首页 > 最新文献

Inflammation and regeneration最新文献

英文 中文
Regulation of intestinal epithelial homeostasis by mesenchymal cells. 间充质细胞对肠上皮稳态的调节
Pub Date : 2024-09-26 DOI: 10.1186/s41232-024-00355-0
Hisako Kayama, Kiyoshi Takeda

The gastrointestinal tract harbors diverse microorganisms in the lumen. Epithelial cells segregate the luminal microorganisms from immune cells in the lamina propria by constructing chemical and physical barriers through the production of various factors to prevent excessive immune responses against microbes. Therefore, perturbations of epithelial integrity are linked to the development of gastrointestinal disorders. Several mesenchymal stromal cell populations, including fibroblasts, myofibroblasts, pericytes, and myocytes, contribute to the establishment and maintenance of epithelial homeostasis in the gut through regulation of the self-renewal, proliferation, and differentiation of intestinal stem cells. Recent studies have revealed alterations in the composition of intestinal mesenchymal stromal cells in patients with inflammatory bowel disease and colorectal cancer. A better understanding of the interplay between mesenchymal stromal cells and epithelial cells associated with intestinal health and diseases will facilitate identification of novel biomarkers and therapeutic targets for gastrointestinal disorders. This review summarizes the key findings obtained to date on the mechanisms by which functionally distinct mesenchymal stromal cells regulate epithelial integrity in intestinal health and diseases at different developmental stages.

胃肠道的管腔中蕴藏着多种微生物。上皮细胞通过产生各种因子构建化学和物理屏障,防止针对微生物的过度免疫反应,从而将管腔微生物与固有层中的免疫细胞隔离开来。因此,上皮完整性的紊乱与胃肠道疾病的发生有关。一些间充质基质细胞群,包括成纤维细胞、肌成纤维细胞、周细胞和肌细胞,通过调节肠道干细胞的自我更新、增殖和分化,有助于建立和维持肠道上皮细胞的稳态。最近的研究发现,炎症性肠病和结肠直肠癌患者肠间充质基质细胞的组成发生了改变。更好地了解与肠道健康和疾病相关的间充质基质细胞和上皮细胞之间的相互作用,将有助于确定胃肠道疾病的新型生物标记物和治疗靶点。本综述总结了迄今为止关于功能独特的间充质基质细胞在不同发育阶段调节肠道健康和疾病的上皮完整性的机制的主要发现。
{"title":"Regulation of intestinal epithelial homeostasis by mesenchymal cells.","authors":"Hisako Kayama, Kiyoshi Takeda","doi":"10.1186/s41232-024-00355-0","DOIUrl":"https://doi.org/10.1186/s41232-024-00355-0","url":null,"abstract":"<p><p>The gastrointestinal tract harbors diverse microorganisms in the lumen. Epithelial cells segregate the luminal microorganisms from immune cells in the lamina propria by constructing chemical and physical barriers through the production of various factors to prevent excessive immune responses against microbes. Therefore, perturbations of epithelial integrity are linked to the development of gastrointestinal disorders. Several mesenchymal stromal cell populations, including fibroblasts, myofibroblasts, pericytes, and myocytes, contribute to the establishment and maintenance of epithelial homeostasis in the gut through regulation of the self-renewal, proliferation, and differentiation of intestinal stem cells. Recent studies have revealed alterations in the composition of intestinal mesenchymal stromal cells in patients with inflammatory bowel disease and colorectal cancer. A better understanding of the interplay between mesenchymal stromal cells and epithelial cells associated with intestinal health and diseases will facilitate identification of novel biomarkers and therapeutic targets for gastrointestinal disorders. This review summarizes the key findings obtained to date on the mechanisms by which functionally distinct mesenchymal stromal cells regulate epithelial integrity in intestinal health and diseases at different developmental stages.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"42"},"PeriodicalIF":0.0,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11426228/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142335199","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
Proteomics of blood extracellular vesicles in inflammatory respiratory diseases for biomarker discovery and new insights into pathophysiology. 对呼吸系统炎症性疾病中的血液细胞外囊泡进行蛋白质组学研究,以发现生物标志物并深入了解病理生理学。
Pub Date : 2024-09-18 DOI: 10.1186/s41232-024-00351-4
Takahiro Kawasaki, Yoshito Takeda, Atsushi Kumanogoh

Background: Inflammatory respiratory diseases, such as interstitial lung disease (ILD), bronchial asthma (BA), chronic obstructive pulmonary disease (COPD), and respiratory infections, remain significant global health concerns owing to their chronic and severe nature. Emerging as a valuable resource, blood extracellular vesicles (EVs) offer insights into disease pathophysiology and biomarker discovery in these conditions.

Main body: This review explores the advancements in blood EV proteomics for inflammatory respiratory diseases, highlighting their potential as non-invasive diagnostic and prognostic tools. Blood EVs offer advantages over traditional serum or plasma samples. Proteomic analyses of blood EVs have revealed numerous biomarkers that can be used to stratify patients, predict disease progression, and identify candidate therapeutic targets. Blood EV proteomics has identified proteins associated with progressive fibrosis in ILD, offering new avenues of treatment. In BA, eosinophil-derived EVs harbor biomarkers crucial for managing eosinophilic inflammation. Research on COPD has also identified proteins that correlate with lung function. Moreover, EVs play a critical role in respiratory infections such as COVID-19, and disease-associated proteins are encapsulated. Thus, proteomic studies have identified key molecules involved in disease severity and immune responses, underscoring their role in monitoring and guiding therapy.

Conclusion: This review highlights the potential of blood EV proteomics as a non-invasive diagnostic and prognostic tool for inflammatory respiratory diseases, providing a promising avenue for improved patient management and therapeutic development.

