masaru Ishii, Takahiro Ochiya, Atsushi Miyajima, Shuji Terai

Background: The progression of liver fibrosis leads to portal hypertension and liver dysfunction. However, no antifibrotic agents have been approved for cirrhosis to date, making them an unmet medical need. Small extracellular vesicles (sEVs) of mesenchymal stem cells (MSCs) are among these candidate agents. In this study, we investigated the effects of sEVs of MSCs, analyzed their distribution in the liver post-administration, whether their effect was dose-dependent, and whether it was possible to collect a large number of sEVs.

Methods: sEVs expressing tdTomato were generated, and their uptake into constituent liver cells was observed in vitro, as well as their sites of uptake and cells in the liver using a mouse model of liver cirrhosis. The efficiency of sEV collection using tangential flow filtration (TFF) and changes in the therapeutic effects of sEVs in a volume-dependent manner were examined.

Results: The sEVs of MSCs accumulated mostly in macrophages in damaged areas of the liver. In addition, the therapeutic effect of sEVs was not necessarily dose-dependent, and it reached a plateau when the dosage exceeded a certain level. Furthermore, although ultracentrifugation was commonly used to collect sEVs for research purposes, we verified that TFF could be used for efficient sEV collection and that their effectiveness is not reduced.

Conclusion: In this study, we identified some unknown aspects regarding the dynamics, collection, and capacity dependence of sEVs. Our results provide important fundamentals for the development of therapies using sEVs and hold potential implications for the therapeutic applications of sEV-based therapies for liver cirrhosis.

背景:肝纤维化的进展导致门静脉高压和肝功能障碍。然而,到目前为止,还没有抗纤维化药物被批准用于肝硬化,这使得它们成为一种未得到满足的医疗需求。间充质干细胞(MSC)的细胞外小泡(sEV)是这些候选药物之一。在本研究中,我们研究了MSCs的sEV的作用,分析了它们在给药后在肝脏中的分布,它们的作用是否具有剂量依赖性,以及是否有可能收集大量的sEV。方法:产生表达tdTomato的sEV,并在体外观察它们对组成肝细胞的摄取,以及它们的摄取部位和肝脏中的细胞。研究了使用切向流过滤(TFF)收集sEV的效率以及sEV治疗效果的体积依赖性变化。结果:间充质干细胞的sEV主要聚集在肝损伤区的巨噬细胞中。此外,sEV的治疗效果不一定是剂量依赖性的,当剂量超过一定水平时,它达到了平稳期。此外,尽管出于研究目的,超速离心通常用于收集sEV,但我们验证了TFF可以用于有效的sEV收集,并且其有效性没有降低。结论:在本研究中,我们确定了sEV的动力学、收集和容量依赖性方面的一些未知方面。我们的研究结果为开发使用sEV的治疗方法提供了重要的基础,并对基于sEV的肝硬化治疗应用具有潜在的意义。
{"title":"Analysis of distribution, collection, and confirmation of capacity dependency of small extracellular vesicles toward a therapy for liver cirrhosis.","authors":"Nobutaka Takeda, Atsunori Tsuchiya, Masaki Mito, Kazuki Natsui, Yui Natusi, Yohei Koseki, Kei Tomiyoshi, Fusako Yamazaki, Yuki Yoshida, Hiroyuki Abe, Masayuki Sano, Taketomo Kido, Yusuke Yoshioka, Junichi Kikuta, Tohru Itoh, Ken Nishimura, Masaru Ishii, Takahiro Ochiya, Atsushi Miyajima, Shuji Terai","doi":"10.1186/s41232-023-00299-x","DOIUrl":"10.1186/s41232-023-00299-x","url":null,"abstract":"<p><strong>Background: </strong>The progression of liver fibrosis leads to portal hypertension and liver dysfunction. However, no antifibrotic agents have been approved for cirrhosis to date, making them an unmet medical need. Small extracellular vesicles (sEVs) of mesenchymal stem cells (MSCs) are among these candidate agents. In this study, we investigated the effects of sEVs of MSCs, analyzed their distribution in the liver post-administration, whether their effect was dose-dependent, and whether it was possible to collect a large number of sEVs.</p><p><strong>Methods: </strong>sEVs expressing tdTomato were generated, and their uptake into constituent liver cells was observed in vitro, as well as their sites of uptake and cells in the liver using a mouse model of liver cirrhosis. The efficiency of sEV collection using tangential flow filtration (TFF) and changes in the therapeutic effects of sEVs in a volume-dependent manner were examined.</p><p><strong>Results: </strong>The sEVs of MSCs accumulated mostly in macrophages in damaged areas of the liver. In addition, the therapeutic effect of sEVs was not necessarily dose-dependent, and it reached a plateau when the dosage exceeded a certain level. Furthermore, although ultracentrifugation was commonly used to collect sEVs for research purposes, we verified that TFF could be used for efficient sEV collection and that their effectiveness is not reduced.</p><p><strong>Conclusion: </strong>In this study, we identified some unknown aspects regarding the dynamics, collection, and capacity dependence of sEVs. Our results provide important fundamentals for the development of therapies using sEVs and hold potential implications for the therapeutic applications of sEV-based therapies for liver cirrhosis.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"43 1","pages":"48"},"PeriodicalIF":0.0,"publicationDate":"2023-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10561446/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41184627","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Placenta mesenchymal stem cell-derived extracellular vesicles alleviate liver fibrosis by inactivating hepatic stellate cells through a miR-378c/SKP2 axis. 胎盘间充质干细胞衍生的细胞外小泡通过miR-378c/SKP2轴失活肝星状细胞来减轻肝纤维化。
Pub Date : 2023-10-05 DOI: 10.1186/s41232-023-00297-z
Wenjie Zheng, Saiyan Bian, Shi Qiu, Colin E Bishop, Meimei Wan, Nuo Xu, Xieyin Sun, Russel Clive Sequeira, Anthony Atala, Zhifeng Gu, Weixin Zhao

