首页 > 最新文献

Inflammation and regeneration最新文献

英文 中文
Cell fusion dynamics: mechanisms of multinucleation in osteoclasts and macrophages. 细胞融合动力学:破骨细胞和巨噬细胞的多核机制
Pub Date : 2024-11-27 DOI: 10.1186/s41232-024-00360-3
Hideaki Sabe, Yasuhito Yahara, Masaru Ishii

Cell-cell fusion is a vital biological process where the membranes of two or more cells merge to form a syncytium. This phenomenon is critical in various physiological and pathological contexts, including embryonic development, tissue repair, immune responses, and the progression of several diseases. Osteoclasts, which are cells from the monocyte/macrophage lineage responsible for bone resorption, have enhanced functionality due to cell fusion. Additionally, other multinucleated giant cells (MGCs) also arise from the fusion of monocytes and macrophages, typically during chronic inflammation and reactions to foreign materials such as prostheses or medical devices. Foreign body giant cells (FBGCs) and Langhans giant cells (LGCs) emerge only under pathological conditions and are involved in phagocytosis, antigen presentation, and the secretion of inflammatory mediators. This review provides a comprehensive overview of the mechanisms underlying the formation of multinucleated cells, with a particular emphasis on macrophages and osteoclasts. Elucidating the intracellular structures, signaling cascades, and fusion-mediating proteins involved in cell-cell fusion enhances our understanding of this fundamental biological process and helps identify potential therapeutic targets for disorders mediated by cell fusion.

细胞-细胞融合是一个重要的生物过程,在这一过程中,两个或多个细胞的细胞膜合并形成一个合胞体。这种现象在胚胎发育、组织修复、免疫反应和多种疾病的进展等各种生理和病理情况下都至关重要。破骨细胞是单核/巨噬细胞系的细胞,负责骨吸收,由于细胞融合,其功能得到增强。此外,其他多核巨细胞(MGC)也是由单核细胞和巨噬细胞融合产生的,通常发生在慢性炎症和对假体或医疗器械等外来材料的反应期间。异物巨细胞(FBGCs)和朗汉斯巨细胞(LGCs)只有在病理情况下才会出现,它们参与吞噬、抗原递呈和炎症介质的分泌。本综述全面概述了多核细胞的形成机制,并特别强调了巨噬细胞和破骨细胞。阐明细胞-细胞融合所涉及的细胞内结构、信号级联和融合介导蛋白,可加深我们对这一基本生物学过程的理解,并有助于确定细胞融合介导的疾病的潜在治疗靶点。
{"title":"Cell fusion dynamics: mechanisms of multinucleation in osteoclasts and macrophages.","authors":"Hideaki Sabe, Yasuhito Yahara, Masaru Ishii","doi":"10.1186/s41232-024-00360-3","DOIUrl":"https://doi.org/10.1186/s41232-024-00360-3","url":null,"abstract":"<p><p>Cell-cell fusion is a vital biological process where the membranes of two or more cells merge to form a syncytium. This phenomenon is critical in various physiological and pathological contexts, including embryonic development, tissue repair, immune responses, and the progression of several diseases. Osteoclasts, which are cells from the monocyte/macrophage lineage responsible for bone resorption, have enhanced functionality due to cell fusion. Additionally, other multinucleated giant cells (MGCs) also arise from the fusion of monocytes and macrophages, typically during chronic inflammation and reactions to foreign materials such as prostheses or medical devices. Foreign body giant cells (FBGCs) and Langhans giant cells (LGCs) emerge only under pathological conditions and are involved in phagocytosis, antigen presentation, and the secretion of inflammatory mediators. This review provides a comprehensive overview of the mechanisms underlying the formation of multinucleated cells, with a particular emphasis on macrophages and osteoclasts. Elucidating the intracellular structures, signaling cascades, and fusion-mediating proteins involved in cell-cell fusion enhances our understanding of this fundamental biological process and helps identify potential therapeutic targets for disorders mediated by cell fusion.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"49"},"PeriodicalIF":0.0,"publicationDate":"2024-11-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142741812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Designer immune cells. 设计免疫细胞
Pub Date : 2024-11-25 DOI: 10.1186/s41232-024-00362-1
Naoki Hosen
{"title":"Designer immune cells.","authors":"Naoki Hosen","doi":"10.1186/s41232-024-00362-1","DOIUrl":"10.1186/s41232-024-00362-1","url":null,"abstract":"","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"48"},"PeriodicalIF":0.0,"publicationDate":"2024-11-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11587678/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142717221","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
Macrophage depletion in inflamed rat knees prevents the activation of synovial mesenchymal stem cells by weakening Nampt and Spp1 signaling. 在发炎的大鼠膝关节中消耗巨噬细胞,可通过削弱 Nampt 和 Spp1 信号来防止滑膜间充质干细胞的活化。
Pub Date : 2024-11-20 DOI: 10.1186/s41232-024-00361-2
Hayato Kodama, Kentaro Endo, Ichiro Sekiya

Background: Macrophages and mesenchymal stem cells (MSCs) engage in crucial interplay during inflammation and have significant roles in tissue regeneration. Synovial MSCs, as key players in joint regeneration, are known to proliferate together with macrophages in synovitis. However, the crosstalk between synovial MSCs and macrophages remains unclear. In this study, we investigated changes in the activation of synovial MSCs in inflamed rat knees following selective depletion of macrophages with clodronate liposomes.