背景:炎症性呼吸系统疾病,如间质性肺病(ILD)、支气管哮喘(BA)、慢性阻塞性肺病(COPD)和呼吸道感染,由于其慢性和严重的性质,仍然是全球关注的重大健康问题。血液细胞外囊泡(EVs)作为一种宝贵的资源,为这些疾病的病理生理学和生物标志物的发现提供了见解:这篇综述探讨了血液EV蛋白质组学在治疗呼吸系统炎症性疾病方面的进展,强调了EV作为非侵入性诊断和预后工具的潜力。与传统的血清或血浆样本相比,血液EV具有优势。血液 EV 蛋白组学分析揭示了许多生物标记物,可用于对患者进行分层、预测疾病进展和确定候选治疗靶点。血EV蛋白质组学发现了与ILD进行性纤维化相关的蛋白质,为治疗提供了新途径。在 BA 中,嗜酸性粒细胞衍生的 EV 隐藏着对控制嗜酸性粒细胞炎症至关重要的生物标记物。对慢性阻塞性肺病的研究也发现了与肺功能相关的蛋白质。此外,EVs 在呼吸道感染(如 COVID-19)中发挥着关键作用,疾病相关蛋白质被包裹其中。因此,蛋白质组学研究发现了涉及疾病严重程度和免疫反应的关键分子,强调了它们在监测和指导治疗中的作用:本综述强调了血液 EV 蛋白质组学作为炎症性呼吸系统疾病的非侵入性诊断和预后工具的潜力,为改善患者管理和治疗开发提供了一条前景广阔的途径。
{"title":"Proteomics of blood extracellular vesicles in inflammatory respiratory diseases for biomarker discovery and new insights into pathophysiology.","authors":"Takahiro Kawasaki, Yoshito Takeda, Atsushi Kumanogoh","doi":"10.1186/s41232-024-00351-4","DOIUrl":"https://doi.org/10.1186/s41232-024-00351-4","url":null,"abstract":"<p><strong>Background: </strong>Inflammatory respiratory diseases, such as interstitial lung disease (ILD), bronchial asthma (BA), chronic obstructive pulmonary disease (COPD), and respiratory infections, remain significant global health concerns owing to their chronic and severe nature. Emerging as a valuable resource, blood extracellular vesicles (EVs) offer insights into disease pathophysiology and biomarker discovery in these conditions.</p><p><strong>Main body: </strong>This review explores the advancements in blood EV proteomics for inflammatory respiratory diseases, highlighting their potential as non-invasive diagnostic and prognostic tools. Blood EVs offer advantages over traditional serum or plasma samples. Proteomic analyses of blood EVs have revealed numerous biomarkers that can be used to stratify patients, predict disease progression, and identify candidate therapeutic targets. Blood EV proteomics has identified proteins associated with progressive fibrosis in ILD, offering new avenues of treatment. In BA, eosinophil-derived EVs harbor biomarkers crucial for managing eosinophilic inflammation. Research on COPD has also identified proteins that correlate with lung function. Moreover, EVs play a critical role in respiratory infections such as COVID-19, and disease-associated proteins are encapsulated. Thus, proteomic studies have identified key molecules involved in disease severity and immune responses, underscoring their role in monitoring and guiding therapy.</p><p><strong>Conclusion: </strong>This review highlights the potential of blood EV proteomics as a non-invasive diagnostic and prognostic tool for inflammatory respiratory diseases, providing a promising avenue for improved patient management and therapeutic development.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"38"},"PeriodicalIF":0.0,"publicationDate":"2024-09-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11409490/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142304838","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
Optimization of transplantation methods using isolated mesenchymal stem/stromal cells: clinical trials of inflammatory bowel diseases as an example. 利用分离的间充质干细胞/基质细胞优化移植方法:以炎症性肠病的临床试验为例。
Pub Date : 2024-08-16 DOI: 10.1186/s41232-024-00350-5
Daisuke Hisamatsu, Akimi Ikeba, Taku Yamato, Yo Mabuchi, Mamoru Watanabe, Chihiro Akazawa

Mesenchymal stem/stromal cells (MSCs) are distributed in various tissues and are used in clinical applications as a source of transplanted cells because of their easy harvestability. Although MSCs express numerous cell-surface antigens, single-cell analyses have revealed a highly heterogeneous cell population depending on the original tissue and donor conditions, including age and interindividual differences. This heterogeneity leads to differences in their functions, such as multipotency and immunomodulatory effects, making it challenging to effectively treat targeted diseases. The therapeutic efficacy of MSCs is controversial and depends on the implantation site. Thus, there is no established recipe for the transplantation of MSCs (including the type of disease, type of origin, method of cell culture, form of transplanted cells, and site of delivery). Our recent preclinical study identified appropriate MSCs and their suitable transplantation routes in a mouse model of inflammatory bowel disease (IBD). Three-dimensional (3D) cultures of MSCs have been demonstrated to enhance their properties and sustain engraftment at the lesion site. In this note, we explore the methods of MSC transplantation for treating IBDs, especially Crohn's disease, from clinical trials published over the past decade. Given the functional changes in MSCs in 3D culture, we also investigate the clinical trials using 3D constructs of MSCs and explore suitable diseases that might benefit from this approach. Furthermore, we discuss the advantages of the prospective isolation of MSCs in terms of interindividual variability. This note highlights the need to define the method of MSC transplantation, including interindividual variability, the culture period, and the transplantation route.

间充质干/基质细胞(MSCs)分布于各种组织中,由于其易于采集,在临床应用中被用作移植细胞的来源。虽然间充质干细胞能表达多种细胞表面抗原,但单细胞分析表明,间充质干细胞群体具有高度异质性,这取决于原始组织和供体条件,包括年龄和个体差异。这种异质性导致其功能(如多潜能性和免疫调节作用)的差异,从而使有效治疗目标疾病具有挑战性。间充质干细胞的疗效存在争议,而且取决于植入部位。因此,间充质干细胞的移植(包括疾病类型、来源类型、细胞培养方法、移植细胞的形式和输送部位)并没有既定的配方。我们最近的临床前研究在炎症性肠病(IBD)小鼠模型中确定了合适的间充质干细胞及其合适的移植途径。间充质干细胞的三维(3D)培养已被证明能增强其特性并维持在病变部位的移植。在本文中,我们将从过去十年发表的临床试验中探讨间充质干细胞移植治疗 IBD(尤其是克罗恩病)的方法。鉴于间充质干细胞在三维培养过程中的功能变化,我们还调查了使用间充质干细胞三维构建体的临床试验,并探讨了可能从这种方法中获益的合适疾病。此外,我们还讨论了前瞻性分离间充质干细胞在个体差异方面的优势。本说明强调了确定间充质干细胞移植方法的必要性,包括个体间变异性、培养期和移植途径。
{"title":"Optimization of transplantation methods using isolated mesenchymal stem/stromal cells: clinical trials of inflammatory bowel diseases as an example.","authors":"Daisuke Hisamatsu, Akimi Ikeba, Taku Yamato, Yo Mabuchi, Mamoru Watanabe, Chihiro Akazawa","doi":"10.1186/s41232-024-00350-5","DOIUrl":"10.1186/s41232-024-00350-5","url":null,"abstract":"<p><p>Mesenchymal stem/stromal cells (MSCs) are distributed in various tissues and are used in clinical applications as a source of transplanted cells because of their easy harvestability. Although MSCs express numerous cell-surface antigens, single-cell analyses have revealed a highly heterogeneous cell population depending on the original tissue and donor conditions, including age and interindividual differences. This heterogeneity leads to differences in their functions, such as multipotency and immunomodulatory effects, making it challenging to effectively treat targeted diseases. The therapeutic efficacy of MSCs is controversial and depends on the implantation site. Thus, there is no established recipe for the transplantation of MSCs (including the type of disease, type of origin, method of cell culture, form of transplanted cells, and site of delivery). Our recent preclinical study identified appropriate MSCs and their suitable transplantation routes in a mouse model of inflammatory bowel disease (IBD). Three-dimensional (3D) cultures of MSCs have been demonstrated to enhance their properties and sustain engraftment at the lesion site. In this note, we explore the methods of MSC transplantation for treating IBDs, especially Crohn's disease, from clinical trials published over the past decade. Given the functional changes in MSCs in 3D culture, we also investigate the clinical trials using 3D constructs of MSCs and explore suitable diseases that might benefit from this approach. Furthermore, we discuss the advantages of the prospective isolation of MSCs in terms of interindividual variability. This note highlights the need to define the method of MSC transplantation, including interindividual variability, the culture period, and the transplantation route.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"37"},"PeriodicalIF":0.0,"publicationDate":"2024-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11328379/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141997100","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
Oncostatin M-driven macrophage-fibroblast circuits as a drug target in autoimmune arthritis. 以肿瘤坏死素 M 驱动的巨噬细胞-成纤维细胞环路作为自身免疫性关节炎的药物靶点
Pub Date : 2024-07-31 DOI: 10.1186/s41232-024-00347-0
Nam Cong-Nhat Huynh, Rui Ling, Masatsugu Komagamine, Tianshu Shi, Masayuki Tsukasaki, Kotaro Matsuda, Kazuo Okamoto, Tatsuo Asano, Ryunosuke Muro, Warunee Pluemsakunthai, George Kollias, Yuko Kaneko, Tsutomu Takeuchi, Sakae Tanaka, Noriko Komatsu, Hiroshi Takayanagi

Background: Recent single-cell RNA sequencing (scRNA-seq) analysis revealed the functional heterogeneity and pathogenic cell subsets in immune cells, synovial fibroblasts and bone cells in rheumatoid arthritis (RA). JAK inhibitors which ameliorate joint inflammation and bone destruction in RA, suppress the activation of various types of cells in vitro. However, the key cellular and molecular mechanisms underlying the potent clinical effects of JAK inhibitors on RA remain to be determined. Our aim is to identify a therapeutic target for JAK inhibitors in vivo.

