Inflammation and regeneration最新文献

筛选
英文 中文
Multi-organ frailty is enhanced by periodontitis-induced inflammaging.
Inflammation and regeneration Pub Date : 2025-02-03 DOI: 10.1186/s41232-025-00366-5
Yoshitaka Kase, Satoru Morikawa, Yuji Okano, Tatsuya Hosoi, Takazumi Yasui, Yoko Taki-Miyashita, Mitsutaka Yakabe, Maraku Goto, Kazuyuki Ishihara, Sumito Ogawa, Taneaki Nakagawa, Hideyuki Okano
{"title":"Multi-organ frailty is enhanced by periodontitis-induced inflammaging.","authors":"Yoshitaka Kase, Satoru Morikawa, Yuji Okano, Tatsuya Hosoi, Takazumi Yasui, Yoko Taki-Miyashita, Mitsutaka Yakabe, Maraku Goto, Kazuyuki Ishihara, Sumito Ogawa, Taneaki Nakagawa, Hideyuki Okano","doi":"10.1186/s41232-025-00366-5","DOIUrl":"https://doi.org/10.1186/s41232-025-00366-5","url":null,"abstract":"<p><strong>Background: </strong>The incidence of periodontitis is high in older individuals. However, its impact on multi-organ frailty remains unclear. We developed mouse models with varying severity and duration of periodontitis to examine its effects.</p><p><strong>Methods: </strong>We generated mouse models with mild and severe periodontitis, categorizing the disease duration into 3-month and 5-month periods for analysis. The organs assessed for frailty included the gastrocnemius muscle, soleus muscle, brain, and femur.</p><p><strong>Results: </strong>Our study found that periodontitis induced systemic inflammation resembling inflammaging and other symptoms characteristic of age-induced frailty. Notably, muscle impairment developed specifically in slow-twitch muscles, and the femur emerged as the most vulnerable bone, exhibiting reduced bone mineral density even with mild and short-duration periodontitis. This condition resulted in the co-occurrence of bone fragility and slow-twitch muscle dysfunction. Cognitive function assessment revealed increased activated microglia and decreased adult neurogenesis in the hippocampus, impairing spatial learning. Thus, periodontitis induced both physical and cognitive frailties. Therapeutic intervention for the periodontitis, which halted the exacerbation of bone resorption markers, did not restore femur bone mineral density.</p><p><strong>Conclusion: </strong>This study underscores the role of periodontitis in inducing multifaceted organ frailty with vulnerability, varying by organ, and the necessity of early intervention, particularly regarding bone density loss.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"45 1","pages":"3"},"PeriodicalIF":0.0,"publicationDate":"2025-02-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143082420","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
Amelioration of liver fibrosis with autologous macrophages induced by IL-34-based condition.
Inflammation and regeneration Pub Date : 2025-01-24 DOI: 10.1186/s41232-025-00364-7
Yuichi Igarashi, Haruka Wada, Masato Muto, Ryohei Sone, Yoshinori Hasegawa, Ken-Ichiro Seino
{"title":"Amelioration of liver fibrosis with autologous macrophages induced by IL-34-based condition.","authors":"Yuichi Igarashi, Haruka Wada, Masato Muto, Ryohei Sone, Yoshinori Hasegawa, Ken-Ichiro Seino","doi":"10.1186/s41232-025-00364-7","DOIUrl":"10.1186/s41232-025-00364-7","url":null,"abstract":"<p><strong>Background: </strong>For the treatment of liver fibrosis, several novel cell therapies have been proposed. Autologous macrophage therapy has been reported as one of the promising treatments. So far, most studies have used colony-stimulating factor 1 (CSF-1) to induce the differentiation of macrophage progenitor cells. The receptor for CSF-1, CSF-1R possesses another ligand, interleukin 34. However, the therapeutic capacity for liver fibrosis by interleukin 34-induced macrophages has not been evaluated.</p><p><strong>Methods: </strong>We have employed acute (bile duct ligation) and chronic (administration of carbon tetrachloride or thioacetamide) liver fibrosis models. Using these models, we evaluated the therapeutic capacity of macrophages induced by interleukin 34-based conditions. In most experiments, interleukin 4 was also added to the differentiation process to induce alternative-activated macrophages. As a mechanism analysis, we have examined liver inflammation and damage, the status of stellate cells, and the immunosuppressive capacity of the macrophages. Human macrophages were differentiated from CD14<sup>+</sup> monocytes and analyzed.