Cell Communication and Signaling最新文献

筛选
英文 中文
Influenza virus infection activates TAK1 to suppress RIPK3-independent apoptosis and RIPK1-dependent necroptosis. 流感病毒感染会激活 TAK1,从而抑制 RIPK3 依赖性凋亡和 RIPK1 依赖性坏死。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2024-07-23 DOI: 10.1186/s12964-024-01727-2
Yuling Sun, Lei Ji, Wei Liu, Jing Sun, Penggang Liu, Xiaoquan Wang, Xiufan Liu, Xiulong Xu
{"title":"Influenza virus infection activates TAK1 to suppress RIPK3-independent apoptosis and RIPK1-dependent necroptosis.","authors":"Yuling Sun, Lei Ji, Wei Liu, Jing Sun, Penggang Liu, Xiaoquan Wang, Xiufan Liu, Xiulong Xu","doi":"10.1186/s12964-024-01727-2","DOIUrl":"10.1186/s12964-024-01727-2","url":null,"abstract":"<p><p>Many DNA viruses develop various strategies to inhibit cell death to facilitate their replication. However, whether influenza A virus (IAV), a fast-replicating RNA virus, attenuates cell death remains unknown. Here, we report that IAV infection induces TAK1 phosphorylation in a murine alveolar epithelial cell line (LET1) and a murine fibroblastoma cell line (L929). The TAK1-specific inhibitor 5Z-7-Oxzeneonal (5Z) and TAK1 knockout significantly enhance IAV-induced apoptosis, as evidenced by increased PARP, caspase-8, and caspase-3 cleavage. TAK1 inhibition also increases necroptosis as evidenced by increased RIPK1<sup>S166</sup>, RIPK3<sup>T231/S232</sup>, and MLKL<sup>S345</sup> phosphorylation. Mechanistically, TAK1 activates IKK, which phosphorylates RIPK1<sup>S25</sup> and inhibits its activation. TAK1 also activates p38 and its downstream kinase MK2, which phosphorylates RIPK1<sup>S321</sup> but does not affect RIPK1 activation. Further investigation revealed that the RIPK1 inhibitor Nec-1 and RIPK1 knockout abrogate IAV-induced apoptosis and necroptosis; re-expression of wild-type but not kinase-dead (KD)-RIPK1 restores IAV-induced cell death. ZBP1 knockout abrogates IAV-induced cell death, whereas RIPK3 knockout inhibits IAV-induced necroptosis but not apoptosis. 5Z treatment enhances IAV-induced cell death and slightly reduces the inflammatory response in the lungs of H1N1 virus-infected mice and prolongs the survival of IAV-infected mice. Our study provides evidence that IAV activates TAK1 to suppress RIPK1-dependent apoptosis and necroptosis, and that RIPK3 is required for IAV-induced necroptosis but not apoptosis in epithelial cells.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-07-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264382/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141753435","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Advancing stroke therapy: innovative approaches with stem cell-derived extracellular vesicles 推进中风治疗:利用干细胞衍生细胞外囊泡的创新方法
IF 8.4 2区 生物学
Cell Communication and Signaling Pub Date : 2024-07-22 DOI: 10.1186/s12964-024-01752-1
Jiahao Song, Da Zhou, Lili Cui, Chuanjie Wu, Lina Jia, Mengqi Wang, Jingrun Li, Jingyuan Ya, Xunming Ji, Ran Meng
{"title":"Advancing stroke therapy: innovative approaches with stem cell-derived extracellular vesicles","authors":"Jiahao Song, Da Zhou, Lili Cui, Chuanjie Wu, Lina Jia, Mengqi Wang, Jingrun Li, Jingyuan Ya, Xunming Ji, Ran Meng","doi":"10.1186/s12964-024-01752-1","DOIUrl":"https://doi.org/10.1186/s12964-024-01752-1","url":null,"abstract":"Stroke is a leading cause of mortality and long-term disability globally, with acute ischemic stroke (AIS) being the most common subtype. Despite significant advances in reperfusion therapies, their limited time window and associated risks underscore the necessity for novel treatment strategies. Stem cell-derived extracellular vesicles (EVs) have emerged as a promising therapeutic approach due to their ability to modulate the post-stroke microenvironment and facilitate neuroprotection and neurorestoration. This review synthesizes current research on the therapeutic potential of stem cell-derived EVs in AIS, focusing on their origin, biogenesis, mechanisms of action, and strategies for enhancing their targeting capacity and therapeutic efficacy. Additionally, we explore innovative combination therapies and discuss both the challenges and prospects of EV-based treatments. Our findings reveal that stem cell-derived EVs exhibit diverse therapeutic effects in AIS, such as promoting neuronal survival, diminishing neuroinflammation, protecting the blood-brain barrier, and