Methods: We performed scRNA-seq analysis of the synovium of collagen-induced arthritis (CIA) mice treated with or without a JAK inhibitor, followed by a computational analysis to identify the drug target cells and signaling pathways. We utilized integrated human RA scRNA-seq datasets and genetically modified mice administered with the JAK inhibitor for the confirmation of our findings.

Results: scRNA-seq analysis revealed that oncostatin M (OSM) driven macrophage-fibroblast interaction is highly activated under arthritic conditions. OSM derived from macrophages, acts on OSM receptor (OSMR)-expressing synovial fibroblasts, activating both inflammatory and tissue-destructive subsets. Inflammatory synovial fibroblasts stimulate macrophages, mainly through IL-6, to exacerbate inflammation. Tissue-destructive synovial fibroblasts promote osteoclast differentiation by producing RANKL to accelerate bone destruction. scRNA-seq analysis also revealed that OSM-signaling in synovial fibroblasts is the main signaling pathway targeted by JAK inhibitors in vivo. Mice specifically lacking OSMR in synovial fibroblasts (Osmr∆Fibro) displayed ameliorated inflammation and joint destruction in arthritis. The JAK inhibitor was effective on the arthritis of the control mice while it had no effect on the arthritis of Osmr∆Fibro mice.

Conclusions: OSM functions as one of the key cytokines mediating pathogenic macrophage-fibroblast interaction. OSM-signaling in synovial fibroblasts is one of the main signaling pathways targeted by JAK inhibitors in vivo. The critical role of fibroblast-OSM signaling in autoimmune arthritis was shown by a combination of mice specifically deficient for OSMR in synovial fibroblasts and administration of the JAK inhibitor. Thus, the OSM-driven synovial macrophage-fibroblast circuit is proven to be a key driver of autoimmune arthritis, serving as a crucial drug target in vivo.

背景:最近的单细胞 RNA 测序(scRNA-seq)分析揭示了类风湿性关节炎(RA)中免疫细胞、滑膜成纤维细胞和骨细胞的功能异质性和致病细胞亚群。JAK 抑制剂可改善类风湿性关节炎的关节炎症和骨质破坏,抑制体外各类细胞的活化。然而,JAK抑制剂对类风湿关节炎产生强大临床效应的关键细胞和分子机制仍有待确定。我们的目标是确定 JAK 抑制剂在体内的治疗靶点:我们对接受或不接受 JAK 抑制剂治疗的胶原诱导性关节炎(CIA)小鼠滑膜进行了 scRNA-seq 分析,随后进行了计算分析,以确定药物靶细胞和信号通路。结果:scRNA-seq分析发现,在关节炎条件下,由oncostatin M(OSM)驱动的巨噬细胞-成纤维细胞相互作用被高度激活。来自巨噬细胞的OSM作用于表达OSM受体(OSMR)的滑膜成纤维细胞,同时激活炎性和组织破坏性亚群。炎症性滑膜成纤维细胞主要通过 IL-6 刺激巨噬细胞,从而加剧炎症。scRNA-seq分析还发现,滑膜成纤维细胞中的OSM信号是JAK抑制剂在体内靶向的主要信号通路。滑膜成纤维细胞中特异性缺乏 OSMR 的小鼠(Osmr∆Fibro)在关节炎中的炎症和关节破坏有所改善。JAK 抑制剂对对照组小鼠的关节炎有效,而对 Osmr∆Fibro 小鼠的关节炎没有影响:结论:OSM是介导致病性巨噬细胞-成纤维细胞相互作用的关键细胞因子之一。滑膜成纤维细胞中的 OSM 信号是 JAK 抑制剂在体内靶向的主要信号通路之一。滑膜成纤维细胞中OSMR特异性缺失的小鼠与JAK抑制剂的联合应用证明了成纤维细胞-OSM信号在自身免疫性关节炎中的关键作用。因此,OSM驱动的滑膜巨噬细胞-成纤维细胞回路被证明是自身免疫性关节炎的关键驱动因素,可作为体内的关键药物靶点。
{"title":"Oncostatin M-driven macrophage-fibroblast circuits as a drug target in autoimmune arthritis.","authors":"Nam Cong-Nhat Huynh, Rui Ling, Masatsugu Komagamine, Tianshu Shi, Masayuki Tsukasaki, Kotaro Matsuda, Kazuo Okamoto, Tatsuo Asano, Ryunosuke Muro, Warunee Pluemsakunthai, George Kollias, Yuko Kaneko, Tsutomu Takeuchi, Sakae Tanaka, Noriko Komatsu, Hiroshi Takayanagi","doi":"10.1186/s41232-024-00347-0","DOIUrl":"10.1186/s41232-024-00347-0","url":null,"abstract":"<p><strong>Background: </strong>Recent single-cell RNA sequencing (scRNA-seq) analysis revealed the functional heterogeneity and pathogenic cell subsets in immune cells, synovial fibroblasts and bone cells in rheumatoid arthritis (RA). JAK inhibitors which ameliorate joint inflammation and bone destruction in RA, suppress the activation of various types of cells in vitro. However, the key cellular and molecular mechanisms underlying the potent clinical effects of JAK inhibitors on RA remain to be determined. Our aim is to identify a therapeutic target for JAK inhibitors in vivo.</p><p><strong>Methods: </strong>We performed scRNA-seq analysis of the synovium of collagen-induced arthritis (CIA) mice treated with or without a JAK inhibitor, followed by a computational analysis to identify the drug target cells and signaling pathways. We utilized integrated human RA scRNA-seq datasets and genetically modified mice administered with the JAK inhibitor for the confirmation of our findings.</p><p><strong>Results: </strong>scRNA-seq analysis revealed that oncostatin M (OSM) driven macrophage-fibroblast interaction is highly activated under arthritic conditions. OSM derived from macrophages, acts on OSM receptor (OSMR)-expressing synovial fibroblasts, activating both inflammatory and tissue-destructive subsets. Inflammatory synovial fibroblasts stimulate macrophages, mainly through IL-6, to exacerbate inflammation. Tissue-destructive synovial fibroblasts promote osteoclast differentiation by producing RANKL to accelerate bone destruction. scRNA-seq analysis also revealed that OSM-signaling in synovial fibroblasts is the main signaling pathway targeted by JAK inhibitors in vivo. Mice specifically lacking OSMR in synovial fibroblasts (Osmr<sup>∆Fibro</sup>) displayed ameliorated inflammation and joint destruction in arthritis. The JAK inhibitor was effective on the arthritis of the control mice while it had no effect on the arthritis of Osmr<sup>∆Fibro</sup> mice.</p><p><strong>Conclusions: </strong>OSM functions as one of the key cytokines mediating pathogenic macrophage-fibroblast interaction. OSM-signaling in synovial fibroblasts is one of the main signaling pathways targeted by JAK inhibitors in vivo. The critical role of fibroblast-OSM signaling in autoimmune arthritis was shown by a combination of mice specifically deficient for OSMR in synovial fibroblasts and administration of the JAK inhibitor. Thus, the OSM-driven synovial macrophage-fibroblast circuit is proven to be a key driver of autoimmune arthritis, serving as a crucial drug target in vivo.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"36"},"PeriodicalIF":0.0,"publicationDate":"2024-07-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11289929/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141857455","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
Adipose-derived mesenchymal stem cell therapy for connective tissue diseases and complications. 脂肪间充质干细胞治疗结缔组织疾病和并发症。
Pub Date : 2024-07-19 DOI: 10.1186/s41232-024-00348-z
Takuya Kotani, Takashi Saito, Takayasu Suzuka, Shogo Matsuda