</p><p><strong>Results: </strong>In both acute and chronic liver damage experiments, interleukin 34-induced macrophages significantly ameliorated liver fibrosis. The addition of interleukin 4 to the differentiation process resulted in an increase of obtained macrophages and a bias to alternative activated macrophages (so-called M2). The alternative activated macrophages (M2-type) showed a reproducible therapeutic effect of liver fibrosis with a suppression of parameters of liver inflammation and damage, stellate cells, and T cell activation. Similar macrophages could be differentiated from human CD14<sup>+</sup> monocytes in the presence of interleukin 34 plus interleukin 4, and a therapeutic effect was observed using a humanized mouse model.</p><p><strong>Conclusions: </strong>Interleukin 34-induced macrophages, particularly when additionally stimulated with interleukin 4, significantly ameliorated the liver fibrosis.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"45 1","pages":"2"},"PeriodicalIF":0.0,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11758727/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143043895","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
Human PBMC-based humanized mice exhibit myositis features and serve as a drug evaluation model. 基于人pbmc的人源化小鼠表现出肌炎特征,可作为药物评价模型。
Inflammation and regeneration Pub Date : 2025-01-15 DOI: 10.1186/s41232-025-00365-6
Akiko Nishidate, Kana Takemoto, Yuki Imura, Mikako Murase, Ryu Yamanaka, Manami Kikuchi, Junpei Anan, Sayuka Kato, Airi Akatsuka, Sachiko Mochizuki, Yuzo Koda
{"title":"Human PBMC-based humanized mice exhibit myositis features and serve as a drug evaluation model.","authors":"Akiko Nishidate, Kana Takemoto, Yuki Imura, Mikako Murase, Ryu Yamanaka, Manami Kikuchi, Junpei Anan, Sayuka Kato, Airi Akatsuka, Sachiko Mochizuki, Yuzo Koda","doi":"10.1186/s41232-025-00365-6","DOIUrl":"10.1186/s41232-025-00365-6","url":null,"abstract":"<p><p>Idiopathic inflammatory myopathies (IIMs) are a group of autoimmune disorders characterized by immune cell infiltration of muscle tissue accompanied by inflammation. Treatment of IIMs is challenging, with few effective therapeutic options due to the lack of appropriate models that successfully recapitulate the features of IIMs observed in humans. In the present study, we demonstrate that immunodeficient mice transplanted with human peripheral blood mononuclear cells (hPBMCs) exhibit the key pathologic features of myositis observed in humans and develop graft-versus-host disease. The hPBMC mice exhibit elevated serum levels of creatine kinase and aspartate transaminase, markers of myositis, and increased expression levels of myositis-related genes, such as vascular cell adhesion molecule 1, intercellular adhesion molecule 1, and serum amyloid A1, in muscle tissues. Histopathologic and flow cytometric analyses reveal the infiltration of CD8<sup>+</sup> T cells in the muscle tissue of hPBMC mice, similar to that observed in patients with myositis, particularly in those with polymyositis. Transplantation of CD8<sup>+</sup> T cell-depleted hPBMC leads to a significant reduction in polymyositis-like symptoms, in agreement with previous studies demonstrating CD8<sup>+</sup> T cells as the main pathologic drivers of polymyositis. Furthermore, the transcriptome analysis of muscle tissue from hPBMC mice reveal extensive upregulation of characteristic genes of polymyositis, providing further support that hPBMC mice accurately reflect the pathophysiology of myositis in humans. Finally, administration of prednisolone or tacrolimus, which are commonly used for IIM treatment, leads to significant alleviation of myositis findings. Therefore, we propose that hPBMC mice should be considered as a model that accurately reflects the pathophysiology of myositis in human patients.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"45 1","pages":"1"},"PeriodicalIF":0.0,"publicationDate":"2025-01-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11734410/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142985498","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