enhancing angiogenesis and neurogenesis. Various strategies, including targeting modifications and cargo modifications, have been developed to improve the efficacy of EVs. Combining EVs with other treatments, such as reperfusion therapy, stem cell transplantation, nanomedicine, and gut microbiome modulation, holds great promise for improving stroke outcomes. However, challenges such as the heterogeneity of EVs and the need for standardized protocols for EV production and quality control remain to be addressed. Stem cell-derived EVs represent a novel therapeutic avenue for AIS, offering the potential to address the limitations of current treatments. Further research is needed to optimize EV-based therapies and translate their benefits to clinical practice, with an emphasis on ensuring safety, overcoming regulatory hurdles, and enhancing the specificity and efficacy of EV delivery to target tissues. ","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.4,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141739830","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: Endometrial senescence is mediated by interleukin 17 receptor B signaling 更正:子宫内膜衰老由白细胞介素 17 受体 B 信号传导介导
IF 8.4 2区 生物学
Cell Communication and Signaling Pub Date : 2024-07-22 DOI: 10.1186/s12964-024-01754-z
Keiko Kawamura, Yumiko Matsumura, Teruhiko Kawamura, Hiromitsu Araki, Norio Hamada, Kazutaka Kuramoto, Hiroshi Yagi, Ichiro Onoyama, Kazuo Asanoma, Kiyoko Kato
{"title":"Correction: Endometrial senescence is mediated by interleukin 17 receptor B signaling","authors":"Keiko Kawamura, Yumiko Matsumura, Teruhiko Kawamura, Hiromitsu Araki, Norio Hamada, Kazutaka Kuramoto, Hiroshi Yagi, Ichiro Onoyama, Kazuo Asanoma, Kiyoko Kato","doi":"10.1186/s12964-024-01754-z","DOIUrl":"https://doi.org/10.1186/s12964-024-01754-z","url":null,"abstract":"<p><b>Correction: Cell Commun Signal 22, 363 (2024)</b>.</p><p><b>https://doi.org/10.1186/s12964-024-01740-5</b>.</p><p> Following publication of the original article [1], the authors reported that the incorrect additional file 12 was published. The correct additional file is published in this correction article and the original article [1] has been corrected.</p><ul data-track-component=\"outbound reference\" data-track-context=\"references section\"><li><p>Kawamura, K., Matsumura, Y., Kawamura, T. et al. Endometrial senescence is mediated by interleukin 17 receptor B signaling. <i>Cell Commun Signal</i><b>22</b>, 363 (2024). https://doi.org/10.1186/s12964-024-01740-5</p></li></ul><p>Download references<svg aria-hidden=\"true\" focusable=\"false\" height=\"16\" role=\"img\" width=\"16\"><use xlink:href=\"#icon-eds-i-download-medium\" xmlns:xlink=\"http://www.w3.org/1999/xlink\"></use></svg></p><h3>Authors and Affiliations</h3><ol><li><p>Department of Obstetrics and Gynecology, Graduate School of Medical Sciences, Kyushu University, Maidashi 3-1-1, Higashi-ku, Fukuoka, 812-8582, Japan</p><p>Keiko Kawamura, Yumiko Matsumura, Teruhiko Kawamura, Norio Hamada, Kazutaka Kuramoto, Hiroshi Yagi, Ichiro Onoyama, Kazuo Asanoma &amp; Kiyoko Kato</p></li><li><p>Department of Business and Technology Management, Faculty of Economics, Kyushu University, Fukuoka, Japan</p><p>Hiromitsu Araki</p></li></ol><span>Authors</span><ol><li><span>Keiko Kawamura</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Yumiko Matsumura</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Teruhiko Kawamura</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Hiromitsu Araki</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Norio Hamada</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Kazutaka Kuramoto</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Hiroshi Yagi</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Ichiro Onoyama</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Kazuo Asanoma</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li><li><span>Kiyoko Kato</span>View author publications<p>You can also search for this author in <span>PubMed<span> </span>Google Scholar</span></p></li></ol><h3>Corresponding author</h3><p>Correspondence","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.4,"publicationDate":"2024-07-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141739829","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
17β-estradiol in colorectal cancer: friend or foe? 结直肠癌中的 17β-雌二醇:敌还是友?