Mesenchymal stem cells (MSCs) may be effective in treating connective tissue disease and associated organ damage, leveraging their anti-inflammatory and immunoregulatory effects. Moreover, MSCs may possess the ability to produce antiapoptotic, proliferative, growth, angiogenic, and antifibrotic factors. Among MSCs, adipose-derived MSCs (ASCs) stand out for their relative ease of harvesting and abundance. Additionally, studies have indicated that compared with bone marrow-derived MSCs, ASCs have superior immunomodulatory, proangiogenic, antiapoptotic, and antioxidative properties. However, relatively few reviews have focused on the efficacy of ASC therapy in treating connective tissue disease (CTD) and interstitial lung disease (ILD). Therefore, this review aims to evaluate evidence from preclinical studies that investigate the effectiveness of MSC therapy, specifically ASC therapy, in managing CTD and ILD. Moreover, we explore the outcomes of documented clinical trials. We also introduce an innovative approach involving the utilization of pharmacologically primed ASCs in the CTD model to address the current challenges associated with ASC therapy.

间充质干细胞(MSCs)具有抗炎和免疫调节作用,可有效治疗结缔组织疾病和相关器官损伤。此外,间充质干细胞可能具有产生抗凋亡、增殖、生长、血管生成和抗纤维化因子的能力。在间充质干细胞中,脂肪来源的间充质干细胞(ASCs)因其相对容易采集和数量丰富而脱颖而出。此外,有研究表明,与骨髓间充质干细胞相比,脂肪间充质干细胞具有更好的免疫调节、促血管生成、抗细胞凋亡和抗氧化特性。然而,有关间充质干细胞治疗结缔组织病(CTD)和间质性肺病(ILD)疗效的综述相对较少。因此,本综述旨在评估来自临床前研究的证据,这些研究调查了间充质干细胞疗法(尤其是 ASC疗法)在治疗结缔组织病和间质性肺病方面的有效性。此外,我们还探讨了有据可查的临床试验结果。我们还介绍了一种创新方法,即在 CTD 模型中利用药理诱导的间充质干细胞来应对目前与间充质干细胞疗法相关的挑战。
{"title":"Adipose-derived mesenchymal stem cell therapy for connective tissue diseases and complications.","authors":"Takuya Kotani, Takashi Saito, Takayasu Suzuka, Shogo Matsuda","doi":"10.1186/s41232-024-00348-z","DOIUrl":"10.1186/s41232-024-00348-z","url":null,"abstract":"<p><p>Mesenchymal stem cells (MSCs) may be effective in treating connective tissue disease and associated organ damage, leveraging their anti-inflammatory and immunoregulatory effects. Moreover, MSCs may possess the ability to produce antiapoptotic, proliferative, growth, angiogenic, and antifibrotic factors. Among MSCs, adipose-derived MSCs (ASCs) stand out for their relative ease of harvesting and abundance. Additionally, studies have indicated that compared with bone marrow-derived MSCs, ASCs have superior immunomodulatory, proangiogenic, antiapoptotic, and antioxidative properties. However, relatively few reviews have focused on the efficacy of ASC therapy in treating connective tissue disease (CTD) and interstitial lung disease (ILD). Therefore, this review aims to evaluate evidence from preclinical studies that investigate the effectiveness of MSC therapy, specifically ASC therapy, in managing CTD and ILD. Moreover, we explore the outcomes of documented clinical trials. We also introduce an innovative approach involving the utilization of pharmacologically primed ASCs in the CTD model to address the current challenges associated with ASC therapy.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"35"},"PeriodicalIF":0.0,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264739/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141725402","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
Pathological mechanisms and novel drug targets in fibrotic interstitial lung disease. 纤维化间质性肺病的病理机制和新型药物靶点。
Pub Date : 2024-07-19 DOI: 10.1186/s41232-024-00345-2
Yasuhiko Nishioka, Jun Araya, Yoshiya Tanaka, Atsushi Kumanogoh

Background: Interstitial lung diseases (ILDs) are a diverse group of conditions characterized by inflammation and fibrosis in the lung. In some patients with ILD, a progressive fibrotic phenotype develops, which is associated with an irreversible decline in lung function and a poor prognosis.

Main body: The pathological mechanisms that underlie this process culminate in fibroblast activation, proliferation, and differentiation into myofibroblasts, which deposit extracellular matrix proteins and result in fibrosis. Upstream of fibroblast activation, epithelial cell injury and immune activation are known initiators of fibrosis progression, with multiple diverse cell types involved. Recent years have seen an increase in our understanding of the complex and interrelated processes that drive fibrosis progression in ILD, in part due to the advent of single-cell RNA sequencing technology and integrative multiomics analyses. Novel pathological mechanisms have been identified, which represent new targets for drugs currently in clinical development. These include phosphodiesterase 4 inhibitors and other molecules that act on intracellular cyclic adenosine monophosphate signaling, as well as inhibitors of the autotaxin-lysophosphatidic acid axis and  α v  integrins. Here, we review current knowledge and recent developments regarding the pathological mechanisms that underlie progressive fibrotic ILD, including potential therapeutic targets.

Conclusion: Knowledge of the pathological mechanisms that drive progressive fibrosis in patients with ILD has expanded, with the role of alveolar endothelial cells, the immune system, and fibroblasts better elucidated. Drugs that target novel mechanisms hold promise for expanding the future therapeutic armamentarium for progressive fibrotic ILD.