Background: Extracellular vesicles derived from mesenchymal stem/stromal cells (MSCs) have shown therapeutic effects on liver fibrosis. This study aimed to evaluate the effects of extracellular vesicles from placenta-derived MSCs (Pd-MSCs-EVs) on liver fibrosis at 3D/2D levels and explore the potential mechanisms.

Methods: The multicellular liver organoids, consisting of hepatocytes, hepatic stellate cells (HSCs), Kupffer cells, and liver sinusoidal endothelial cells, were observed for growth status, morphological changes, and metabolism. Human transformation growth factor- beta 1 (TGF-β1) was used to induce fibrosis at optimal concentration. The anti-fibrosis effects of Pd-MSCs-EVs were evaluated in liver organoids and HSCs models. Anti-fibrotic content of Pd-MSCs-EVs was identified by multiple experimental validations.

Results: TGF-β1 induced fibrosis in liver organoids, while Pd-MSCs-EVs significantly alleviated fibrotic phenotypes. Following serial verifications, miR-378c was identified as a potential key anti-fibrosis content. In contrast, miR-378c depletion decreased the anti-fibrotic effects of Pd-MSCs-EVs. Additionally, Pd-MSCs-EVs administration repressed TGF-β1-mediated HSCs activation at 2D or 3D levels. Mechanistically, exosomal miR-378c inactivated HSCs by inhibiting epithelial-mesenchymal transition (EMT) through stabilizing E-cadherin via targeting its E3 ubiquitin ligase S-Phase Kinase Associated Protein 2 (SKP2).

Conclusion: Pd-MSCs-EVs ameliorated TGF-β1-induced fibrosis by deactivating HSCs in a miR-378c/SKP2-dependent manner, which may be an efficient therapeutic candidate for liver fibrosis.