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2024-07-19 DOI: 10.1186/s12964-024-01745-0
Zihong Wu, Chong Xiao, Jiamei Wang, Min Zhou, Fengming You, Xueke Li
{"title":"17β-estradiol in colorectal cancer: friend or foe?","authors":"Zihong Wu, Chong Xiao, Jiamei Wang, Min Zhou, Fengming You, Xueke Li","doi":"10.1186/s12964-024-01745-0","DOIUrl":"10.1186/s12964-024-01745-0","url":null,"abstract":"<p><p>Colorectal cancer (CRC) is a common gastrointestinal malignancy with higher incidence and mortality rates in men compared to women, potentially due to the effects of estrogen signaling. There is substantial evidence supporting the significant role of 17β-Estradiol (E2) in reducing CRC risk in females, although this perspective remains debated. E2 has been demonstrated to inhibit CRC cell proliferation and migration at the cellular level by enhancing DNA mismatch repair, modulating key gene expression, triggering cell cycle arrest, and reducing activity of migration factors. Furthermore, E2 contributes to promote a tumor microenvironment unfavorable for CRC growth by stimulating ERβ expression, reducing inflammatory responses, reversing immunosuppression, and altering the gut microbiome composition. Conversely, under conditions of high oxidative stress, hypoxia, and nutritional deficiencies, E2 may facilitate CRC development through GPER-mediated non-genomic signaling. E2's influence on CRC involves the genomic and non-genomic signals mediated by ERβ and GPER, respectively, leading to its dual roles in anticancer activity and carcinogenesis. This review aims to summarize the potential mechanisms by which E2 directly or indirectly impacts CRC development, providing insights into the phenomenon of sexual dimorphism in CRC and suggesting potential strategies for prevention and treatment.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264751/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141728364","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting TANK-binding kinase 1 attenuates painful diabetic neuropathy via inhibiting microglia pyroptosis. 靶向 TANK 结合激酶 1 可通过抑制小胶质细胞的脓毒症减轻糖尿病神经病变的疼痛。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2024-07-19 DOI: 10.1186/s12964-024-01723-6
Qinming Liao, Yimei Yang, Yilu Li, Jun Zhang, Keke Fan, Yihao Guo, Jun Chen, Yinhao Chen, Pian Zhu, Lijin Huang, Zhongjie Liu
{"title":"Targeting TANK-binding kinase 1 attenuates painful diabetic neuropathy via inhibiting microglia pyroptosis.","authors":"Qinming Liao, Yimei Yang, Yilu Li, Jun Zhang, Keke Fan, Yihao Guo, Jun Chen, Yinhao Chen, Pian Zhu, Lijin Huang, Zhongjie Liu","doi":"10.1186/s12964-024-01723-6","DOIUrl":"10.1186/s12964-024-01723-6","url":null,"abstract":"<p><strong>Background: </strong>Painful diabetic neuropathy (PDN) is closely linked to inflammation, which has been demonstrated to be associated with pyroptosis. Emerging evidence has implicated TANK-binding kinase 1 (TBK1) in various inflammatory diseases. However, it remains unknown whether activated TBK1 causes hyperalgesia via pyroptosis.</p><p><strong>Methods: </strong>PDN mice model of type 1 or type 2 diabetic was induced by C57BL/6J or BKS-DB mice with Lepr gene mutation. For type 2 diabetes PDN model, TBK1-siRNA, Caspase-1 inhibitor Ac-YVAD-cmk or TBK1 inhibitor amlexanox (AMX) were delivered by intrathecal injection or intragastric administration. The pain threshold and plantar skin blood perfusion were evaluated through animal experiments. The assessments of spinal cord, dorsal root ganglion, sciatic nerve, plantar skin and serum included western blotting, immunofluorescence, ELISA, and transmission electron microscopy.</p><p><strong>Results: </strong>In the PDN mouse model, we found that TBK1 was significantly activated in the spinal dorsal horn (SDH) and mainly located in microglia, and intrathecal injection of chemically modified TBK1-siRNA could improve hyperalgesia. Herein, we described the mechanism that TBK1 could activate the noncanonical nuclear factor κB (NF-κB) pathway, mediate the activation of NLRP3 inflammasome, trigger microglia pyroptosis, and ultimately induce PDN, which could be reversed following TBK1-siRNA injection. We also found that systemic administration of AMX, a TBK1 inhibitor, could effectively improve peripheral nerve injury. These results revealed the key role of TBK1 in PDN and that TBK1 inhibitor AMX could be a potential strategy for treating PDN.