背景:间质性肺疾病(ILDs)是一组以肺部炎症和纤维化为特征的多种疾病。一些 ILD 患者会出现进行性纤维化表型,这与肺功能不可逆转的下降和不良预后有关:这一过程的病理机制最终导致成纤维细胞活化、增殖并分化为肌成纤维细胞,从而沉积细胞外基质蛋白并导致纤维化。在成纤维细胞活化的上游,上皮细胞损伤和免疫激活是纤维化进展的已知启动因子,涉及多种不同类型的细胞。近年来,我们对推动 ILD 纤维化进展的复杂而相互关联的过程有了更多的了解,这部分归功于单细胞 RNA 测序技术和综合多组学分析的出现。新的病理机制已被确定,成为目前临床开发药物的新靶点。这些药物包括磷酸二酯酶 4 抑制剂和其他作用于细胞内环磷酸腺苷信号转导的分子,以及自旋素-来磷脂酸轴和α v整合素的抑制剂。在此,我们回顾了有关进展性纤维化性 ILD 的病理机制的现有知识和最新进展,包括潜在的治疗靶点:结论:随着肺泡内皮细胞、免疫系统和成纤维细胞的作用得到进一步阐明,人们对导致 ILD 患者进行性纤维化的病理机制的认识也在不断扩大。针对新机制的药物有望扩大进展性纤维化 ILD 的未来治疗范围。
{"title":"Pathological mechanisms and novel drug targets in fibrotic interstitial lung disease.","authors":"Yasuhiko Nishioka, Jun Araya, Yoshiya Tanaka, Atsushi Kumanogoh","doi":"10.1186/s41232-024-00345-2","DOIUrl":"10.1186/s41232-024-00345-2","url":null,"abstract":"<p><strong>Background: </strong>Interstitial lung diseases (ILDs) are a diverse group of conditions characterized by inflammation and fibrosis in the lung. In some patients with ILD, a progressive fibrotic phenotype develops, which is associated with an irreversible decline in lung function and a poor prognosis.</p><p><strong>Main body: </strong>The pathological mechanisms that underlie this process culminate in fibroblast activation, proliferation, and differentiation into myofibroblasts, which deposit extracellular matrix proteins and result in fibrosis. Upstream of fibroblast activation, epithelial cell injury and immune activation are known initiators of fibrosis progression, with multiple diverse cell types involved. Recent years have seen an increase in our understanding of the complex and interrelated processes that drive fibrosis progression in ILD, in part due to the advent of single-cell RNA sequencing technology and integrative multiomics analyses. Novel pathological mechanisms have been identified, which represent new targets for drugs currently in clinical development. These include phosphodiesterase 4 inhibitors and other molecules that act on intracellular cyclic adenosine monophosphate signaling, as well as inhibitors of the autotaxin-lysophosphatidic acid axis and  <math><msub><mi>α</mi> <mi>v</mi></msub> </math>  integrins. Here, we review current knowledge and recent developments regarding the pathological mechanisms that underlie progressive fibrotic ILD, including potential therapeutic targets.</p><p><strong>Conclusion: </strong>Knowledge of the pathological mechanisms that drive progressive fibrosis in patients with ILD has expanded, with the role of alveolar endothelial cells, the immune system, and fibroblasts better elucidated. Drugs that target novel mechanisms hold promise for expanding the future therapeutic armamentarium for progressive fibrotic ILD.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"34"},"PeriodicalIF":0.0,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264521/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141725403","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
In vivo neural regeneration via AAV-NeuroD1 gene delivery to astrocytes in neonatal hypoxic-ischemic brain injury. 在新生儿缺氧缺血性脑损伤中通过 AAV-NeuroD1 基因传递到星形胶质细胞实现体内神经再生。
Pub Date : 2024-07-16 DOI: 10.1186/s41232-024-00349-y
Miri Kim, Seokmin Oh, Songyeon Kim, Il-Sun Kim, Joowon Kim, Jungho Han, Ji Woong Ahn, Seungsoo Chung, Jae-Hyung Jang, Jeong Eun Shin, Kook In Park

Background: Neonatal hypoxic-ischemic brain injury (HIBI) is a significant contributor to neonatal mortality and long-term neurodevelopmental disability, characterized by massive neuronal loss and reactive astrogliosis. Current therapeutic approaches for neonatal HIBI have been limited to general supportive therapy because of the lack of methods to compensate for irreversible neuronal loss. This study aimed to establish a feasible regenerative therapy for neonatal HIBI utilizing in vivo direct neuronal reprogramming technology.

Methods: Neonatal HIBI was induced in ICR mice at postnatal day 7 by permanent right common carotid artery occlusion and exposure to hypoxia with 8% oxygen and 92% nitrogen for 90 min. Three days after the injury, NeuroD1 was delivered to reactive astrocytes of the injury site using the astrocyte-tropic adeno-associated viral (AAV) vector AAVShH19. AAVShH19 was engineered with the Cre-FLEX system for long-term tracking of infected cells.

Results: AAVShH19-mediated ectopic NeuroD1 expression effectively converted astrocytes into GABAergic neurons, and the converted cells exhibited electrophysiological properties and synaptic transmitters. Additionally, we found that NeuroD1-mediated in vivo direct neuronal reprogramming protected injured host neurons and altered the host environment, i.e., decreased the numbers of activated microglia, reactive astrocytes, and toxic A1-type astrocytes, and decreased the expression of pro-inflammatory factors. Furthermore, NeuroD1-treated mice exhibited significantly improved motor functions.

Conclusions: This study demonstrates that NeuroD1-mediated in vivo direct neuronal reprogramming technology through AAV gene delivery can be a novel regenerative therapy for neonatal HIBI.

背景:新生儿缺氧缺血性脑损伤(HIBI)是导致新生儿死亡和长期神经发育障碍的重要因素,其特点是神经元大量缺失和反应性星形胶质细胞增多。目前对新生儿缺氧性脑损伤的治疗方法仅限于一般的支持疗法,因为缺乏对不可逆的神经元损失进行补偿的方法。本研究旨在利用体内直接神经元重编程技术为新生儿HIBI建立一种可行的再生疗法:方法:在出生后第7天,通过永久性右颈总动脉闭塞和暴露于8%氧气和92%氮气的低氧环境中90分钟,诱导ICR小鼠新生儿HIBI。损伤三天后,使用星形胶质细胞趋向性腺相关病毒(AAV)载体 AAVShH19 将 NeuroD1 运送到损伤部位的反应性星形胶质细胞中。AAVShH19采用了Cre-FLEX系统,用于长期追踪受感染的细胞:结果:AAVShH19介导的异位NeuroD1表达有效地将星形胶质细胞转化为GABA能神经元,转化后的细胞表现出电生理特性和突触递质。此外,我们还发现,NeuroD1 介导的体内神经元直接重编程保护了受伤的宿主神经元,并改变了宿主环境,即减少了活化小胶质细胞、反应性星形胶质细胞和毒性 A1 型星形胶质细胞的数量,并降低了促炎因子的表达。此外,经 NeuroD1 处理的小鼠的运动功能也有明显改善:本研究表明,通过 AAV 基因递送,NeuroD1 介导的体内神经元直接重编程技术可以成为新生儿 HIBI 的一种新型再生疗法。
{"title":"In vivo neural regeneration via AAV-NeuroD1 gene delivery to astrocytes in neonatal hypoxic-ischemic brain injury.","authors":"Miri Kim, Seokmin Oh, Songyeon Kim, Il-Sun Kim, Joowon Kim, Jungho Han, Ji Woong Ahn, Seungsoo Chung, Jae-Hyung Jang, Jeong Eun Shin, Kook In Park","doi":"10.1186/s41232-024-00349-y","DOIUrl":"10.1186/s41232-024-00349-y","url":null,"abstract":"<p><strong>Background: </strong>Neonatal hypoxic-ischemic brain injury (HIBI) is a significant contributor to neonatal mortality and long-term neurodevelopmental disability, characterized by massive neuronal loss and reactive astrogliosis. Current therapeutic approaches for neonatal HIBI have been limited to general supportive therapy because of the lack of methods to compensate for irreversible neuronal loss. This study aimed to establish a feasible regenerative therapy for neonatal HIBI utilizing in vivo direct neuronal reprogramming technology.</p><p><strong>Methods: </strong>Neonatal HIBI was induced in ICR mice at postnatal day 7 by permanent right common carotid artery occlusion and exposure to hypoxia with 8% oxygen and 92% nitrogen for 90 min. Three days after the injury, NeuroD1 was delivered to reactive astrocytes of the injury site using the astrocyte-tropic adeno-associated viral (AAV) vector AAVShH19. AAVShH19 was engineered with the Cre-FLEX system for long-term tracking of infected cells.</p><p><strong>Results: </strong>AAVShH19-mediated ectopic NeuroD1 expression effectively converted astrocytes into GABAergic neurons, and the converted cells exhibited electrophysiological properties and synaptic transmitters. Additionally, we found that NeuroD1-mediated in vivo direct neuronal reprogramming protected injured host neurons and altered the host environment, i.e., decreased the numbers of activated microglia, reactive astrocytes, and toxic A1-type astrocytes, and decreased the expression of pro-inflammatory factors. Furthermore, NeuroD1-treated mice exhibited significantly improved motor functions.</p><p><strong>Conclusions: </strong>This study demonstrates that NeuroD1-mediated in vivo direct neuronal reprogramming technology through AAV gene delivery can be a novel regenerative therapy for neonatal HIBI.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"33"},"PeriodicalIF":0.0,"publicationDate":"2024-07-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11253351/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141629546","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
Proteomic insights into extracellular vesicles in ALS for therapeutic potential of Ropinirole and biomarker discovery. 通过蛋白质组学深入了解 ALS 细胞外囊泡,发现罗匹尼罗的治疗潜力和生物标记物。
Pub Date : 2024-07-12 DOI: 10.1186/s41232-024-00346-1
Chris Kato, Koji Ueda, Satoru Morimoto, Shinichi Takahashi, Shiho Nakamura, Fumiko Ozawa, Daisuke Ito, Yugaku Daté, Kensuke Okada, Naoki Kobayashi, Jin Nakahara, Hideyuki Okano