背景:来源于间充质干/基质细胞的细胞外小泡已显示出对肝纤维化的治疗作用。本研究旨在从3D/2D水平评估胎盘来源的间充质干细胞(Pd-MSCs-EVs)的细胞外小泡对肝纤维化的影响,并探讨其潜在机制。方法:观察由肝细胞、肝星状细胞、库普弗细胞和肝窦内皮细胞组成的多细胞肝类器官的生长状态、形态变化和代谢。人转化生长因子-β1(TGF-β1)在最佳浓度下诱导纤维化。在肝类器官和HSC模型中评估了Pd-MSCs-EVs的抗纤维化作用。通过多种实验验证鉴定了Pd-MSCs-EVs的抗纤维化含量。结果:TGF-β1诱导肝类器官纤维化,而Pd-MSCs-EVs显著减轻纤维化表型。经过一系列验证,miR-378c被鉴定为潜在的关键抗纤维化内容物。相反,miR-378c的缺失降低了Pd-MSCs-EVs的抗纤维化作用。此外,Pd-MSCs-EVs给药在2D或3D水平上抑制TGF-β1介导的HSC活化。从机制上讲,外泌体miR-378c通过靶向其E3泛素连接酶S相激酶相关蛋白2(SKP2)稳定E-钙粘蛋白来抑制上皮-间充质转化(EMT),从而使HSC失活,其可能是肝纤维化的有效治疗候选物。
{"title":"Placenta mesenchymal stem cell-derived extracellular vesicles alleviate liver fibrosis by inactivating hepatic stellate cells through a miR-378c/SKP2 axis.","authors":"Wenjie Zheng, Saiyan Bian, Shi Qiu, Colin E Bishop, Meimei Wan, Nuo Xu, Xieyin Sun, Russel Clive Sequeira, Anthony Atala, Zhifeng Gu, Weixin Zhao","doi":"10.1186/s41232-023-00297-z","DOIUrl":"10.1186/s41232-023-00297-z","url":null,"abstract":"<p><strong>Background: </strong>Extracellular vesicles derived from mesenchymal stem/stromal cells (MSCs) have shown therapeutic effects on liver fibrosis. This study aimed to evaluate the effects of extracellular vesicles from placenta-derived MSCs (Pd-MSCs-EVs) on liver fibrosis at 3D/2D levels and explore the potential mechanisms.</p><p><strong>Methods: </strong>The multicellular liver organoids, consisting of hepatocytes, hepatic stellate cells (HSCs), Kupffer cells, and liver sinusoidal endothelial cells, were observed for growth status, morphological changes, and metabolism. Human transformation growth factor- beta 1 (TGF-β1) was used to induce fibrosis at optimal concentration. The anti-fibrosis effects of Pd-MSCs-EVs were evaluated in liver organoids and HSCs models. Anti-fibrotic content of Pd-MSCs-EVs was identified by multiple experimental validations.</p><p><strong>Results: </strong>TGF-β1 induced fibrosis in liver organoids, while Pd-MSCs-EVs significantly alleviated fibrotic phenotypes. Following serial verifications, miR-378c was identified as a potential key anti-fibrosis content. In contrast, miR-378c depletion decreased the anti-fibrotic effects of Pd-MSCs-EVs. Additionally, Pd-MSCs-EVs administration repressed TGF-β1-mediated HSCs activation at 2D or 3D levels. Mechanistically, exosomal miR-378c inactivated HSCs by inhibiting epithelial-mesenchymal transition (EMT) through stabilizing E-cadherin via targeting its E3 ubiquitin ligase S-Phase Kinase Associated Protein 2 (SKP2).</p><p><strong>Conclusion: </strong>Pd-MSCs-EVs ameliorated TGF-β1-induced fibrosis by deactivating HSCs in a miR-378c/SKP2-dependent manner, which may be an efficient therapeutic candidate for liver fibrosis.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"43 1","pages":"47"},"PeriodicalIF":0.0,"publicationDate":"2023-10-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10557276/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41173303","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A niche-mimicking polymer hydrogel-based approach to identify molecular targets for tackling human pancreatic cancer stem cells. 一种基于镍亚胺聚合物水凝胶的方法,用于识别对付人类胰腺癌症干细胞的分子靶点。
Pub Date : 2023-09-27 DOI: 10.1186/s41232-023-00296-0
Yoshitaka Murota, Mariko Nagane, Mei Wu, Mithun Santra, Seshasailam Venkateswaran, Shinji Tanaka, Mark Bradley, Tetsuya Taga, Kouichi Tabu