Methods: Acute inflammation was induced in rat knee joints by injection of carrageenan (day 0). Clodronate liposomes were administered intra-articularly on days 1 and 4 to deplete macrophages, with empty liposomes as a control. Knee joints were collected on day 7 for evaluation by histology, flow cytometry, and colony-forming assays. Concurrently, synovial MSCs were cultured and subjected to proliferation assays, flow cytometry, and chondrogenesis assessments. We also analyzed their crosstalk using single-cell RNA sequencing (scRNA-seq).

Results: Clodronate liposome treatment significantly reduced CD68-positive macrophage numbers and suppressed synovitis. Immunohistochemistry and flow cytometry showed decreased expression of CD68 (a macrophage marker) and CD44 and CD271 (MSC markers) in the clodronate group, while CD73 expression remained unchanged. The number of colony-forming cells per 1000 nucleated cells and per gram of synovium was significantly lower in the clodronate group than in the control group. Cultured synovial MSCs from both groups showed comparable proliferation, surface antigen expression, and chondrogenic capacity. scRNA-seq identified seven distinct synovial fibroblast (SF) subsets, with a notable decrease in the Mki67+ SF subset, corresponding to synovial MSCs, in the clodronate group. Clodronate treatment downregulated genes related to extracellular matrix organization and anabolic pathways in Mki67+ SF. Cell-cell communication analysis revealed diminished Nampt and Spp1 signaling interaction between macrophages and Mki67+ SF and diminished Ccl7, Spp1, and Csf1 signaling interaction between Mki67+ SF and macrophages in the clodronate group. Spp1 and Nampt promoted the proliferation and/or chondrogenesis of synovial MSCs.

Conclusions: Macrophage depletion with clodronate liposomes suppressed synovitis and reduced the number and activity of synovial MSCs, highlighting the significance of macrophage-derived Nampt and Spp1 signals in synovial MSC activation. These findings offer potential therapeutic strategies to promote joint tissue regeneration by enhancing beneficial signals between macrophages and synovial MSCs.

背景:巨噬细胞和间充质干细胞(MSCs)在炎症过程中会发生重要的相互作用,并在组织再生中发挥重要作用。滑膜间充质干细胞作为关节再生的关键角色,已知会在滑膜炎中与巨噬细胞一起增殖。然而,滑膜间充质干细胞与巨噬细胞之间的相互影响仍不清楚。在这项研究中,我们研究了用克洛膦酸脂质体选择性消耗巨噬细胞后,炎症大鼠膝关节滑膜间充质干细胞活化的变化:方法:通过注射卡拉胶诱导大鼠膝关节急性炎症(第 0 天)。第 1 天和第 4 天在关节内注射氯膦酸脂质体以消耗巨噬细胞,空脂质体作为对照。第 7 天收集膝关节,通过组织学、流式细胞术和集落形成试验进行评估。同时培养滑膜间充质干细胞,并对其进行增殖测定、流式细胞术和软骨形成评估。我们还利用单细胞 RNA 测序(scRNA-seq)分析了它们之间的相互影响:结果:氯膦酸脂质体治疗显著减少了CD68阳性巨噬细胞的数量,并抑制了滑膜炎。免疫组化和流式细胞术显示,氯膦酸钠组中 CD68(一种巨噬细胞标记物)、CD44 和 CD271(间充质干细胞标记物)的表达减少,而 CD73 的表达保持不变。每 1000 个有核细胞和每克滑膜中的集落形成细胞数在氯膦酸钠组明显低于对照组。scRNA-seq鉴定出了7个不同的滑膜成纤维细胞(SF)亚群,其中氯膦酸钠组中与滑膜间充质干细胞相对应的Mki67+ SF亚群明显减少。氯膦酸盐治疗下调了与细胞外基质组织和 Mki67+ SF 合成代谢途径相关的基因。细胞-细胞通讯分析显示,在氯膦酸钠组中,巨噬细胞与 Mki67+ SF 之间的 Nampt 和 Spp1 信号交互作用减弱,Mki67+ SF 与巨噬细胞之间的 Ccl7、Spp1 和 Csf1 信号交互作用减弱。Spp1和Nampt促进了滑膜间充质干细胞的增殖和/或软骨形成:结论:用氯膦酸脂质体消耗巨噬细胞可抑制滑膜炎,并减少滑膜间充质干细胞的数量和活性,突出了巨噬细胞衍生的Nampt和Spp1信号在滑膜间充质干细胞活化中的重要性。这些发现为通过增强巨噬细胞和滑膜间充质干细胞之间的有益信号来促进关节组织再生提供了潜在的治疗策略。
{"title":"Macrophage depletion in inflamed rat knees prevents the activation of synovial mesenchymal stem cells by weakening Nampt and Spp1 signaling.","authors":"Hayato Kodama, Kentaro Endo, Ichiro Sekiya","doi":"10.1186/s41232-024-00361-2","DOIUrl":"10.1186/s41232-024-00361-2","url":null,"abstract":"<p><strong>Background: </strong>Macrophages and mesenchymal stem cells (MSCs) engage in crucial interplay during inflammation and have significant roles in tissue regeneration. Synovial MSCs, as key players in joint regeneration, are known to proliferate together with macrophages in synovitis. However, the crosstalk between synovial MSCs and macrophages remains unclear. In this study, we investigated changes in the activation of synovial MSCs in inflamed rat knees following selective depletion of macrophages with clodronate liposomes.</p><p><strong>Methods: </strong>Acute inflammation was induced in rat knee joints by injection of carrageenan (day 0). Clodronate liposomes were administered intra-articularly on days 1 and 4 to deplete macrophages, with empty liposomes as a control. Knee joints were collected on day 7 for evaluation by histology, flow cytometry, and colony-forming assays. Concurrently, synovial MSCs were cultured and subjected to proliferation assays, flow cytometry, and chondrogenesis assessments. We also analyzed their crosstalk using single-cell RNA sequencing (scRNA-seq).</p><p><strong>Results: </strong>Clodronate liposome treatment significantly reduced CD68-positive macrophage numbers and suppressed synovitis. Immunohistochemistry and flow cytometry showed decreased expression of CD68 (a macrophage marker) and CD44 and CD271 (MSC markers) in the clodronate group, while CD73 expression remained unchanged. The number of colony-forming cells per 1000 nucleated cells and per gram of synovium was significantly lower in the clodronate group than in the control group. Cultured synovial MSCs from both groups showed comparable proliferation, surface antigen expression, and chondrogenic capacity. scRNA-seq identified seven distinct synovial fibroblast (SF) subsets, with a notable decrease in the Mki67<sup>+</sup> SF subset, corresponding to synovial MSCs, in the clodronate group. Clodronate treatment downregulated genes related to extracellular matrix organization and anabolic pathways in Mki67<sup>+</sup> SF. Cell-cell communication analysis revealed diminished Nampt and Spp1 signaling interaction between macrophages and Mki67<sup>+</sup> SF and diminished Ccl7, Spp1, and Csf1 signaling interaction between Mki67<sup>+</sup> SF and macrophages in the clodronate group. Spp1 and Nampt promoted the proliferation and/or chondrogenesis of synovial MSCs.</p><p><strong>Conclusions: </strong>Macrophage depletion with clodronate liposomes suppressed synovitis and reduced the number and activity of synovial MSCs, highlighting the significance of macrophage-derived Nampt and Spp1 signals in synovial MSC activation. These findings offer potential therapeutic strategies to promote joint tissue regeneration by enhancing beneficial signals between macrophages and synovial MSCs.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"47"},"PeriodicalIF":0.0,"publicationDate":"2024-11-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11577658/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142678161","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
The new era for the research on the regulation of microorganism-induced inflammation. 微生物诱发炎症调控研究的新时代。
Pub Date : 2024-11-12 DOI: 10.1186/s41232-024-00359-w
Kiyoshi Takeda
{"title":"The new era for the research on the regulation of microorganism-induced inflammation.","authors":"Kiyoshi Takeda","doi":"10.1186/s41232-024-00359-w","DOIUrl":"https://doi.org/10.1186/s41232-024-00359-w","url":null,"abstract":"","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"46"},"PeriodicalIF":0.0,"publicationDate":"2024-11-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11555862/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142635104","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
Focusing on exosomes to overcome the existing bottlenecks of CAR-T cell therapy. 关注外泌体,克服 CAR-T 细胞疗法的现有瓶颈。
Pub Date : 2024-11-04 DOI: 10.1186/s41232-024-00358-x
Si-Heng Zhang, Ling-Long Peng, Yi-Fei Chen, Yan Xu, Vahid Moradi