Background: Extracellular vesicles (EVs) hold the potential for elucidating the pathogenesis of amyotrophic lateral sclerosis (ALS) and serve as biomarkers. Notably, the comparative and longitudinal alterations in the protein profiles of EVs in serum (sEVs) and cerebrospinal fluid (CSF; cEVs) of sporadic ALS (SALS) patients remain uncharted. Ropinirole hydrochloride (ROPI; dopamine D2 receptor [D2R] agonist), a new anti-ALS drug candidate identified through induced pluripotent stem cell (iPSC)-based drug discovery, has been suggested to inhibit ALS disease progression in the Ropinirole Hydrochloride Remedy for Amyotrophic Lateral Sclerosis (ROPALS) trial, but its mechanism of action is not well understood. Therefore, we tried to reveal longitudinal changes with disease progression and the effects of ROPI on protein profiles of EVs.

Methods: We collected serum and CSF at fixed intervals from ten controls and from 20 SALS patients participating in the ROPALS trial. Comprehensive proteomic analysis of EVs, extracted from these samples, was conducted using liquid chromatography/mass spectrometer (LC/MS). Furthermore, we generated iPSC-derived astrocytes (iPasts) and performed RNA sequencing on astrocytes with or without ROPI treatment.

Results: The findings revealed notable disparities yet high congruity in sEVs and cEVs protein profiles concerning disease status, time and ROPI administration. In SALS, both sEVs and cEVs presented elevated levels of inflammation-related proteins but reduced levels associated with unfolded protein response (UPR). These results mirrored the longitudinal changes after disease onset and correlated with the revised ALS Functional Rating Scale (ALSFRS-R) at sampling time, suggesting a link to the onset and progression of SALS. ROPI appeared to counteract these changes, attenuating inflammation-related protein levels and boosting those tied to UPR in SALS, proposing an anti-ALS impact on EV protein profiles. Reverse translational research using iPasts indicated that these changes may partly reflect the DRD2-dependent neuroinflammatory inhibitory effects of ROPI. We have also identified biomarkers that predict diagnosis and disease progression by machine learning-driven biomarker search.

Conclusions: Despite the limited sample size, this study pioneers in reporting time-series proteomic alterations in serum and CSF EVs from SALS patients, offering comprehensive insights into SALS pathogenesis, ROPI-induced changes, and potential prognostic and diagnostic biomarkers.

背景:细胞外囊泡(EVs)具有阐明肌萎缩性脊髓侧索硬化症(ALS)发病机制和作为生物标记物的潜力。值得注意的是,散发性肌萎缩侧索硬化症(SALS)患者血清(sEVs)和脑脊液(CSF;cEVs)中的EVs蛋白谱的比较和纵向变化仍是未知数。盐酸罗匹尼罗(ROPI;多巴胺D2受体[D2R]激动剂)是通过基于诱导多能干细胞(iPSC)的药物研发发现的一种新型抗ALS候选药物,在盐酸罗匹尼罗治疗肌萎缩性脊髓侧索硬化症(ROPALS)试验中被认为可抑制ALS疾病进展,但其作用机制尚不十分清楚。因此,我们试图揭示疾病进展的纵向变化以及 ROPI 对 EVs 蛋白特征的影响:方法:我们以固定的时间间隔收集了 10 名对照组和 20 名参与 ROPALS 试验的 SALS 患者的血清和脑脊液。我们使用液相色谱/质谱仪(LC/MS)对从这些样本中提取的 EVs 进行了全面的蛋白质组学分析。此外,我们还生成了 iPSC 衍生星形胶质细胞(iPasts),并对接受或未接受 ROPI 治疗的星形胶质细胞进行了 RNA 测序:结果:研究结果表明,sEVs 和 cEVs 蛋白特征在疾病状态、时间和 ROPI 施用方面存在显著差异,但又高度一致。在 SALS 中,sEVs 和 cEVs 的炎症相关蛋白水平均升高,但与未折叠蛋白反应(UPR)相关的蛋白水平降低。这些结果反映了发病后的纵向变化,并与取样时的修订 ALS 功能评分量表(ALSFRS-R)相关,表明这与 SALS 的发病和进展有关。ROPI似乎能抵消这些变化,降低炎症相关蛋白水平,提高与SALS中UPR相关的蛋白水平,从而对EV蛋白谱产生抗ALS影响。使用iPasts进行的反向转化研究表明,这些变化可能部分反映了ROPI的DRD2依赖性神经炎症抑制作用。我们还通过机器学习驱动的生物标记物搜索,确定了可预测诊断和疾病进展的生物标记物:尽管样本量有限,但这项研究开创性地报告了SALS患者血清和脑脊液EVs的时间序列蛋白质组学变化,为SALS发病机制、ROPI诱导的变化以及潜在的预后和诊断生物标志物提供了全面的见解。
{"title":"Proteomic insights into extracellular vesicles in ALS for therapeutic potential of Ropinirole and biomarker discovery.","authors":"Chris Kato, Koji Ueda, Satoru Morimoto, Shinichi Takahashi, Shiho Nakamura, Fumiko Ozawa, Daisuke Ito, Yugaku Daté, Kensuke Okada, Naoki Kobayashi, Jin Nakahara, Hideyuki Okano","doi":"10.1186/s41232-024-00346-1","DOIUrl":"10.1186/s41232-024-00346-1","url":null,"abstract":"<p><strong>Background: </strong>Extracellular vesicles (EVs) hold the potential for elucidating the pathogenesis of amyotrophic lateral sclerosis (ALS) and serve as biomarkers. Notably, the comparative and longitudinal alterations in the protein profiles of EVs in serum (sEVs) and cerebrospinal fluid (CSF; cEVs) of sporadic ALS (SALS) patients remain uncharted. Ropinirole hydrochloride (ROPI; dopamine D2 receptor [D2R] agonist), a new anti-ALS drug candidate identified through induced pluripotent stem cell (iPSC)-based drug discovery, has been suggested to inhibit ALS disease progression in the Ropinirole Hydrochloride Remedy for Amyotrophic Lateral Sclerosis (ROPALS) trial, but its mechanism of action is not well understood. Therefore, we tried to reveal longitudinal changes with disease progression and the effects of ROPI on protein profiles of EVs.</p><p><strong>Methods: </strong>We collected serum and CSF at fixed intervals from ten controls and from 20 SALS patients participating in the ROPALS trial. Comprehensive proteomic analysis of EVs, extracted from these samples, was conducted using liquid chromatography/mass spectrometer (LC/MS). Furthermore, we generated iPSC-derived astrocytes (iPasts) and performed RNA sequencing on astrocytes with or without ROPI treatment.</p><p><strong>Results: </strong>The findings revealed notable disparities yet high congruity in sEVs and cEVs protein profiles concerning disease status, time and ROPI administration. In SALS, both sEVs and cEVs presented elevated levels of inflammation-related proteins but reduced levels associated with unfolded protein response (UPR). These results mirrored the longitudinal changes after disease onset and correlated with the revised ALS Functional Rating Scale (ALSFRS-R) at sampling time, suggesting a link to the onset and progression of SALS. ROPI appeared to counteract these changes, attenuating inflammation-related protein levels and boosting those tied to UPR in SALS, proposing an anti-ALS impact on EV protein profiles. Reverse translational research using iPasts indicated that these changes may partly reflect the DRD2-dependent neuroinflammatory inhibitory effects of ROPI. We have also identified biomarkers that predict diagnosis and disease progression by machine learning-driven biomarker search.</p><p><strong>Conclusions: </strong>Despite the limited sample size, this study pioneers in reporting time-series proteomic alterations in serum and CSF EVs from SALS patients, offering comprehensive insights into SALS pathogenesis, ROPI-induced changes, and potential prognostic and diagnostic biomarkers.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"32"},"PeriodicalIF":0.0,"publicationDate":"2024-07-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11241965/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141602356","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
Autocrine insulin-like growth factor 2 signaling as a potential target in the associated development of pulmonary emphysema and cancer in smokers. 胰岛素样生长因子 2 的自分泌信号是吸烟者肺气肿和癌症相关发展的潜在靶点。
Pub Date : 2024-06-21 DOI: 10.1186/s41232-024-00344-3
Hye-Jin Boo, Hye-Young Min, Heung-Bin Lim, Euni Lee, Ho-Young Lee