Background: Pancreatic adenocarcinoma (PAAD) is one of the most fatal human cancers, but effective therapies remain to be established. Cancer stem cells (CSCs) are highly resistant to anti-cancer drugs and a deeper understanding of their microenvironmental niche has been considered important to provide understanding and solutions to cancer eradication. However, as the CSC niche is composed of a wide variety of biological and physicochemical factors, the development of multidisciplinary tools that recapitulate their complex features is indispensable. Synthetic polymers have been studied as attractive biomaterials due to their tunable biofunctionalities, while hydrogelation technique further renders upon them a diversity of physical properties, making them an attractive tool for analysis of the CSC niche.

Methods: To develop innovative materials that recapitulate the CSC niche in pancreatic cancers, we performed polymer microarray analysis to identify niche-mimicking scaffolds that preferentially supported the growth of CSCs. The niche-mimicking activity of the identified polymers was further optimized by polyethylene glycol (PEG)-based hydrogelation. To reveal the biological mechanisms behind the activity of the optimized hydrogels towards CSCs, proteins binding onto the hydrogel were analyzed by liquid chromatography with tandem mass spectrometry (LC-MS/MS), and the potential therapeutic targets were validated by looking at gene expression and patients' outcome in the TCGA database.

Results: PA531, a heteropolymer composed of 2-methoxyethyl methacrylate (MEMA) and 2-(diethylamino)ethyl methacrylate (DEAEMA) (5.5:4.5) that specifically supports the growth and maintenance of CSCs was identified by polymer microarray screening using the human PAAD cell line KLM1. The polymer PA531 was converted into five hydrogels (PA531-HG1 to HG5) and developed to give an optimized scaffold with the highest CSC niche-mimicking activities. From this polymer that recapitulated CSC binding and control, the proteins fetuin-B and angiotensinogen were identified as candidate target molecules with clinical significance due to the correlation between gene expression levels and prognosis in PAAD patients and the proteins associated with the niche-mimicking polymer.

Conclusion: This study screened for biofunctional polymers suitable for recapitulation of the pancreatic CSC niche and one hydrogel with high niche-mimicking abilities was successfully fabricated. Two soluble factors with clinical significance were identified as potential candidates for biomarkers and therapeutic targets in pancreatic cancers. Such a biomaterial-based approach could be a new platform in drug discovery and therapy development against CSCs, via targeting of their niche.