</p><p><strong>Conclusions: </strong>Our findings revealed a novel causal role of TBK1 in pathogenesis of PDN, which raises the possibility of applying amlexanox to selectively target TBK1 as a potential therapeutic strategy for PDN.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-07-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11264750/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141728365","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
IRF1 regulation of ZBP1 links mitochondrial DNA and chondrocyte damage in osteoarthritis. IRF1 对 ZBP1 的调控将线粒体 DNA 与骨关节炎中软骨细胞的损伤联系在一起。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2024-07-18 DOI: 10.1186/s12964-024-01744-1
Kai Sun, Fan Lu, Liangcai Hou, Xiong Zhang, Chunran Pan, Haigang Liu, Zehang Zheng, Zhou Guo, Zhaoxuan Ruan, Yanjun Hou, Jinming Zhang, Fengjing Guo, Wentao Zhu
{"title":"IRF1 regulation of ZBP1 links mitochondrial DNA and chondrocyte damage in osteoarthritis.","authors":"Kai Sun, Fan Lu, Liangcai Hou, Xiong Zhang, Chunran Pan, Haigang Liu, Zehang Zheng, Zhou Guo, Zhaoxuan Ruan, Yanjun Hou, Jinming Zhang, Fengjing Guo, Wentao Zhu","doi":"10.1186/s12964-024-01744-1","DOIUrl":"10.1186/s12964-024-01744-1","url":null,"abstract":"<p><strong>Background: </strong>Z-DNA binding protein 1 (ZBP1) is a nucleic acid sensor that is involved in multiple inflammatory diseases, but whether and how it contributes to osteoarthritis (OA) are unclear.</p><p><strong>Methods: </strong>Cartilage tissues were harvested from patients with OA and a murine model of OA to evaluate ZBP1 expression. Subsequently, the functional role and mechanism of ZBP1 were examined in primary chondrocytes, and the role of ZBP1 in OA was explored in mouse models.</p><p><strong>Results: </strong>We showed the upregulation of ZBP1 in articular cartilage originating from OA patients and mice with OA after destabilization of the medial meniscus (DMM) surgery. Specifically, knockdown of ZBP1 alleviated chondrocyte damage and protected mice from DMM-induced OA. Mechanistically, tumor necrosis factor alpha induced ZBP1 overexpression in an interferon regulatory factor 1 (IRF1)-dependent manner and elicited the activation of ZBP1 via mitochondrial DNA (mtDNA) release and ZBP1 binding. The upregulated and activated ZBP1 could interact with receptor-interacting protein kinase 1 and activate the transforming growth factor-beta-activated kinase 1-NF-κB signaling pathway, which led to chondrocyte inflammation and extracellular matrix degradation. Moreover, inhibition of the mtDNA-IRF1-ZBP1 axis with Cyclosporine A, a blocker of mtDNA release, could delay the progression of DMM-induced OA.</p><p><strong>Conclusions: </strong>Our data revealed the pathological role of the mtDNA-IRF1-ZBP1 axis in OA chondrocytes, suggesting that inhibition of this axis could be a viable therapeutic approach for OA.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-07-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11256489/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141725131","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting CCL2-CCR2 signaling pathway alleviates macrophage dysfunction in COPD via PI3K-AKT axis. 靶向 CCL2-CCR2 信号通路可通过 PI3K-AKT 轴缓解慢性阻塞性肺病的巨噬细胞功能障碍。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2024-07-17 DOI: 10.1186/s12964-024-01746-z
Yue Dong, Ying Dong, Chengyue Zhu, Lan Yang, Hanlin Wang, Junqing Li, Zixuan Zheng, Hanwei Zhao, Wanji Xie, Meiting Chen, Zhijun Jie, Jia Li, Yi Zang, Jindong Shi