Ca2+ signaling in vascular smooth muscle and endothelial cells in blood vessel remodeling: a review. 血管重构中血管平滑肌和内皮细胞Ca2+信号的研究进展
Inflammation and regeneration Pub Date : 2024-12-27 DOI: 10.1186/s41232-024-00363-0
Yoshiaki Suzuki, Wayne R Giles, Gerald W Zamponi, Rubii Kondo, Yuji Imaizumi, Hisao Yamamura
{"title":"Ca<sup>2+</sup> signaling in vascular smooth muscle and endothelial cells in blood vessel remodeling: a review.","authors":"Yoshiaki Suzuki, Wayne R Giles, Gerald W Zamponi, Rubii Kondo, Yuji Imaizumi, Hisao Yamamura","doi":"10.1186/s41232-024-00363-0","DOIUrl":"10.1186/s41232-024-00363-0","url":null,"abstract":"<p><p>Vascular smooth muscle cells (VSMCs) and endothelial cells (ECs) act together to regulate blood pressure and systemic blood flow by appropriately adjusting blood vessel diameter in response to biochemical or biomechanical stimuli. Ion channels that are expressed in these cells regulate membrane potential and cytosolic Ca<sup>2+</sup> concentration ([Ca<sup>2+</sup>]<sub>cyt</sub>) in response to such stimuli. The subsets of these ion channels involved in Ca<sup>2+</sup> signaling often form molecular complexes with intracellular molecules via scaffolding proteins. This allows Ca<sup>2+</sup> signaling to be tightly controlled in localized areas within the cell, resulting in a balanced vascular tone. When hypertensive stimuli are applied to blood vessels for extended periods, gene expression in these vascular cells can change dramatically. For example, alteration in ion channel expression often induces electrical remodeling that produces a depolarization of the membrane potential and elevated [Ca<sup>2+</sup>]<sub>cyt</sub>. Coupled with endothelial dysfunction blood vessels undergo functional remodeling characterized by enhanced vasoconstriction. In addition, pathological challenges to vascular cells can induce inflammatory gene products that may promote leukocyte infiltration, in part through Ca<sup>2+</sup>-dependent pathways. Macrophages accumulating in the vascular adventitia promote fibrosis through extracellular matrix turnover, and cause structural remodeling of blood vessels. This functional and structural remodeling often leads to chronic hypertension affecting not only blood vessels, but also multiple organs including the brain, kidneys, and heart, thus increasing the risk of severe cardiovascular events. In this review, we outline recent advances in multidisciplinary research concerning Ca<sup>2+</sup> signaling in VSMCs and ECs, with an emphasis on the mechanisms underlying functional and structural vascular remodeling.</p>","PeriodicalId":94041,"journal":{"name":"Inflammation and regeneration","volume":"44 1","pages":"50"},"PeriodicalIF":0.0,"publicationDate":"2024-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11673324/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142901133","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
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
Inflammation and regeneration Pub Date : 2024-11-27 DOI: 10.1186/s41232-024-00360-3
Hideaki Sabe, Yasuhito Yahara, Masaru Ishii
{"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":"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":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11600601/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142741812","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
Designer immune cells. 设计免疫细胞
Inflammation and regeneration 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。
Inflammation and regeneration Pub Date : 2024-11-20 DOI: 10.1186/s41232-024-00361-2
Hayato Kodama, Kentaro Endo, Ichiro Sekiya
{"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。
Inflammation and regeneration 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。
Inflammation and regeneration 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
{"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。
Inflammation and regeneration Pub Date : 2024-10-31 DOI: 10.1186/s41232-024-00357-y
Yining Liu, Wei Zhang, Nan Lin, Zelei Yang, Yanxin Liu, Huaxia Chen
{"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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信