背景:胰腺癌(PAAD)是人类最致命的癌症之一,但有效的治疗方法仍有待建立。癌症干细胞(CSCs)对抗癌药物具有高度耐药性,对其微环境生态位的深入了解被认为对于提供对癌症根除的理解和解决方案至关重要。然而,由于CSC生态位由各种各样的生物和物理化学因素组成,开发概括其复杂特征的多学科工具是必不可少的。合成聚合物由于其可调的生物功能而被研究为有吸引力的生物材料,而水凝胶技术进一步赋予了它们多样的物理性质,使其成为分析CSC生态位的有吸引力的工具。方法:为了开发能够概括胰腺癌CSC小生境的创新材料,我们进行了聚合物微阵列分析,以确定优先支持CSC生长的小生境模拟支架。通过聚乙二醇(PEG)基水凝胶进一步优化了所鉴定聚合物的生态位模拟活性。为了揭示优化的水凝胶对CSCs活性背后的生物学机制,通过液相色谱-串联质谱法(LC-MS/MS)分析了与水凝胶结合的蛋白质,并通过查看TCGA数据库中的基因表达和患者结果来验证潜在的治疗靶点。结果:使用人PAAD细胞系KLM1,通过聚合物微阵列筛选,鉴定了一种由甲基丙烯酸2-甲氧基乙酯(MEMA)和甲基丙烯酸2-(二乙氨基)乙酯(DEAEMA)(5.5:4.5)组成的异聚物PA531,该异聚物特异性支持CSCs的生长和维持。将聚合物PA531转化为五种水凝胶(PA531-HG1至HG5),并开发出具有最高CSC小生境模拟活性的优化支架。从这种概括CSC结合和控制的聚合物中,由于PAAD患者的基因表达水平和预后之间的相关性,以及与模拟生态位的聚合物相关的蛋白质,蛋白胎蛋白-B和血管紧张素原被鉴定为具有临床意义的候选靶分子。结论:本研究筛选了适用于重现胰腺CSC生态位的生物功能聚合物,并成功制备了一种具有高生态位模拟能力的水凝胶。两种具有临床意义的可溶性因子被确定为胰腺癌生物标志物和治疗靶点的潜在候选者。这种基于生物材料的方法可以通过靶向CSC的小生境,成为针对CSC的药物发现和治疗开发的新平台。
{"title":"A niche-mimicking polymer hydrogel-based approach to identify molecular targets for tackling human pancreatic cancer stem cells.","authors":"Yoshitaka Murota, Mariko Nagane, Mei Wu, Mithun Santra, Seshasailam Venkateswaran, Shinji Tanaka, Mark Bradley, Tetsuya Taga, Kouichi Tabu","doi":"10.1186/s41232-023-00296-0","DOIUrl":"10.1186/s41232-023-00296-0","url":null,"abstract":"<p><strong>Background: </strong>Pancreatic adenocarcinoma (PAAD) is one of the most fatal human cancers, but effective therapies remain to be established. Cancer stem cells (CSCs) are highly resistant to anti-cancer drugs and a deeper understanding of their microenvironmental niche has been considered important to provide understanding and solutions to cancer eradication. However, as the CSC niche is composed of a wide variety of biological and physicochemical factors, the development of multidisciplinary tools that recapitulate their complex features is indispensable. Synthetic polymers have been studied as attractive biomaterials due to their tunable biofunctionalities, while hydrogelation technique further renders upon them a diversity of physical properties, making them an attractive tool for analysis of the CSC niche.</p><p><strong>Methods: </strong>To develop innovative materials that recapitulate the CSC niche in pancreatic cancers, we performed polymer microarray analysis to identify niche-mimicking scaffolds that preferentially supported the growth of CSCs. The niche-mimicking activity of the identified polymers was further optimized by polyethylene glycol (PEG)-based hydrogelation. To reveal the biological mechanisms behind the activity of the optimized hydrogels towards CSCs, proteins binding onto the hydrogel were analyzed by liquid chromatography with tandem mass spectrometry (LC-MS/MS), and the potential therapeutic targets were validated by looking at gene expression and patients' outcome in the TCGA database.</p><p><strong>Results: </strong>PA531, a heteropolymer composed of 2-methoxyethyl methacrylate (MEMA) and 2-(diethylamino)ethyl methacrylate (DEAEMA) (5.5:4.5) that specifically supports the growth and maintenance of CSCs was identified by polymer microarray screening using the human PAAD cell line KLM1. The polymer PA531 was converted into five hydrogels (PA531-HG1 to HG5) and developed to give an optimized scaffold with the highest CSC niche-mimicking activities. From this polymer that recapitulated CSC binding and control, the proteins fetuin-B and angiotensinogen were identified as candidate target molecules with clinical significance due to the correlation between gene expression levels and prognosis in PAAD patients and the proteins associated with the niche-mimicking polymer.</p><p><strong>Conclusion: </strong>This study screened for biofunctional polymers suitable for recapitulation of the pancreatic CSC niche and one hydrogel with high niche-mimicking abilities was successfully fabricated. Two soluble factors with clinical significance were identified as potential candidates for biomarkers and therapeutic targets in pancreatic cancers. Such a biomaterial-based approach could be a new platform in drug discovery and therapy development against CSCs, via targeting of their niche.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"43 1","pages":"46"},"PeriodicalIF":0.0,"publicationDate":"2023-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10523636/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41160308","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Divergent roles of the Hippo pathway in the pathogenesis of idiopathic pulmonary fibrosis: tissue homeostasis and fibrosis. Hippo通路在特发性肺纤维化发病机制中的不同作用:组织稳态和纤维化。
Pub Date : 2023-09-21 DOI: 10.1186/s41232-023-00295-1
Ryusuke Kizawa, Jun Araya, Yu Fujita