{"title":"Targeting CCL2-CCR2 signaling pathway alleviates macrophage dysfunction in COPD via PI3K-AKT axis.","authors":"Yue Dong, Ying Dong, Chengyue Zhu, Lan Yang, Hanlin Wang, Junqing Li, Zixuan Zheng, Hanwei Zhao, Wanji Xie, Meiting Chen, Zhijun Jie, Jia Li, Yi Zang, Jindong Shi","doi":"10.1186/s12964-024-01746-z","DOIUrl":"10.1186/s12964-024-01746-z","url":null,"abstract":"<p><strong>Background: </strong>Chronic obstructive pulmonary disease (COPD) remains a leading cause of morbidity and mortality worldwide, characterized by persistent respiratory symptoms and airflow limitation. The involvement of C-C motif chemokine ligand 2 (CCL2) in COPD pathogenesis, particularly in macrophage regulation and activation, is poorly understood despite its recognized role in chronic inflammation. Our study aims to elucidate the regulatory role and molecular mechanisms of CCL2 in the pathogenesis of COPD, providing new insights for therapeutic strategies.</p><p><strong>Methods: </strong>This study focused on the CCL2-CCR2 signaling pathway, exploring its role in COPD pathogenesis using both Ccl2 knockout (KO) mice and pharmacological inhibitors. To dissect the underlying mechanisms, we employed various in vitro and in vivo methods to analyze the secretion patterns and pathogenic effects of CCL2 and its downstream molecular signaling through the CCL2-CCR2 axis.</p><p><strong>Results: </strong>Elevated Ccl2 expression was confirmed in the lungs of COPD mice and was associated with enhanced recruitment and activation of macrophages. Deletion of Ccl2 in knockout mice, as well as treatment with a Ccr2 inhibitor, resulted in protection against CS- and LPS-induced alveolar injury and airway remodeling. Mechanistically, CCL2 was predominantly secreted by bronchial epithelial cells in a process dependent on STAT1 phosphorylation and acted through the CCR2 receptor on macrophages. This interaction activated the PI3K-AKT signaling pathway, which was pivotal for macrophage activation and the secretion of inflammatory cytokines, further influencing the progression of COPD.</p><p><strong>Conclusions: </strong>The study highlighted the crucial role of CCL2 in mediating inflammatory responses and remodeling in COPD. It enhanced our understanding of COPD's molecular mechanisms, particularly how CCL2's interaction with the CCR2 activates critical signaling pathways. Targeting the CCL2-CCR2 axis emerged as a promising strategy to alleviate COPD pathology.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11253350/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141629332","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
miR-29a-3p orchestrates key signaling pathways for enhanced migration of human mesenchymal stem cells. miR-29a-3p 可协调关键信号通路,促进人类间充质干细胞的迁移。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2024-07-17 DOI: 10.1186/s12964-024-01737-0
Dayeon Kang, Taehwan Kim, Ga-Eun Choi, Arum Park, Jin Yoon, Jinho Yu, Nayoung Suh
{"title":"miR-29a-3p orchestrates key signaling pathways for enhanced migration of human mesenchymal stem cells.","authors":"Dayeon Kang, Taehwan Kim, Ga-Eun Choi, Arum Park, Jin Yoon, Jinho Yu, Nayoung Suh","doi":"10.1186/s12964-024-01737-0","DOIUrl":"10.1186/s12964-024-01737-0","url":null,"abstract":"<p><strong>Background: </strong>The homing of human mesenchymal stem cells (hMSCs) is crucial for their therapeutic efficacy and is characterized by the orchestrated regulation of multiple signaling modules. However, the principal upstream regulators that synchronize these signaling pathways and their mechanisms during cellular migration remain largely unexplored.</p><p><strong>Methods: </strong>miR-29a-3p was exogenously expressed in either wild-type or DiGeorge syndrome critical region 8 (DGCR8) knockdown hMSCs. Multiple pathway components were analyzed using Western blotting, immunohistochemistry, and real-time quantitative PCR. hMSC migration was assessed both in vitro and in vivo through wound healing, Transwell, contraction, and in vivo migration assays. Extensive bioinformatic analyses using gene set enrichment analysis and Ingenuity pathway analysis identified enriched pathways, upstream regulators, and downstream targets.