Since chimeric antigen receptor T (CAR-T) cells were introduced three decades ago, the treatment using these cells has led to outstanding outcomes, and at the moment, CAR-T cell therapy is a well-established mainstay for treating CD19 + malignancies and multiple myeloma. Despite the astonishing results of CAR-T cell therapy in B-cell-derived malignancies, several bottlenecks must be overcome to promote its safety and efficacy and broaden its applicability. These bottlenecks include cumbersome production process, safety concerns of viral vectors, poor efficacy in treating solid tumors, life-threatening side effects, and dysfunctionality of infused CAR-T cells over time. Exosomes are nano-sized vesicles that are secreted by all living cells and play an essential role in cellular crosstalk by bridging between cells. In this review, we discuss how the existing bottlenecks of CAR-T cell therapy can be overcome by focusing on exosomes. First, we delve into the effect of tumor-derived exosomes on the CAR-T cell function and discuss how inhibiting their secretion can enhance the efficacy of CAR-T cell therapy. Afterward, the application of exosomes to the manufacturing of CAR-T cells in a non-viral approach is discussed. We also review the latest advancements in ex vivo activation and cultivation of CAR-T cells using exosomes, as well as the potential of engineered exosomes to in vivo induction or boost the in vivo proliferation of CAR-T cells. Finally, we discuss how CAR-engineered exosomes can be used as a versatile tool for the direct killing of tumor cells or delivering intended therapeutic payloads in a targeted manner.