Idiopathic pulmonary fibrosis (IPF) is a progressive aging-related lung disease with a poor prognosis. Despite extensive research, the cause of IPF remains largely unknown and treatment strategies are limited. Proposed mechanisms of the pathogenesis of IPF are a combination of excessive accumulation of the extracellular matrix and dysfunctional lung tissue regeneration. Epithelial cell dysfunction, in addition to fibroblast activation, is considered a key process in the progression of IPF. Epithelial cells normally maintain homeostasis of the lung tissue through regulated proliferation, differentiation, cell death, and cellular senescence. However, various stresses can cause repetitive damage to lung epithelial cells, leading to dysfunctional regeneration and acquisition of profibrotic functions. The Hippo pathway is a central signaling pathway that maintains tissue homeostasis and plays an essential role in fundamental biological processes. Dysregulation of the Hippo pathway has been implicated in various diseases, including IPF. However, the role of the Hippo pathway in the pathogenesis of IPF remains unclear, particularly given the pathway's opposing effects on the 2 key pathogenic mechanisms of IPF: epithelial cell dysfunction and fibroblast activation. A deeper understanding of the relationship between the Hippo pathway and the pathogenesis of IPF will pave the way for novel Hippo-targeted therapies.

特发性肺纤维化(IPF)是一种进展性与衰老相关的肺部疾病,预后不良。尽管进行了广泛的研究,但IPF的病因在很大程度上仍然未知,治疗策略也有限。所提出的IPF发病机制是细胞外基质过度积累和肺组织再生功能障碍的结合。除了成纤维细胞活化外,上皮细胞功能障碍被认为是IPF进展的关键过程。上皮细胞通常通过调节增殖、分化、细胞死亡和细胞衰老来维持肺组织的稳态。然而,各种压力会对肺上皮细胞造成重复性损伤,导致再生和促纤维化功能获得功能障碍。Hippo通路是维持组织稳态的中枢信号通路,在基本生物过程中发挥着重要作用。Hippo通路的失调与包括IPF在内的各种疾病有关。然而,Hippo通路在IPF发病机制中的作用尚不清楚,特别是考虑到该通路对IPF的两个关键致病机制(上皮细胞功能障碍和成纤维细胞活化)的相反作用。更深入地了解Hippo通路与IPF发病机制之间的关系,将为新的Hippo靶向治疗铺平道路。
{"title":"Divergent roles of the Hippo pathway in the pathogenesis of idiopathic pulmonary fibrosis: tissue homeostasis and fibrosis.","authors":"Ryusuke Kizawa, Jun Araya, Yu Fujita","doi":"10.1186/s41232-023-00295-1","DOIUrl":"10.1186/s41232-023-00295-1","url":null,"abstract":"<p><p>Idiopathic pulmonary fibrosis (IPF) is a progressive aging-related lung disease with a poor prognosis. Despite extensive research, the cause of IPF remains largely unknown and treatment strategies are limited. Proposed mechanisms of the pathogenesis of IPF are a combination of excessive accumulation of the extracellular matrix and dysfunctional lung tissue regeneration. Epithelial cell dysfunction, in addition to fibroblast activation, is considered a key process in the progression of IPF. Epithelial cells normally maintain homeostasis of the lung tissue through regulated proliferation, differentiation, cell death, and cellular senescence. However, various stresses can cause repetitive damage to lung epithelial cells, leading to dysfunctional regeneration and acquisition of profibrotic functions. The Hippo pathway is a central signaling pathway that maintains tissue homeostasis and plays an essential role in fundamental biological processes. Dysregulation of the Hippo pathway has been implicated in various diseases, including IPF. However, the role of the Hippo pathway in the pathogenesis of IPF remains unclear, particularly given the pathway's opposing effects on the 2 key pathogenic mechanisms of IPF: epithelial cell dysfunction and fibroblast activation. A deeper understanding of the relationship between the Hippo pathway and the pathogenesis of IPF will pave the way for novel Hippo-targeted therapies.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"43 1","pages":"45"},"PeriodicalIF":0.0,"publicationDate":"2023-09-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10512581/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"41174573","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","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
Inflammation and regeneration
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