Background: Tobacco smoking causes pulmonary inflammation, resulting in emphysema, an independent risk factor for lung cancer. Induction of insulin-like growth factor 2 (IGF2) in response to lung injury by tobacco carcinogens, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol and polycyclic aromatic hydrocarbon benzo[a]pyrene in combination (NB), is critical for the proliferation of alveolar type 2 cells (AT2s) for lung repair. However, persistent IGF2 overexpression during NB-induced severe injury results in hyperproliferation of AT2s without coordinated AT2-to-AT1 differentiation, disrupting alveolar repair, which leads to the concurrent development of emphysema and lung cancer. The current study aims to verify the role of IGF2 signaling in the associated development of emphysema and cancer and develop effective pharmaceuticals for the diseases using animal models that recapitulate the characteristics of these chronic diseases.

Methods: The pathogenesis of pulmonary emphysema and cancer was analyzed by lung function testing, histological evaluation, in situ zymography, dihydroethidium staining, and immunofluorescence and immunohistochemistry analyses utilizing mouse models of emphysema and cancer established by moderate exposure to NB for up to seven months.

Results: Moderate NB exposure induced IGF2 expression in AT2s during the development of pulmonary emphysema and lung cancer in mice. Using AT2-specific insulin receptor knockout mice, we verified the causative role of sustained IGF2 signaling activation in AT2s in emphysema development. IGF2-targeting strategies, including voltage-dependent calcium channel blocker (CCB) and a neutralizing antibody, significantly suppressed the NB-induced development of emphysema and lung cancer. A publicly available database revealed an inverse correlation between the use of calcium channel blockers and a COPD diagnosis.

Conclusions: Our work confirms sustained IGF2 signaling activation in AT2s couples impaired lung repair to the concurrent development of emphysema and cancer in mice. Additionally, CCB and IGF2-specific neutralizing antibodies are effective pharmaceuticals for the two diseases.

背景:吸烟会引起肺部炎症,导致肺气肿,而肺气肿是肺癌的一个独立危险因素。烟草致癌物质 4-(甲基亚硝基氨基)-1-(3-吡啶基)-1-丁醇和多环芳香烃苯并[a]芘(NB)共同诱导胰岛素样生长因子 2(IGF2)以应对肺损伤,这对于肺泡 2 型细胞(AT2s)的增殖和肺修复至关重要。然而,在 NB 诱导的严重损伤过程中,IGF2 的持续过表达会导致 AT2 细胞过度增殖,而 AT2 向 AT1 的分化却不协调,从而破坏肺泡修复,导致肺气肿和肺癌的同时发生。本研究旨在验证 IGF2 信号在肺气肿和癌症相关发展中的作用,并利用再现这些慢性疾病特征的动物模型开发治疗这些疾病的有效药物:方法:通过肺功能测试、组织学评估、原位酶谱分析、二氢乙啶染色以及免疫荧光和免疫组化分析,利用中度暴露于NB长达7个月的肺气肿和癌症小鼠模型分析肺气肿和癌症的发病机制:结果:在小鼠肺气肿和肺癌的发展过程中,适度暴露于 NB 会诱导 AT2 中 IGF2 的表达。通过使用 AT2 特异性胰岛素受体基因敲除小鼠,我们验证了 AT2 中持续的 IGF2 信号激活在肺气肿发生中的致病作用。IGF2靶向策略,包括电压依赖性钙通道阻滞剂(CCB)和中和抗体,显著抑制了NB诱导的肺气肿和肺癌的发展。公开数据库显示,钙通道阻滞剂的使用与慢性阻塞性肺病的诊断之间存在反相关性:我们的研究证实,AT2s 中 IGF2 信号的持续激活将肺修复功能受损与小鼠肺气肿和癌症的同时发生联系在一起。此外,CCB 和 IGF2 特异性中和抗体是治疗这两种疾病的有效药物。
{"title":"Autocrine insulin-like growth factor 2 signaling as a potential target in the associated development of pulmonary emphysema and cancer in smokers.","authors":"Hye-Jin Boo, Hye-Young Min, Heung-Bin Lim, Euni Lee, Ho-Young Lee","doi":"10.1186/s41232-024-00344-3","DOIUrl":"10.1186/s41232-024-00344-3","url":null,"abstract":"<p><strong>Background: </strong>Tobacco smoking causes pulmonary inflammation, resulting in emphysema, an independent risk factor for lung cancer. Induction of insulin-like growth factor 2 (IGF2) in response to lung injury by tobacco carcinogens, 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol and polycyclic aromatic hydrocarbon benzo[a]pyrene in combination (NB), is critical for the proliferation of alveolar type 2 cells (AT2s) for lung repair. However, persistent IGF2 overexpression during NB-induced severe injury results in hyperproliferation of AT2s without coordinated AT2-to-AT1 differentiation, disrupting alveolar repair, which leads to the concurrent development of emphysema and lung cancer. The current study aims to verify the role of IGF2 signaling in the associated development of emphysema and cancer and develop effective pharmaceuticals for the diseases using animal models that recapitulate the characteristics of these chronic diseases.</p><p><strong>Methods: </strong>The pathogenesis of pulmonary emphysema and cancer was analyzed by lung function testing, histological evaluation, in situ zymography, dihydroethidium staining, and immunofluorescence and immunohistochemistry analyses utilizing mouse models of emphysema and cancer established by moderate exposure to NB for up to seven months.</p><p><strong>Results: </strong>Moderate NB exposure induced IGF2 expression in AT2s during the development of pulmonary emphysema and lung cancer in mice. Using AT2-specific insulin receptor knockout mice, we verified the causative role of sustained IGF2 signaling activation in AT2s in emphysema development. IGF2-targeting strategies, including voltage-dependent calcium channel blocker (CCB) and a neutralizing antibody, significantly suppressed the NB-induced development of emphysema and lung cancer. A publicly available database revealed an inverse correlation between the use of calcium channel blockers and a COPD diagnosis.</p><p><strong>Conclusions: </strong>Our work confirms sustained IGF2 signaling activation in AT2s couples impaired lung repair to the concurrent development of emphysema and cancer in mice. Additionally, CCB and IGF2-specific neutralizing antibodies are effective pharmaceuticals for the two diseases.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"31"},"PeriodicalIF":0.0,"publicationDate":"2024-06-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11191215/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141433670","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
Epigenetic targets of Janus kinase inhibitors are linked to genetic risks of rheumatoid arthritis. Janus 激酶抑制剂的表观遗传靶点与类风湿性关节炎的遗传风险有关。
Pub Date : 2024-06-04 DOI: 10.1186/s41232-024-00337-2
Haruka Tsuchiya, Mineto Ota, Haruka Takahashi, Hiroaki Hatano, Megumi Ogawa, Sotaro Nakajima, Risa Yoshihara, Tomohisa Okamura, Shuji Sumitomo, Keishi Fujio