自 30 年前引入嵌合抗原受体 T(CAR-T)细胞以来,利用这些细胞进行的治疗取得了卓越的疗效,目前,CAR-T 细胞疗法已成为治疗 CD19 + 恶性肿瘤和多发性骨髓瘤的成熟疗法。尽管 CAR-T 细胞疗法在 B 细胞衍生恶性肿瘤中取得了惊人的疗效,但要提高其安全性和有效性并扩大其适用范围,还必须克服几个瓶颈。这些瓶颈包括繁琐的生产过程、病毒载体的安全性问题、治疗实体瘤的疗效不佳、危及生命的副作用以及输注的CAR-T细胞随着时间的推移会出现功能障碍。外泌体是所有活细胞分泌的纳米级囊泡,通过在细胞间架桥,在细胞串联中发挥着重要作用。在这篇综述中,我们将讨论如何通过关注外泌体来克服 CAR-T 细胞疗法的现有瓶颈。首先,我们深入探讨了肿瘤衍生的外泌体对 CAR-T 细胞功能的影响,并讨论了抑制外泌体分泌如何提高 CAR-T 细胞疗法的疗效。随后,我们讨论了外泌体在非病毒疗法中用于制造 CAR-T 细胞的应用。我们还回顾了利用外泌体体外激活和培养 CAR-T 细胞的最新进展,以及工程外泌体在体内诱导或促进 CAR-T 细胞体内增殖的潜力。最后,我们将讨论 CAR 工程外泌体如何作为一种多功能工具,直接杀死肿瘤细胞或以靶向方式递送预期的治疗载荷。
{"title":"Focusing on exosomes to overcome the existing bottlenecks of CAR-T cell therapy.","authors":"Si-Heng Zhang, Ling-Long Peng, Yi-Fei Chen, Yan Xu, Vahid Moradi","doi":"10.1186/s41232-024-00358-x","DOIUrl":"10.1186/s41232-024-00358-x","url":null,"abstract":"<p><p>Since chimeric antigen receptor T (CAR-T) cells were introduced three decades ago, the treatment using these cells has led to outstanding outcomes, and at the moment, CAR-T cell therapy is a well-established mainstay for treating CD19 + malignancies and multiple myeloma. Despite the astonishing results of CAR-T cell therapy in B-cell-derived malignancies, several bottlenecks must be overcome to promote its safety and efficacy and broaden its applicability. These bottlenecks include cumbersome production process, safety concerns of viral vectors, poor efficacy in treating solid tumors, life-threatening side effects, and dysfunctionality of infused CAR-T cells over time. Exosomes are nano-sized vesicles that are secreted by all living cells and play an essential role in cellular crosstalk by bridging between cells. In this review, we discuss how the existing bottlenecks of CAR-T cell therapy can be overcome by focusing on exosomes. First, we delve into the effect of tumor-derived exosomes on the CAR-T cell function and discuss how inhibiting their secretion can enhance the efficacy of CAR-T cell therapy. Afterward, the application of exosomes to the manufacturing of CAR-T cells in a non-viral approach is discussed. We also review the latest advancements in ex vivo activation and cultivation of CAR-T cells using exosomes, as well as the potential of engineered exosomes to in vivo induction or boost the in vivo proliferation of CAR-T cells. Finally, we discuss how CAR-engineered exosomes can be used as a versatile tool for the direct killing of tumor cells or delivering intended therapeutic payloads in a targeted manner.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"45"},"PeriodicalIF":0.0,"publicationDate":"2024-11-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11533312/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142570837","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
SPARC activates p38γ signaling to promote PFKFB3 protein stabilization and contributes to keloid fibroblast glycolysis. SPARC 激活 p38γ 信号,促进 PFKFB3 蛋白稳定,并有助于瘢痕成纤维细胞糖酵解。
Pub Date : 2024-10-31 DOI: 10.1186/s41232-024-00357-y
Yining Liu, Wei Zhang, Nan Lin, Zelei Yang, Yanxin Liu, Huaxia Chen

Background: Keloids are currently challenging to treat because they recur after resection which may affect patients' quality of life. At present, no universal consensus on treatment regimen has been established. Thus, finding new molecular mechanisms underlying keloid formation is imminent. This study aimed to explore the function of secreted protein acidic and cysteine rich (SPARC) on keloids and its behind exact mechanisms.

Methods: The expression of SPARC, p38γ, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), α-SMA, and Ki67 in patients with keloid and bleomycin (BLM)-induced fibrosis mice was assessed utilizing western blot, qRT-PCR, and immunohistochemical staining. After transfected with pcDNA-SPARC, si-SPARC-1#, si-SPARC-2#, and si-p38γ, and treated with glycolytic inhibitor (2-DG) or p38 inhibitor (SB203580), CCK-8, EdU, transwell, and western blot were utilized for assessing the proliferation, migration, and collagen production of keloid fibroblasts (KFs).

Results: SPARC, p38γ, and PFKFB3 were highly expressed in patients with keloid and BLM-induced fibrosis mice. SPARC promoted the proliferation, migration, and collagen production of KFs via inducing glycolysis. Moreover, SPARC could activate p38γ signaling to stabilize PFKFB3 protein expression in KFs. Next, we demonstrated that SPARC promoted the proliferation, migration, collagen production, and glycolysis of KFs via regulating p38γ signaling. In addition, in BLM-induced fibrosis mice, inhibition of p38γ and PFKFB3 relieved skin fibrosis.

Conclusions: Our findings indicated that SPARC could activate p38γ pathway to stabilize the expression of PFKFB3, and thus promote the glycolysis of KFs and the progression of keloid.