</p><p><strong>Results: </strong>The global depletion of microRNAs (miRNAs) due to DGCR8 gene silencing, a critical component of miRNA biogenesis, significantly impaired hMSC migration. The bioinformatics analysis identified miR-29a-3p as a pivotal upstream regulator. Its overexpression in DGCR8-knockdown hMSCs markedly improved their migration capabilities. Our data demonstrate that miR-29a-3p enhances cell migration by directly inhibiting two key phosphatases: protein tyrosine phosphatase receptor type kappa (PTPRK) and phosphatase and tensin homolog (PTEN). The ectopic expression of miR-29a-3p stabilized the polarization of the Golgi apparatus and actin cytoskeleton during wound healing. It also altered actomyosin contractility and cellular traction forces by changing the distribution and phosphorylation of myosin light chain 2. Additionally, it regulated focal adhesions by modulating the levels of PTPRK and paxillin. In immunocompromised mice, the migration of hMSCs overexpressing miR-29a-3p toward a chemoattractant significantly increased.</p><p><strong>Conclusions: </strong>Our findings identify miR-29a-3p as a key upstream regulator that governs hMSC migration. Specifically, it was found to modulate principal signaling pathways, including polarization, actin cytoskeleton, contractility, and adhesion, both in vitro and in vivo, thereby reinforcing migration regulatory circuits.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-07-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11256664/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141635940","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Endometrial senescence is mediated by interleukin 17 receptor B signaling. 子宫内膜衰老是由白细胞介素 17 受体 B 信号传导介导的。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2024-07-15 DOI: 10.1186/s12964-024-01740-5
Keiko Kawamura, Yumiko Matsumura, Teruhiko Kawamura, Hiromitsu Araki, Norio Hamada, Kazutaka Kuramoto, Hiroshi Yagi, Ichiro Onoyama, Kazuo Asanoma, Kiyoko Kato
{"title":"Endometrial senescence is mediated by interleukin 17 receptor B signaling.","authors":"Keiko Kawamura, Yumiko Matsumura, Teruhiko Kawamura, Hiromitsu Araki, Norio Hamada, Kazutaka Kuramoto, Hiroshi Yagi, Ichiro Onoyama, Kazuo Asanoma, Kiyoko Kato","doi":"10.1186/s12964-024-01740-5","DOIUrl":"10.1186/s12964-024-01740-5","url":null,"abstract":"<p><strong>Background: </strong>We previously identified Il17RB, a member of the IL17 superfamily, as a candidate marker gene for endometrial aging. While IL17RB has been linked to inflammation and malignancies in several organ systems, its function in the endometrium has not been investigated and is thus poorly understood. In the present study, we performed a functional analysis of this receptor with the aim of determining the effects of its age-associated overexpression on the uterine environment.</p><p><strong>Methods: </strong>We analyzed IL17RB-related signaling pathways and downstream gene expression in an immortalized human endometrial glandular epithelial cell line (\"hEM\") forced to express the receptor via lentiviral transduction (\"IL17RB-hEM\"). We also prepared endometrial organoids from human endometrial tissue sourced from hysterectomy patients (\"patient-derived EOs\") and exposed them to cytokines that are upregulated by IL17RB expression to investigate changes in organoid-forming capacity and senescence markers. We analyzed RNA-seq data (GEO accession number GSE132886) from our previous study to identify the signaling pathways associated with altered IL17RB expression. We also analyzed the effects of the JNK pathway on organoid-forming capacity.