Background: Current strategies that target cytokines (e.g., tumor necrosis factor (TNF)-α), or signaling molecules (e.g., Janus kinase (JAK)) have advanced the management for allergies and autoimmune diseases. Nevertheless, the molecular mechanism that underpins its clinical efficacy have largely remained elusive, especially in the local tissue environment. Here, we aimed to identify the genetic, epigenetic, and immunological targets of JAK inhibitors (JAKis), focusing on their effects on synovial fibroblasts (SFs), the major local effectors associated with destructive joint inflammation in rheumatoid arthritis (RA).

Methods: SFs were activated by cytokines related to inflammation in RA, and were treated with three types of JAKis or a TNF-α inhibitor (TNFi). Dynamic changes in transcriptome and chromatin accessibility were profiled across samples to identify drug targets. Furthermore, the putative targets were validated using luciferase assays and clustered regularly interspaced short palindromic repeat (CRISPR)-based genome editing.

Results: We found that both JAKis and the TNFi targeted the inflammatory module including IL6. Conversely, specific gene signatures that were preferentially inhibited by either of the drug classes were identified. Strikingly, RA risk enhancers for CD40 and TRAF1 were distinctively regulated by JAKis and the TNFi. We performed luciferase assays and CRISPR-based genome editing, and successfully fine-mapped the single causal variants in these loci, rs6074022-CD40 and rs7021049-TRAF1.

Conclusions: JAKis and the TNFi had a direct impact on different RA risk enhancers, and we identified nucleotide-resolution targets for both drugs. Distinctive targets of clinically effective drugs could be useful for tailoring the application of these drugs and future design of more efficient treatment strategies.

背景:目前针对细胞因子(如肿瘤坏死因子 (TNF)-α) 或信号分子(如 Janus 激酶 (JAK))的治疗策略推动了过敏症和自身免疫性疾病的治疗。然而,支撑其临床疗效的分子机制在很大程度上仍然难以捉摸,尤其是在局部组织环境中。在此,我们旨在确定 JAK 抑制剂(JAKis)的遗传学、表观遗传学和免疫学靶点,重点研究其对滑膜成纤维细胞(SFs)的影响,SFs 是类风湿性关节炎(RA)中与破坏性关节炎症相关的主要局部效应因子:方法:类风湿性关节炎(RA)患者的滑膜成纤维细胞被与炎症相关的细胞因子激活,并接受三种 JAKis 或 TNF-α 抑制剂(TNFi)的治疗。研究人员分析了各样本转录组和染色质可及性的动态变化,以确定药物靶点。此外,还利用荧光素酶测定法和基于聚类规则间隔短回文重复(CRISPR)的基因组编辑对推定靶点进行了验证:我们发现,JAKis 和 TNFi 都以包括 IL6 在内的炎症模块为靶点。相反,我们还发现了这两类药物优先抑制的特定基因特征。令人震惊的是,JAKis 和 TNFi 对 CD40 和 TRAF1 的 RA 风险增强子的调节作用截然不同。我们进行了荧光素酶测定和基于CRISPR的基因组编辑,并成功地精细绘制了这些位点(rs6074022-CD40和rs7021049-TRAF1)的单个致病变异体:JAKis和TNFi对不同的RA风险增强因子有直接影响,我们确定了这两种药物的核苷酸分辨率靶点。临床有效药物的独特靶点有助于调整这些药物的应用和未来更有效治疗策略的设计。
{"title":"Epigenetic targets of Janus kinase inhibitors are linked to genetic risks of rheumatoid arthritis.","authors":"Haruka Tsuchiya, Mineto Ota, Haruka Takahashi, Hiroaki Hatano, Megumi Ogawa, Sotaro Nakajima, Risa Yoshihara, Tomohisa Okamura, Shuji Sumitomo, Keishi Fujio","doi":"10.1186/s41232-024-00337-2","DOIUrl":"10.1186/s41232-024-00337-2","url":null,"abstract":"<p><strong>Background: </strong>Current strategies that target cytokines (e.g., tumor necrosis factor (TNF)-α), or signaling molecules (e.g., Janus kinase (JAK)) have advanced the management for allergies and autoimmune diseases. Nevertheless, the molecular mechanism that underpins its clinical efficacy have largely remained elusive, especially in the local tissue environment. Here, we aimed to identify the genetic, epigenetic, and immunological targets of JAK inhibitors (JAKis), focusing on their effects on synovial fibroblasts (SFs), the major local effectors associated with destructive joint inflammation in rheumatoid arthritis (RA).</p><p><strong>Methods: </strong>SFs were activated by cytokines related to inflammation in RA, and were treated with three types of JAKis or a TNF-α inhibitor (TNFi). Dynamic changes in transcriptome and chromatin accessibility were profiled across samples to identify drug targets. Furthermore, the putative targets were validated using luciferase assays and clustered regularly interspaced short palindromic repeat (CRISPR)-based genome editing.</p><p><strong>Results: </strong>We found that both JAKis and the TNFi targeted the inflammatory module including IL6. Conversely, specific gene signatures that were preferentially inhibited by either of the drug classes were identified. Strikingly, RA risk enhancers for CD40 and TRAF1 were distinctively regulated by JAKis and the TNFi. We performed luciferase assays and CRISPR-based genome editing, and successfully fine-mapped the single causal variants in these loci, rs6074022-CD40 and rs7021049-TRAF1.</p><p><strong>Conclusions: </strong>JAKis and the TNFi had a direct impact on different RA risk enhancers, and we identified nucleotide-resolution targets for both drugs. Distinctive targets of clinically effective drugs could be useful for tailoring the application of these drugs and future design of more efficient treatment strategies.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"29"},"PeriodicalIF":0.0,"publicationDate":"2024-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11149281/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141238853","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