背景:瘢痕疙瘩是目前治疗的难题,因为切除后会复发,这可能会影响患者的生活质量。目前,治疗方案尚未达成普遍共识。因此,寻找瘢痕疙瘩形成的新分子机制迫在眉睫。本研究旨在探讨富含半胱氨酸的酸性分泌蛋白(SPARC)对瘢痕疙瘩的作用及其背后的确切机制:方法:采用Western blot、qRT-PCR和免疫组化染色等方法评估了瘢痕疙瘩患者和博莱霉素(BLM)诱导的纤维化小鼠体内SPARC、p38γ、6-磷酸果糖-2-激酶/果糖-2,6-二磷酸酶3(PFKFB3)、α-SMA和Ki67的表达。转染 pcDNA-SPARC、si-SPARC-1#、si-SPARC-2# 和 si-p38γ 并用糖酵解抑制剂(2-DG)或 p38 抑制剂(SB203580)处理后,利用 CCK-8、EdU、transwell 和 western blot 评估瘢痕疙瘩成纤维细胞(KFs)的增殖、迁移和胶原生成:结果:SPARC、p38γ和PFKFB3在瘢痕疙瘩患者和BLM诱导的纤维化小鼠中高表达。SPARC 通过诱导糖酵解促进 KFs 的增殖、迁移和胶原蛋白生成。此外,SPARC 还能激活 p38γ 信号转导,稳定 KFs 中 PFKFB3 蛋白的表达。接下来,我们证明了 SPARC 通过调节 p38γ 信号传导促进了 KFs 的增殖、迁移、胶原生成和糖酵解。此外,在BLM诱导的纤维化小鼠中,抑制p38γ和PFKFB3可缓解皮肤纤维化:我们的研究结果表明,SPARC可激活p38γ通路以稳定PFKFB3的表达,从而促进KFs的糖酵解和瘢痕疙瘩的进展。
{"title":"SPARC activates p38γ signaling to promote PFKFB3 protein stabilization and contributes to keloid fibroblast glycolysis.","authors":"Yining Liu, Wei Zhang, Nan Lin, Zelei Yang, Yanxin Liu, Huaxia Chen","doi":"10.1186/s41232-024-00357-y","DOIUrl":"10.1186/s41232-024-00357-y","url":null,"abstract":"<p><strong>Background: </strong>Keloids are currently challenging to treat because they recur after resection which may affect patients' quality of life. At present, no universal consensus on treatment regimen has been established. Thus, finding new molecular mechanisms underlying keloid formation is imminent. This study aimed to explore the function of secreted protein acidic and cysteine rich (SPARC) on keloids and its behind exact mechanisms.</p><p><strong>Methods: </strong>The expression of SPARC, p38γ, 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), α-SMA, and Ki67 in patients with keloid and bleomycin (BLM)-induced fibrosis mice was assessed utilizing western blot, qRT-PCR, and immunohistochemical staining. After transfected with pcDNA-SPARC, si-SPARC-1#, si-SPARC-2#, and si-p38γ, and treated with glycolytic inhibitor (2-DG) or p38 inhibitor (SB203580), CCK-8, EdU, transwell, and western blot were utilized for assessing the proliferation, migration, and collagen production of keloid fibroblasts (KFs).</p><p><strong>Results: </strong>SPARC, p38γ, and PFKFB3 were highly expressed in patients with keloid and BLM-induced fibrosis mice. SPARC promoted the proliferation, migration, and collagen production of KFs via inducing glycolysis. Moreover, SPARC could activate p38γ signaling to stabilize PFKFB3 protein expression in KFs. Next, we demonstrated that SPARC promoted the proliferation, migration, collagen production, and glycolysis of KFs via regulating p38γ signaling. In addition, in BLM-induced fibrosis mice, inhibition of p38γ and PFKFB3 relieved skin fibrosis.</p><p><strong>Conclusions: </strong>Our findings indicated that SPARC could activate p38γ pathway to stabilize the expression of PFKFB3, and thus promote the glycolysis of KFs and the progression of keloid.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"44"},"PeriodicalIF":0.0,"publicationDate":"2024-10-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11529245/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142559856","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
Deletion of TP signaling in macrophages delays liver repair following APAP-induced liver injury by reducing accumulation of reparative macrophage and production of HGF. 巨噬细胞中 TP 信号的缺失会减少修复性巨噬细胞的聚集和 HGF 的产生,从而延迟 APAP 诱导的肝损伤后的肝修复。
Pub Date : 2024-10-03 DOI: 10.1186/s41232-024-00356-z
Mina Tanabe, Kanako Hosono, Atsushi Yamashita, Yoshiya Ito, Masataka Majima, Shuh Narumiya, Chika Kusano, Hideki Amano

Background: Acetaminophen (APAP)-induced liver injury is the most common cause of acute liver failure. Macrophages are key players in liver restoration following APAP-induced liver injury. Thromboxane A2 (TXA2) and its receptor, thromboxane prostanoid (TP) receptor, have been shown to be involved in tissue repair. However, whether TP signaling plays a role in liver repair after APAP hepatotoxicity by affecting macrophage function remains unclear.

Methods: Male TP knockout (TP-/-) and C57BL/6 wild-type (WT) mice were treated with APAP (300 mg/kg). In addition, macrophage-specific TP-knockout (TP△mac) and control WT mice were treated with APAP. We explored changes in liver inflammation, liver repair, and macrophage accumulation in mice treated with APAP.

Results: Compared with WT mice, TP-/- mice showed aggravated liver injury as indicated by increased levels of alanine transaminase (ALT) and necrotic area as well as delayed liver repair as indicated by decreased expression of proliferating cell nuclear antigen (PCNA). Macrophage deletion exacerbated APAP-induced liver injury and impaired liver repair. Transplantation of TP-deficient bone marrow (BM) cells to WT or TP-/- mice aggravated APAP hepatotoxicity with suppressed accumulation of macrophages, while transplantation of WT-BM cells to WT or TP-/- mice attenuated APAP-induced liver injury with accumulation of macrophages in the injured regions. Macrophage-specific TP-/- mice exacerbated liver injury and delayed liver repair, which was associated with increased pro-inflammatory macrophages and decreased reparative macrophages and hepatocyte growth factor (HGF) expression. In vitro, TP signaling facilitated macrophage polarization to a reparative phenotype. Transfer of cultured BM-derived macrophages from control mice to macrophage-specific TP-/- mice attenuated APAP-induced liver injury and promoted liver repair. HGF treatment mitigated APAP-induced inflammation and promoted liver repair after APAP-induced liver injury.

Conclusions: Deletion of TP signaling in macrophages delays liver repair following APAP-induced liver injury, which is associated with reduced accumulation of reparative macrophages and the hepatotrophic factor HGF. Specific activation of TP signaling in macrophages may be a potential therapeutic target for liver repair and regeneration after APAP hepatotoxicity.