</p><p><strong>Results: </strong>Stimulation with interleukin 17B enhanced the NF-κB pathway in IL17RB-hEM, resulting in significantly elevated expression of the genes encoding the senescence associated secretory phenotype (SASP) factors IL6, IL8, and IL1β. Of these cytokines, IL1β inhibited endometrial organoid growth. Bioinformatics analysis showed that the JNK signaling pathway was associated with age-related variation in IL17RB expression. When IL17RB-positive cells were cultured in the presence of IL17B, their organoid-forming capacity was slightly but non-significantly lower than in unexposed IL17RB-positive cells, but when IL17B was paired with a JNK inhibitor (SP600125), it was restored to control levels. Further, IL1β exposure significantly reduced organoid-forming capacity and increased p21 expression in endometrial organoids relative to non-exposure (control), but when IL1β was paired with SP600125, both indicators were restored to levels comparable to the control condition.</p><p><strong>Conclusions: </strong>We have revealed an association between IL17RB, whose expression increases in the endometrial glandular epithelium with advancing age, and cellular senescence. Using human endometrial organoids as in vitro model, we found that IL1β inhibits cell proliferation and leads to endometrial senescence via the JNK pathway.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247761/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141621866","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Dihydroorotase MoPyr4 is required for development, pathogenicity, and autophagy in rice blast fungus. 稻瘟病真菌的发育、致病性和自噬都需要二氢烟酸酶MoPyr4。
IF 8.2 2区 生物学
Cell Communication and Signaling Pub Date : 2024-07-15 DOI: 10.1186/s12964-024-01741-4
Jing-Yi Wang, Ying-Ying Cai, Lin Li, Xue-Ming Zhu, Zi-Fang Shen, Zi-He Wang, Jian Liao, Jian-Ping Lu, Xiao-Hong Liu, Fu-Cheng Lin
{"title":"Dihydroorotase MoPyr4 is required for development, pathogenicity, and autophagy in rice blast fungus.","authors":"Jing-Yi Wang, Ying-Ying Cai, Lin Li, Xue-Ming Zhu, Zi-Fang Shen, Zi-He Wang, Jian Liao, Jian-Ping Lu, Xiao-Hong Liu, Fu-Cheng Lin","doi":"10.1186/s12964-024-01741-4","DOIUrl":"10.1186/s12964-024-01741-4","url":null,"abstract":"<p><p>Dihydroorotase (DHOase) is the third enzyme in the six enzymatic reaction steps of the endogenous pyrimidine nucleotide de novo biosynthesis pathway, which is a metabolic pathway conserved in both bacteria and eukaryotes. However, research on the biological function of DHOase in plant pathogenic fungi is very limited. In this study, we identified and named MoPyr4, a homologous protein of Saccharomyces cerevisiae DHOase Ura4, in the rice blast fungus Magnaporthe oryzae and investigated its ability to regulate fungal growth, pathogenicity, and autophagy. Deletion of MoPYR4 led to defects in growth, conidiation, appressorium formation, the transfer and degradation of glycogen and lipid droplets, appressorium turgor accumulation, and invasive hypha expansion in M. oryzae, which eventually resulted in weakened fungal pathogenicity. Long-term replenishment of exogenous uridine-5'-phosphate (UMP) can effectively restore the phenotype and virulence of the ΔMopyr4 mutant. Further study revealed that MoPyr4 also participated in the regulation of the Pmk1-MAPK signaling pathway, co-localized with peroxisomes for the oxidative stress response, and was involved in the regulation of the Osm1-MAPK signaling pathway in response to hyperosmotic stress. In addition, MoPyr4 interacted with MoAtg5, the core protein involved in autophagy, and positively regulated autophagic degradation. Taken together, our results suggested that MoPyr4 for UMP biosynthesis was crucial for the development and pathogenicity of M. oryzae. We also revealed that MoPyr4 played an essential role in the external stress response and pathogenic mechanism through participation in the Pmk1-MAPK signaling pathway, peroxisome-related oxidative stress response mechanism, the Osm1-MAPK signaling pathway and the autophagy pathway.</p>","PeriodicalId":55268,"journal":{"name":"Cell Communication and Signaling","volume":null,"pages":null},"PeriodicalIF":8.2,"publicationDate":"2024-07-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11247805/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141621851","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
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