背景:对乙酰氨基酚(APAP)引起的肝损伤是急性肝衰竭最常见的原因。巨噬细胞是 APAP 诱导肝损伤后肝脏恢复的关键角色。血栓素 A2(TXA2)及其受体--血栓素类前列腺素(TP)受体已被证明参与组织修复。然而,TP 信号是否通过影响巨噬细胞功能在 APAP 肝毒性后的肝脏修复中发挥作用仍不清楚:方法:雄性 TP 基因敲除(TP-/-)小鼠和 C57BL/6 野生型(WT)小鼠均接受 APAP(300 mg/kg)治疗。此外,巨噬细胞特异性TP基因敲除(TP△mac)小鼠和对照WT小鼠也接受了APAP治疗。我们探讨了使用 APAP 治疗的小鼠肝脏炎症、肝脏修复和巨噬细胞积累的变化:结果:与 WT 小鼠相比,TP-/- 小鼠的丙氨酸转氨酶(ALT)水平和坏死面积增加,表明肝损伤加重;增殖细胞核抗原(PCNA)表达减少,表明肝修复延迟。巨噬细胞的缺失加剧了 APAP 诱导的肝损伤并损害了肝修复。将 TP 缺失的骨髓(BM)细胞移植给 WT 或 TP-/- 小鼠会加重 APAP 的肝毒性,同时抑制巨噬细胞的聚集;而将 WT-BM 细胞移植给 WT 或 TP-/- 小鼠会减轻 APAP 诱导的肝损伤,同时在损伤区域聚集巨噬细胞。巨噬细胞特异性 TP-/- 小鼠加剧了肝损伤并延迟了肝修复,这与促炎性巨噬细胞增加、修复性巨噬细胞和肝细胞生长因子(HGF)表达减少有关。在体外,TP 信号促进巨噬细胞极化为修复表型。将培养的巨噬细胞从对照组小鼠转移到巨噬细胞特异性 TP-/- 小鼠体内,可减轻 APAP 引起的肝损伤并促进肝修复。HGF治疗减轻了APAP诱导的炎症,促进了APAP诱导的肝损伤后的肝修复:结论:巨噬细胞中 TP 信号的缺失会延迟 APAP 诱导的肝损伤后的肝修复,这与修复性巨噬细胞和肝营养因子 HGF 的积累减少有关。特异性激活巨噬细胞中的 TP 信号可能是 APAP 肝毒性后肝脏修复和再生的潜在治疗靶点。
{"title":"Deletion of TP signaling in macrophages delays liver repair following APAP-induced liver injury by reducing accumulation of reparative macrophage and production of HGF.","authors":"Mina Tanabe, Kanako Hosono, Atsushi Yamashita, Yoshiya Ito, Masataka Majima, Shuh Narumiya, Chika Kusano, Hideki Amano","doi":"10.1186/s41232-024-00356-z","DOIUrl":"10.1186/s41232-024-00356-z","url":null,"abstract":"<p><strong>Background: </strong>Acetaminophen (APAP)-induced liver injury is the most common cause of acute liver failure. Macrophages are key players in liver restoration following APAP-induced liver injury. Thromboxane A<sub>2</sub> (TXA<sub>2</sub>) and its receptor, thromboxane prostanoid (TP) receptor, have been shown to be involved in tissue repair. However, whether TP signaling plays a role in liver repair after APAP hepatotoxicity by affecting macrophage function remains unclear.</p><p><strong>Methods: </strong>Male TP knockout (TP<sup>-/-</sup>) and C57BL/6 wild-type (WT) mice were treated with APAP (300 mg/kg). In addition, macrophage-specific TP-knockout (TP<sup>△mac</sup>) and control WT mice were treated with APAP. We explored changes in liver inflammation, liver repair, and macrophage accumulation in mice treated with APAP.</p><p><strong>Results: </strong>Compared with WT mice, TP<sup>-/-</sup> mice showed aggravated liver injury as indicated by increased levels of alanine transaminase (ALT) and necrotic area as well as delayed liver repair as indicated by decreased expression of proliferating cell nuclear antigen (PCNA). Macrophage deletion exacerbated APAP-induced liver injury and impaired liver repair. Transplantation of TP-deficient bone marrow (BM) cells to WT or TP<sup>-/-</sup> mice aggravated APAP hepatotoxicity with suppressed accumulation of macrophages, while transplantation of WT-BM cells to WT or TP<sup>-/-</sup> mice attenuated APAP-induced liver injury with accumulation of macrophages in the injured regions. Macrophage-specific TP<sup>-/-</sup> mice exacerbated liver injury and delayed liver repair, which was associated with increased pro-inflammatory macrophages and decreased reparative macrophages and hepatocyte growth factor (HGF) expression. In vitro, TP signaling facilitated macrophage polarization to a reparative phenotype. Transfer of cultured BM-derived macrophages from control mice to macrophage-specific TP<sup>-/-</sup> mice attenuated APAP-induced liver injury and promoted liver repair. HGF treatment mitigated APAP-induced inflammation and promoted liver repair after APAP-induced liver injury.</p><p><strong>Conclusions: </strong>Deletion of TP signaling in macrophages delays liver repair following APAP-induced liver injury, which is associated with reduced accumulation of reparative macrophages and the hepatotrophic factor HGF. Specific activation of TP signaling in macrophages may be a potential therapeutic target for liver repair and regeneration after APAP hepatotoxicity.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"43"},"PeriodicalIF":0.0,"publicationDate":"2024-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11451145/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142373929","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
Beyond the bulk: overview and novel insights into the dynamics of muscle satellite cells during muscle regeneration. 超越体积:肌肉再生过程中肌肉卫星细胞动态的概述和新见解。
Pub Date : 2024-09-26 DOI: 10.1186/s41232-024-00354-1
Woo Seok Byun, Jinu Lee, Jea-Hyun Baek

Skeletal muscle possesses remarkable regenerative capabilities, fully recovering within a month following severe acute damage. Central to this process are muscle satellite cells (MuSCs), a resident population of somatic stem cells capable of self-renewal and differentiation. Despite the highly predictable course of muscle regeneration, evaluating this process has been challenging due to the heterogeneous nature of myogenic precursors and the limited insight provided by traditional markers with overlapping expression patterns. Notably, recent advancements in single-cell technologies, such as single-cell (scRNA-seq) and single-nucleus RNA sequencing (snRNA-seq), have revolutionized muscle research. These approaches allow for comprehensive profiling of individual cells, unveiling dynamic heterogeneity among myogenic precursors and their contributions to regeneration. Through single-cell transcriptome analyses, researchers gain valuable insights into cellular diversity and functional dynamics of MuSCs post-injury. This review aims to consolidate classical and new insights into the heterogeneity of myogenic precursors, including the latest discoveries from novel single-cell technologies.

骨骼肌具有惊人的再生能力,在严重急性损伤后一个月内就能完全恢复。这一过程的核心是肌肉卫星细胞(MuSCs),这是一种能够自我更新和分化的体细胞干细胞常驻群。尽管肌肉再生过程具有高度可预测性,但由于肌肉生成前体的异质性,以及表达模式重叠的传统标记物提供的洞察力有限,评估这一过程一直具有挑战性。值得注意的是,单细胞技术(如单细胞(scRNA-seq)和单核 RNA 测序(snRNA-seq))的最新进展彻底改变了肌肉研究。这些方法可对单个细胞进行全面分析,揭示肌原性前体的动态异质性及其对再生的贡献。通过单细胞转录组分析,研究人员获得了有关损伤后造血干细胞的细胞多样性和功能动态的宝贵见解。本综述旨在整合对肌原纤维前体异质性的经典见解和新见解,包括新型单细胞技术的最新发现。
{"title":"Beyond the bulk: overview and novel insights into the dynamics of muscle satellite cells during muscle regeneration.","authors":"Woo Seok Byun, Jinu Lee, Jea-Hyun Baek","doi":"10.1186/s41232-024-00354-1","DOIUrl":"https://doi.org/10.1186/s41232-024-00354-1","url":null,"abstract":"<p><p>Skeletal muscle possesses remarkable regenerative capabilities, fully recovering within a month following severe acute damage. Central to this process are muscle satellite cells (MuSCs), a resident population of somatic stem cells capable of self-renewal and differentiation. Despite the highly predictable course of muscle regeneration, evaluating this process has been challenging due to the heterogeneous nature of myogenic precursors and the limited insight provided by traditional markers with overlapping expression patterns. Notably, recent advancements in single-cell technologies, such as single-cell (scRNA-seq) and single-nucleus RNA sequencing (snRNA-seq), have revolutionized muscle research. These approaches allow for comprehensive profiling of individual cells, unveiling dynamic heterogeneity among myogenic precursors and their contributions to regeneration. Through single-cell transcriptome analyses, researchers gain valuable insights into cellular diversity and functional dynamics of MuSCs post-injury. This review aims to consolidate classical and new insights into the heterogeneity of myogenic precursors, including the latest discoveries from novel single-cell technologies.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"39"},"PeriodicalIF":0.0,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11426090/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142335196","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
Engraftment of human mesenchymal stem cells in a severely immunodeficient mouse. 人类间充质干细胞在严重免疫缺陷小鼠体内的移植。
Pub Date : 2024-09-26 DOI: 10.1186/s41232-024-00353-2
Yuko Kato, Yusuke Ohno, Ryoji Ito, Takeshi Taketani, Yumi Matsuzaki, Satoru Miyagi

The transplantation of human mesenchymal stromal/stem cells (hMSCs) has potential as a curative and permanent therapy for congenital skeletal diseases. However, the self-renewal and differentiation capacities of hMSCs markedly vary. Therefore, cell proliferation and trilineage differentiation capacities were tested in vitro to characterize hMSCs before their clinical use. However, it remains unclear whether the ability of hMSCs in vitro accurately predicts that in living animals. The xenograft model is an alternative method for validating clinical MSCs. Nevertheless, the protocol still needs refinement, and it has yet to be established whether hMSCs, which are expanded in culture for clinical use, retain the ability to engraft and differentiate into adipogenic, osteogenic, and chondrogenic lineage cells in transplantation settings. In the present study, to establish a robust xenograft model of MSCs, we examined the delivery routes of hMSCs and the immunological state of recipients. The intra-arterial injection of hMSCs into X-ray-irradiated (IR) NOG, a severely immunodeficient mouse, achieved the highest engraftment but failed to sustain long-term engraftment. We demonstrated that graft cells localized to a collagenase-released fraction (CR), in which endogenous colony-forming cells reside. We also showed that Pdgfrα+Sca1+ MSCs (PαS), which reside in the CR fraction, resisted IR. These results show that our protocol enables hMSCs to fulfill a high level of engraftment in mouse bone marrow in the short term. In contrast, long-term reconstitution was restricted, at least partially, because of IR-resistant endogenous MSCs.

移植人类间充质基质/干细胞(hMSCs)具有治疗先天性骨骼疾病的潜力。然而,间充质干细胞的自我更新和分化能力存在明显差异。因此,在将 hMSCs 用于临床之前,我们对其细胞增殖和三系分化能力进行了体外测试,以确定其特征。然而,目前仍不清楚 hMSCs 在体外的能力是否能准确预测其在活体动物体内的能力。异种移植模型是验证临床间充质干细胞的另一种方法。尽管如此,该方法仍需改进,而且临床使用的经培养扩增的 hMSCs 在移植环境中是否仍能保留其吞噬和分化成成脂、成骨和软骨系细胞的能力也有待确定。在本研究中,为了建立稳健的间充质干细胞异种移植模型,我们研究了 hMSCs 的输送途径和受体的免疫状态。将 hMSCs 经动脉内注射到经 X 射线照射(IR)的 NOG(一种严重免疫缺陷的小鼠)体内可获得最高的接种率,但无法维持长期接种。我们证实,移植物细胞定位在胶原酶释放部分(CR),内源性集落形成细胞就存在于该部分。我们还发现,存在于 CR 部分的 Pdgfrα+Sca1+ 间充质干细胞(PαS)可抵抗 IR。这些结果表明,我们的方案能使 hMSCs 在短期内实现在小鼠骨髓中的高水平移植。与此相反,由于内源性间充质干细胞对红外有抵抗力,长期重建至少部分受到了限制。
{"title":"Engraftment of human mesenchymal stem cells in a severely immunodeficient mouse.","authors":"Yuko Kato, Yusuke Ohno, Ryoji Ito, Takeshi Taketani, Yumi Matsuzaki, Satoru Miyagi","doi":"10.1186/s41232-024-00353-2","DOIUrl":"https://doi.org/10.1186/s41232-024-00353-2","url":null,"abstract":"<p><p>The transplantation of human mesenchymal stromal/stem cells (hMSCs) has potential as a curative and permanent therapy for congenital skeletal diseases. However, the self-renewal and differentiation capacities of hMSCs markedly vary. Therefore, cell proliferation and trilineage differentiation capacities were tested in vitro to characterize hMSCs before their clinical use. However, it remains unclear whether the ability of hMSCs in vitro accurately predicts that in living animals. The xenograft model is an alternative method for validating clinical MSCs. Nevertheless, the protocol still needs refinement, and it has yet to be established whether hMSCs, which are expanded in culture for clinical use, retain the ability to engraft and differentiate into adipogenic, osteogenic, and chondrogenic lineage cells in transplantation settings. In the present study, to establish a robust xenograft model of MSCs, we examined the delivery routes of hMSCs and the immunological state of recipients. The intra-arterial injection of hMSCs into X-ray-irradiated (IR) NOG, a severely immunodeficient mouse, achieved the highest engraftment but failed to sustain long-term engraftment. We demonstrated that graft cells localized to a collagenase-released fraction (CR), in which endogenous colony-forming cells reside. We also showed that Pdgfrα<sup>+</sup>Sca1<sup>+</sup> MSCs (PαS), which reside in the CR fraction, resisted IR. These results show that our protocol enables hMSCs to fulfill a high level of engraftment in mouse bone marrow in the short term. In contrast, long-term reconstitution was restricted, at least partially, because of IR-resistant endogenous MSCs.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"40"},"PeriodicalIF":0.0,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11426203/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142335197","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
Peripheral and central regulation of neuro-immune crosstalk. 神经免疫串扰的外周和中枢调控
Pub Date : 2024-09-26 DOI: 10.1186/s41232-024-00352-3
Mayuko Izumi, Yoshimitsu Nakanishi, Sujin Kang, Atsushi Kumanogoh

The neural and immune systems sense and respond to external stimuli to maintain tissue homeostasis. These systems do not function independently but rather interact with each other to effectively exert biological actions and prevent disease pathogenesis, such as metabolic, inflammatory, and infectious disorders. Mutual communication between these systems is also affected by tissue niche-specific signals that reflect the tissue environment. However, the regulatory mechanisms underlying these interactions are not completely understood. In addition to the peripheral regulation of neuro-immune crosstalk, recent studies have reported that the central nervous system plays essential roles in the regulation of systemic neuro-immune interactions. In this review, we provide an overview of the molecular basis of peripheral and systemic neuro-immune crosstalk and explore how these multilayered interactions are maintained.

神经系统和免疫系统能够感知外部刺激并做出反应,以维持组织的平衡。这些系统并不是独立运作的,而是相互影响,以有效地发挥生物作用和预防疾病的发病,如代谢、炎症和感染性疾病。这些系统之间的相互交流还受到反映组织环境的组织特异性信号的影响。然而,这些相互作用的调控机制尚未完全明了。除了神经-免疫串扰的外周调控外,最近的研究还发现,中枢神经系统在调控全身神经-免疫相互作用中发挥着至关重要的作用。在这篇综述中,我们将概述外周和全身神经-免疫相互协作的分子基础,并探讨如何维持这些多层次的相互作用。
{"title":"Peripheral and central regulation of neuro-immune crosstalk.","authors":"Mayuko Izumi, Yoshimitsu Nakanishi, Sujin Kang, Atsushi Kumanogoh","doi":"10.1186/s41232-024-00352-3","DOIUrl":"https://doi.org/10.1186/s41232-024-00352-3","url":null,"abstract":"<p><p>The neural and immune systems sense and respond to external stimuli to maintain tissue homeostasis. These systems do not function independently but rather interact with each other to effectively exert biological actions and prevent disease pathogenesis, such as metabolic, inflammatory, and infectious disorders. Mutual communication between these systems is also affected by tissue niche-specific signals that reflect the tissue environment. However, the regulatory mechanisms underlying these interactions are not completely understood. In addition to the peripheral regulation of neuro-immune crosstalk, recent studies have reported that the central nervous system plays essential roles in the regulation of systemic neuro-immune interactions. In this review, we provide an overview of the molecular basis of peripheral and systemic neuro-immune crosstalk and explore how these multilayered interactions are maintained.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"41"},"PeriodicalIF":0.0,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11426056/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142335198","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